09.01.2013 Views

2 - TI Pharma

2 - TI Pharma

2 - TI Pharma

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

Diabetogenic Effects of Glucocorticoid Drugs:<br />

The Knowns and The Unknowns<br />

Daniël Henri van Raalte


Diabetogenic Effects of Glucocorticoid Drugs:<br />

The Knowns and The Unknowns<br />

The studies described in this thesis were performed within the framework of the Dutch Top<br />

Institute <strong>Pharma</strong>, project T1-106.<br />

ISBN: 978-94-6191-423-1<br />

Author: Daniël Henri van Raalte<br />

Cover Design: Lotje van Lieshout<br />

Lay-out: Cyriel van Oorschot, Ipskamp drukkers BV<br />

Printed by: Ipskamp Drukkers BV<br />

© D.H. van Raalte, Amsterdam, The Netherlands, 2012.<br />

No part of this thesis may be reproduced, stored or transmitted in any form or by any means,<br />

without prior permission of the author.<br />

Financial support by the Dutch Diabetes Research Foundation and the Dutch Arthritis Foundation<br />

for the publication of this thesis is gratefully acknowledged.<br />

The printing of this thesis was additionally supported by: Boehringer Ingelheim B.V., Eli Lilly<br />

Nederland B.V., GlaxoSmithKline B.V., Novartis <strong>Pharma</strong> B.V., Posthumus Meyjes fonds, Sanofi<br />

–Aventis B.V., Servier Nederland B.V., Vrije Universiteit.


VRIjE UNIVERSITEIT<br />

Diabetogenic Effects of Glucocorticoid Drugs:<br />

The Knowns and The Unknowns<br />

ACADEMISCH PROEFSCHRIFT<br />

ter verkrijging van de graad Doctor aan<br />

de Vrije Universiteit Amsterdam,<br />

op gezag van de rector magnificus<br />

prof.dr. L.M. Bouter,<br />

in het openbaar te verdedigen<br />

ten overstaan van de promotiecommissie<br />

van de Faculteit der Geneeskunde<br />

op vrijdag 23 november 2012 om 13.45 uur<br />

in de aula van de universiteit,<br />

De Boelelaan 1105<br />

door<br />

Daniël Henri van Raalte<br />

geboren te Amsterdam


Promotor: prof.dr. M. Diamant


Leescommissie: prof.dr. M. Boers<br />

prof.dr. A.K. Groen<br />

prof.dr. R.j. Heine<br />

prof.dr. M.H.H. Kramer<br />

dr. D.M. Ouwens<br />

dr. C. Rustemeijer<br />

prof.dr. H.P. Sauerwein<br />

“I not only use all the brains that I have, but all that I can borrow” –<br />

Thomas Woodrow Wilson


CoNTENT<br />

Part I Introduction<br />

Chapter 1 General introduction and outline of the thesis 11<br />

Chapter 2 Novel insights into glucocorticoid-mediated diabetogenic 23<br />

effects: towards expansion of therapeutic options?<br />

Part II Glucocorticoids and islet-cell fuction<br />

Chapter 3 Prednisolone-induced beta-cell dysfunction is associated with 55<br />

impaired endoplasmic reticulum homeostasis in INS-1E cells.<br />

Chapter 4 Acute and 2-week exposure to prednisolone impair different 75<br />

aspects of beta-cell function in healthy men.<br />

Chapter 5 Islet-cell dysfunction induced by glucocorticoid treatment: 91<br />

role for altered sympathovagal balance?<br />

Chapter 6 Glucocorticoid receptor gene polymorphisms are associated 115<br />

with reduced first-phase glucose-stimulated insulin secretion<br />

and disposition index in women, but not in men.<br />

Chapter 7 GLP-1 receptor agonist treatment prevents glucocorticoid 133<br />

induced glucose intolerance and islet-cell dysfunction in<br />

humans.<br />

Part III Mechanisms of glucocorticoid-induced insulin resistance<br />

Chapter 8 Low-dose glucocorticoid treatment affects multiple aspects<br />

of intermediary metabolism in healthy humans: a randomized<br />

controlled trial.<br />

151<br />

Chapter 9 Glycosphingolipid metabolism and mitochondrial function do<br />

not explain glucocorticoid-induced insulin resistance in<br />

healthy men<br />

175<br />

Chapter 10 Glucocorticoid treatment impairs microvascular function in<br />

healthy males, in association with its adverse effects on<br />

glucose metabolism and blood pressure.<br />

193<br />

Chapter 11 Glucocorticoids alter subcutaneous adipose tissue function<br />

and adipokine secretion in vivo in healthy men<br />

217


Chapter 12 Angiopoietine-like protein 4 is differentially regulated by 231<br />

glucocorticoids and insulin in vitro and in vivo in healthy<br />

humans.<br />

Part IV Metabolic effects of glucocorticoids and interaction with<br />

systemic inflammation<br />

Chapter 13 Metabolic effects of high-dose prednisolone treatment in early<br />

rheumatoid arthritis: diabetogenic effects and inflammation<br />

reduction on the balance.<br />

249<br />

Chapter 14 Glucose tolerance, insulin sensitivity and beta-cell function<br />

in patients with rheumatoid arthritis treated with or without<br />

low-to-medium dose glucocorticoids.<br />

267<br />

Part V Discussion<br />

Chapter 15 Diabetogenic effects of glucocorticoid drugs: the knowns<br />

and the unknowns<br />

289<br />

Nederlandse samenvatting (Dutch summary) 329<br />

Dankwoord (Acknowledgements) 339<br />

Biography 347<br />

List of Publications 351<br />

List of co-author affiliations 357<br />

Color Figures 365


PART<br />

Introduction<br />

I


Chapter 1<br />

General Introduction and Outline of the Thesis<br />

D.H. van Raalte and M.Diamant


Chapter 1 General Introduction<br />

The involvement of hormones secreted by the adrenal cortex in intermediary metabolism was<br />

first established in 1908, when the French investigators Bierry and Malloizel discovered that<br />

adrenalectomized dogs developed lower fasting glucose levels [1]. In 1910, hypoglycemia was<br />

first described in association with Addison’s disease [2]. In the following decades, the critical<br />

role of the adrenal cortex in intermediary metabolism and energy homeostasis was further<br />

characterized [3]. A major advance was made in 1936 with the simultaneous isolation of the<br />

inactive form of the adrenal hormone cortisol, so called cortisone, by the Polish-born, Swiss<br />

chemist Tadeusz Reichstein [4] and the American scientist Edward Calvin Kendall [5]. This<br />

breakthrough enabled further experiments into the various physiological roles of adrenal<br />

cortex hormones. Not only were the metabolic effects of cortisone further explored, cortisone<br />

was also shown to reduce stress responses and traumatic shock in rats [6].<br />

The amount of cortisone that could be isolated from bovine adrenal glands, however, was<br />

small, and the need to produce adrenocorticosteroids through synthetic methods became soon<br />

apparent. This process was fuelled by the US entry into the Second World War, when rumors<br />

circulated that Luftwaffe pilots were taking adrenal extracts to increase their resistance to<br />

oxygen deprivation at high altitudes. Although this rumor was never confirmed, it induced an<br />

all-out quest for large-scale synthesis of active adrenal hormone, which was given even higher<br />

priority than the development of penicillin and anti-malarials [7,8].<br />

In 1946, Lewis Hastings Sarett of Merck Research Laboratories succeeded in synthetically<br />

producing cortisone from desoxycholic acid [9]. By the summer of 1948, sufficient material<br />

was produced to initiate the first studies in humans. In clinical trials conducted in the Mayo<br />

clinic by the rheumatologist Philip Showalter Hench, a collaborator of Kendall, cortisone<br />

treatment for the first time improved the symptoms of Addison’s disease. In addition, Hench<br />

tested cortisone in patients with rheumatoid arthritis, driven by observations that joint<br />

complains were reduced during pregnancy and jaundice, conditions in which sex steroids and<br />

bile acids are increased, substances that were found closely related to the novel discovered<br />

steroid cortisone. Indeed, cortisone treatment induced a spectacular reduction in joint<br />

tenderness and swelling in chronic rheumatoid arthritis patients [10,11]. In the following<br />

years, the use of adrenal cortex hormones formulations was successfully introduced in the<br />

treatment of an increasing number of diseases due to their potent anti-inflammatory and<br />

immunosuppressive effects. Tadeusz Reichstein, Philip Showalter Hench and Edward Calvin<br />

Kendall shared the Nobel Prize for Physiology or Medicine in 1950 ‘for research on the<br />

structure and biological effects of adrenal cortex hormones’. In the same year, cortisone was<br />

officially launched as a pharmacological agent.<br />

12


Figure 1. Overview of the therapeutic and adverse effects induced by glucocorticoid treatment. (See<br />

page 366 for full color figure)<br />

However, with its increasing use, it became quickly apparent that high-dose glucocorticoid<br />

(GC) treatment induced several undesired effects, ranging from salt and water retention<br />

to increased gastric acidity and psychosis. To this end, in the 1950-1960s, newer synthetic<br />

GC compounds were developed, including prednisolone and dexamethasone, which were<br />

designed to induce fewer side effects. Prednisolone, which is characterized by a 1,2 double bond<br />

in the A-ring, was first produced in 1955 by Schering Corporation and was introduced into the<br />

market a decade later under the brand name Meticortelone. When this structure additionally<br />

undergoes 9α-fluorination, 1-dehydrogenation and 16α-methylation, dexamethasone is<br />

yielded and this compound was introduced for clinical use a year later. Although these newer<br />

formulation significantly reduced side effects as compared to cortisone, many important side<br />

effects of GC treatment remain present today (Figure 1). Of these adverse effects, unfavorable<br />

changes in cardiometabolic parameters are prominent and include the development of<br />

glucose intolerance, diabetes, visceral adiposity, dyslipidemia, skeletal muscle atrophy and<br />

hypertension [12]. The mechanisms underlying these so called diabetogenic effects of GCs<br />

regarding glucose, lipid and protein metabolism were studied extensively in the 1960-1970s<br />

13<br />

1<br />

Chapter 1


Chapter 1 General Introduction<br />

and were mainly attributed to GC-induced insulin resistance [13-21]. After this period of<br />

intensive research on the metabolic effects of GCs, research slowed down in this area for<br />

about two decades (Figure 2).<br />

Figure 2. The number of new publications on Pubmed with search terms “glucocorticoids” and “diabetes”<br />

shown per 5-year intervals. After 1975, the amount of research performed on the topic declined<br />

somewhat, but in recent years, the subject has received full attention.<br />

In the last 15 years, however, research into the diabetogenic effects of GCs has been revived,<br />

mainly due to two different reasons. First, it became clear that deregulated (tissue) GC<br />

metabolism may play a role in ‘idiopathic’ obesity and metabolic syndrome, encouraged by<br />

the observation that several features of these conditions strikingly resemble the phenotype of<br />

chronic GC excess. It was postulated by Björntorp that low-grade chronic stress, characterized<br />

by (slightly) increased GC activity, may contribute to visceral obesity, the metabolic syndrome<br />

and cardiovascular disease [22]. In addition, increased tissue cortisol levels, generated from<br />

the inactive metabolite cortisone by enhanced activity of the enzyme 11-beta hydroxysteroid<br />

dehydrogenase (HSD) type 1, were demonstrated in adipose tissue derived from rodent<br />

models of obesity and from obese humans [23]. This makes the hormone 11-beta HSD type 1 an<br />

attractive therapeutic candidate for the treatment of obesity and its metabolic consequences.<br />

As such, a battery of pharmaceutical companies is currently developing compounds that may<br />

reduce tissue cortisol levels by targeting 11-beta HSD type 1. The amount of research done on<br />

this topic has skyrocketed during the last decade (Figure 3).<br />

Second, improved understanding of the mode of action of GCs on the molecular and<br />

cellular level has led to the development of so-called dissociated glucocorticoid receptor<br />

(GR) agonists (Figure 4). In 1985, it was discovered that GCs exert their actions through<br />

14


inding to the intracellular GR, following which this complex migrates to the nucleus, where<br />

it regulates target gene expression [24,25]. Additionally, it became clear that the antiinflammatory<br />

actions and dysmetabolic effects of GCs may be the result of different genomic<br />

actions, i.e. transrepression and transactivation, respectively. This suggests that these effects<br />

may potentially be separated, which forms the basis of these novel GR agonists [25-32].<br />

Several compounds with a dissociated profile are currently under development by different<br />

pharmaceutical companies and have shown promising results in disease and side effect<br />

animal models, where they reduced inflammation without altering glucose levels [33].<br />

Figure 3. The number of new publications on Pubmed with search term “11-beta hydroxysteroid<br />

dehydrogenase” shown per decade. The number of publications on this topic has skyrocketed in the past<br />

2 decades.<br />

Figure 4. The number of new publications on Pubmed with search term “selective glucocorticoid<br />

receptor agonists” shown per decade. In the last decade, information on the development of these novel<br />

compounds has found its way to the public domain.<br />

15<br />

1<br />

Chapter 1


Chapter 1 General Introduction<br />

The work done in this thesis should be seen in light of the development of these dissociated<br />

GR agonists and was conducted within the framework of a relatively novel organization. In<br />

2005, the fifth Dutch Technological Top Institute was founded by the name of Top Institute<br />

<strong>Pharma</strong>, in which the Dutch government, several pharmaceutical companies and all Dutch<br />

universities were brought together with the goal to develop so called priority medicines. In<br />

our Top Institute <strong>Pharma</strong> Project consortium (T1.106), we collaborated with NV Organon<br />

(more recently incorporated first by Schering-Plough and later by Merck Sharp & Dohme),<br />

who are currently developing GR agonists with a dissociated profile.<br />

The clinical development of these novel compounds, however, faces important difficulties.<br />

The mechanisms by which prednisolone induces metabolic changes remain largely unknown.<br />

In addition, due to the differences in the type of GC administered, treatment duration, mode<br />

of administration and chosen dose, several contrasting reports exist in the literature. Also,<br />

since many studies were conducted more than 4 decades ago, it may be difficult to compare<br />

recent studies with the results obtained from those early studies due to different research<br />

methods and techniques that are currently being employed. Thus, in order to be able to<br />

further develop the dissociated GR agonists, it is highly important to first obtain detailed<br />

information regarding the metabolic side effects induced by prednisolone treatment and to<br />

select biomarkers that reflect these metabolic side effects in a dose-dependent manner. Only<br />

after these indicators of efficacy and safety have become available, it will be possible to design<br />

compounds with an improved therapeutic index compared to the currently-available classic<br />

GR agonists.<br />

In the present thesis, of which the PANTHEON (Prednisolone peripherAl effects oN glucose<br />

meTabolism, metabolic Hormones, insulin sEnsitivity and secretioN)-trials form the<br />

backbone, we aimed to assess the peripheral metabolic effects of both low-and high-dose GC<br />

treatment in healthy volunteers and patients with rheumatoid arthritis, focusing specifically<br />

on the effects of prednisolone on islet-cell function and insulin sensitivity of various metabolic<br />

pathways.<br />

In chapter 2 of this thesis, an overview of the knowledge regarding the diabetogenic side<br />

effects of GC treatment is presented that was available at the time when our studies were<br />

initiated. Part II describes the effects of GC treatment on pancreatic islet-cell function. In<br />

chapter 3 mechanisms underlying the deleterious effects of GCs on beta-cell function are<br />

addressed in INS-1E cells in vitro, in particular the involvement of endoplasmic reticulum<br />

stress. In chapter 4, the effects of both acute and more prolonged high-dose GC treatment on<br />

beta-cell function are described, using mathematical modeling analysis of glucose, insulin and<br />

C-peptide concentrations obtained from standardized meal challenge tests. In chapter 5, we<br />

16


further explored the effects of both low- and high-dose GC treatment on both alpha- and betacell<br />

function in healthy volunteers using both intravenous hyperglycemic clamp studies and<br />

meal challenge tests. In chapter 6, in order to further delineate the role of GR signaling in betacell<br />

function, we investigated the effects of single nucleotide polymorphisms (SNPs) in the GR<br />

gene on measures of beta-cell function in a cohort of 449 subjects. In chapter 7, we explored<br />

the potential of the novel class of glucose-lowering agents the glucagon-like peptide (GLP)-<br />

1 receptor agonists to prevent the dysmetabolic effects induced by GC treatment in healthy<br />

subjects. Part III of the thesis focuses on GC-induced insulin resistance and the mechanisms<br />

that may contribute to this well-known but only partly understood side effect of GC treatment.<br />

In chapter 8, the effects of both low- and high-dose GC treatment on several aspects of<br />

intermediary metabolism is addressed. Possible mediators of GC-induced insulin resistance in<br />

skeletal muscle, including impaired sphingolipid metabolism and mitochondrial dysfunction,<br />

are investigated in chapter 9. In chapter 10 the possible role of microvascular dysfunction<br />

induced by GC treatment was addressed and in chapter 11 we investigated the effects of GC<br />

treatment on adipose tissue function and adipokine secretion. In chapter 12, the role of the<br />

novel endocrine factor angiopoietin-like protein 4 in GC-induced metabolic alterations was<br />

studied, by combining an in vivo (randomized controlled trial in healthy individuals) and in<br />

vitro (cell culture) approach. In part IV of this thesis, using an oral glucose tolerance test<br />

to assess both insulin secretory response and surrogate markers of insulin resistance, the<br />

diabetogenic effects of GC treatment were investigated in two different rheumatoid arthritis<br />

populations, allowing to study the interaction with systemic inflammation. In chapter 13, the<br />

acute effects of high-dose prednisolone treatment on glucose tolerance, insulin sensitivity and<br />

insulin secretion in patients with early and active rheumatoid arthritis are described, whereas<br />

in chapter 14, the effects of chronic GC treatment on glucose tolerance, insulin sensitivity and<br />

insulin secretion are assessed in patients with long-standing rheumatoid arthritis.<br />

In the general discussion (part V, chapter 15), an overview will be presented of the<br />

pathophysiology of glucose intolerance induced by GC treatment. Additionally, implications<br />

will be discussed for the development of dissociated GR agonists. Finally, since these<br />

compounds are still under development, a proposition will be made how GC-induced diabetes<br />

may best be treated in clinical practice.<br />

17<br />

1<br />

Chapter 1


Chapter 1 General Introduction<br />

REFERENCES<br />

1. Bierry H, Malloizel L. Hypoglycemic après décapsulation, effets de l’injection d’adrénaline sur les<br />

animaux décapsules. Co R Soc Biol. 1908; 65:232-4.<br />

2. Porges O. Über hypoglykämie bei Morbus Addison Sowie bei nebennierenlosen Hunden. Z Klin Med.<br />

1910; 69:341-4<br />

3. Long C, Katzin B, Fry E. The adrenal cortex and carbohydrate metabolism. Endocrinology. 1940;<br />

26:309-44<br />

4. Reichstein T. Über bestandteile der nebennieren-rind, VI. Trennungsmethoden sowie isolierung der<br />

substanzen Fa, H und j.. Helvetica Chimica Acta. 1936; 19:1107-26.<br />

5. Mason HL, Meyers CS, Kendall EC. The chemistry of crystalline substances isolated from the<br />

suprarenal gland. j Biol Chem. 1936; 114:613-631<br />

6. Selye H, Dosne C, Bassett L, Whittaker j. On the therapeutic value of adrenal cortical hormones in<br />

traumatic shock and allied conditions. Can Med Assoc j. 1940; 43:1-8<br />

7. Hillier SG. Diamonds are forever: the cortisone legacy. j Endocrinol. 2007; 195(1):1-6<br />

8. Quirke V. Making British cortisone: Glaxo and the development of corticosteroids in Britain in the<br />

1950s-1960s. Stud Hist Philos Biol Biomed Sci. 2005; 36(4):645-74<br />

9. Sarett LH. Partial synthesis of pregnene-4-triol-17(beta), 20(beta), 21-dione-3,11 and pregnene-4diol-17(beta),<br />

21-trione-3,11,20 monoacetate? j Biol Chem. 1946; 162:601-31<br />

10. Hench PS, Kendall EC, Slocumb CH, Polley HF. The effect of a hormone of the adrenal cortex<br />

(17-hydroxy-11-dehydrocorticosterone; compound E) and of pituitary adrenocorticotropic<br />

hormone on rheumatoid arthritis. Mayo Clin Proc. 1949; 24(8):181-97<br />

11. Hench PS, Kendall EC, Slocumb CH, Polley HF. Effects of cortisone acetate and pituitary ACTH on<br />

rheumatoid arthritis, rheumatic fever and certain other conditions. Arch Intern Med (Chic). 1950;<br />

85(4):545-666<br />

12. Schacke H, Docke WD, Asadullah K. Mechanisms involved in the side effects of glucocorticoids.<br />

<strong>Pharma</strong>col Ther. 2002; 96(1):23-43<br />

13. Exton jH, Mallette LE, jefferson LS, Wong EH, Friedmann N, Miller TB, jr., Park CR. The hormonal<br />

control of hepatic gluconeogenesis. Recent Prog Horm Res. 1970; 26:411-461<br />

14. Fain jN. Effects of dexamethasone and 2-deoxy-d-glucose on fructose and gluctose metabolism by<br />

incubated adipose tissue j Biol Chem. 1964; 239:958-62<br />

15. Ginsburg j, Paton A. Effects of insulin after adrenalectomy. Lancet. 1956; 271(6941):491-4<br />

16. Issekutz B, jr., Borkow I. Effect of glucagon and glucose load on glucose kinetics, plasma FFA, and<br />

insulin in dogs treated with methylprednisolone. Metabolism. 1973; 22(1):39-49<br />

18


17. Kaplan SA, Shimizu CS. Effects of cortisol on amino acids in skeletal muscle and plasma.<br />

Endocrinology. 1963; 72:267-72<br />

18. Lecocq FR, Mebane D, Madison LL. The acute effect of hydrocortisone on hepatic glucose output and<br />

peripheral glucose utilization j Clin Invest. 1964; 43:237-46<br />

19. Munck A. Glucocorticoid inhibition of glucose uptake by peripheral tissues: old and new evidence,<br />

molecular mechanisms, and physiological significance. Perspect Biol Med. 1971; 14(2):265-9<br />

20. Ninomiya R, Forbath NF, Hetenyi G, jr. Effect of adrenal steroids on glucose kinetics in normal and<br />

diabetic dogs. Diabetes. 1965; 14(11):729-39<br />

21. Tomas FM, Munro HN, Young VR. Effect of glucocorticoid administration on the rate of muscle<br />

protein breakdown in vivo in rats, as measured by urinary excretion of N tau-methylhistidine.<br />

Biochem j. 1979; 178(1):139-46<br />

22. Björntorp P. The associations between obesity, adipose tissue distribution and disease. Acta Med<br />

Scand Suppl. 1988; 723:121-34.<br />

23. Morton NM, Seckl jR. 11beta-hydroxysteroid dehydrogenase type 1 and obesity. Front Horm Res.<br />

2008; 36:146-64<br />

24. Hollenberg SM, Weinberger C, Ong ES, Cerelli G, Oro A, Lebo R, Thompson EB, Rosenfeld MG, Evans<br />

RM. Primary structure and expression of a function human glucocorticoid receptor cDNA. Nature.<br />

1985; 318(6047):635-41.<br />

25. Rosen j, Miner jN. The search for safer glucocorticoid receptor ligands. Endocr Rev 2005; 26(3):452-<br />

64<br />

26. McMaster A, Ray DW. Modelling the glucocorticoid receptor and producing therapeutic agents with<br />

anti-inflammatory effects but reduced side-effects. Exp Physiol. 2007; 92(2):299-309<br />

27. Lowenberg M, Stahn C, Hommes DW, Buttgereit F. Novel insights into mechanisms of glucocorticoid<br />

action and the development of new glucocorticoid receptor ligands. Steroids. 2008; 73(9-10):1025-<br />

9<br />

28. Schacke H, Rehwinkel H, Asadullah K, Cato AC. Insight into the molecular mechanisms of<br />

glucocorticoid receptor action promotes identification of novel ligands with an improved therapeutic<br />

index. Exp Dermatol. 2006; 15(8):565-73<br />

29. Schacke H, Berger M, Rehwinkel H, Asadullah K. Selective glucocorticoid receptor agonists (SEGRAs):<br />

novel ligands with an improved therapeutic index. Mol Cell Endocrinol 2007; 275(1-2):109-17<br />

30. Song IH, Gold R, Straub RH, Burmester GR, Buttgereit F. New glucocorticoids on the horizon: repress,<br />

don’t activate! J Rheumatol. 2005; 32(7):1199-1207<br />

31. Stahn C, Lowenberg M, Hommes DW, Buttgereit F. Molecular mechanisms of glucocorticoid action<br />

and selective glucocorticoid receptor agonists. Mol Cell Endocrinol. 2007; 275(1-2):71-8<br />

19<br />

1<br />

Chapter 1


Chapter 1 General Introduction<br />

32. Thompson CF, Quraishi N, Ali A, Mosley RT, Tata jR, Hammond ML, Balkovec jM, Einstein M, Ge L,<br />

20<br />

Harris G, Kelly TM, Mazur P, Pandit S, Santoro j, Sitlani A, Wang C, Williamson j, Miller DK, Yamin<br />

TT, Thompson CM, O’Neill EA, Zaller D, Forrest MJ, Carballo-Jane E, Luell S. Novel glucocorticoids<br />

containing a 6,5-bicyclic core fused to a pyrazole ring: synthesis, in vitro profile, molecular modeling<br />

studies, and in vivo experiments. Bioorg Med Chem Lett. 2007; 17(12):3354-61.<br />

33. van Lierop Mj, Alkema W, Laskewitz Aj, Dijkema R, van der Maaden HM, Smit Mj, Conti PGM, jams<br />

CGjM, Timmers M, van Boeckel SCAA, Lusher Sj, McGuire R, vanSchaik RC, de Vlieg j, Smeets RL,<br />

Hofstra CL, Boots AMH, van Duin M, Ingelse BA,Schoonen WGEJ, Grefhorst A, van Dijk T, Kuipers<br />

F, Dokter WHA. A novel non-steroidalselective glucocorticoid receptor modulator with full antiinflammatory<br />

properties andimproved therapeutic index. Manuscript submitted. 2012.


Chapter 2<br />

Novel Insights into Glucocorticoid-Mediated<br />

Diabetogenic Effects: Towards Expansion of<br />

Therapeutic Options?<br />

D.H. van Raalte, D.M. Ouwens and M.Diamant<br />

European Journal of Clinical Investigation. 2009; 39(2):81-93


Chapter 2 Glucocorticoids: metabolic side effects<br />

24<br />

ABSTRACT<br />

At pharmacological concentrations, glucocorticoids (GCs) display potent anti-inflammatory<br />

effects, and are therefore frequently prescribed by physicians to treat a wide variety of<br />

diseases. Despite excellent efficacy, GC therapy is hampered by their notorious metabolic<br />

side-effect profile. Chronic exposure to increased levels of circulating GCs is associated<br />

with central adiposity, dyslipidemia, skeletal muscle wasting, insulin resistance, glucose<br />

intolerance and overt diabetes. Remarkably, many of these side effects of GC treatment<br />

resemble the various components of the metabolic syndrome (MetS), in which indeed subtle<br />

disturbances in the hypothalamic-pituitary-adrenal (HPA) axis and/or increased tissue<br />

sensitivity to GCs have been reported. Recent developments have led to renewed interest<br />

in the mechanisms of GCs diabetogenic effects. First, ‘selective dissociating glucocorticoid<br />

receptor (GR) ligands’, which aim to segregate GCs anti-inflammatory and metabolic<br />

actions, are currently being developed. Second, at present, selective 11β-hydroxysteroid<br />

dehydrogenase type 1 (11β-HSD1) inhibitors, which may reduce local GC concentrations<br />

by inhibiting cortisone to cortisol conversion, are evaluated in clinical trials as a novel<br />

treatment modality for the MetS.<br />

In this review, we provide an update of the current knowledge on the mechanisms that<br />

underlie GC-induced dysmetabolic effects. In particular, recent progress in research into<br />

the role of GCs in the pathogenesis of insulin resistance and beta-cell dysfunction will be<br />

discussed.


1. INTRoDUC<strong>TI</strong>oN<br />

Glucocorticoid (GC) hormones are secreted by the cortex of the adrenal gland, under control<br />

of the hypothalamic-pituitary-adrenal (HPA) axis. They play essential roles in glucose, lipid<br />

and protein metabolism and contribute to energy homeostasis. In the postabsorptive state,<br />

GCs provide substrate for oxidative metabolism by increasing lipolysis, proteolysis and<br />

hepatic glucose production [1].<br />

At supraphysiological concentrations, GCs display strong anti-inflammatory and<br />

immunosuppressive actions. Hence, GCs are, since decades, the cornerstone in the treatment<br />

of numerous inflammatory conditions. Annually, approximately 10 million new prescriptions<br />

for oral corticosteroids are issued in the United States. Despite their excellent efficacy, GC<br />

use is limited by their side-effect profile, which is dependent of the administered dose and<br />

duration of treatment [2]. Among these side effects are metabolic derangements, including<br />

the development of central adiposity [3], hepatic steatosis [4], dyslipidemia characterized by<br />

increased plasma levels of triglyceride rich lipoproteins (TRL) and nonesterified fatty acids<br />

(NEFA) [5,6], increased breakdown of skeletal muscle mass [7], insulin resistance, glucose<br />

intolerance and overt diabetes in susceptible individuals [2]. In addition, GC-induced beta-cell<br />

dysfunction, which may determine the progression from insulin resistance to hyperglycemia,<br />

has been the topic of extensive research more recently [8,9].<br />

A similar cluster of metabolic abnormalities is also present in subjects with the metabolic<br />

syndrome (MetS) [10]. Although in one study glucose intolerant subjects were shown to<br />

have higher cortisol levels during an oral glucose tolerance test (OGTT) than healthy controls<br />

[11], at present, no clear association was established between excess plasma levels of GCs<br />

and ‘idiopathic’ obesity and/or the MetS [12]. In the last decade, however, the importance of<br />

factors that modulate the biological effects of GCs in target tissues has been recognized [13]. In<br />

this respect, the 11β-hydroxysteroid dehydrogenase (11β-HSD) enzymes, which interconvert<br />

cortisol and its inactive metabolite cortisone, are of particular interest. 11β-HSD type 1, which<br />

is predominantly expressed in liver and adipose tissue, amplifies local GC action by conversion<br />

of cortisone to cortisol, whereas 11β-HSD type 2, which is mainly expressed in the kidney,<br />

reduces GC-induced effects by converting cortisol to cortisone [14]. Indeed, in several studies<br />

in both rodents and humans, 11β-HSD1 expression and activity in subcutaneous adipose<br />

tissue (SCAT) were shown to be positively associated with insulin resistance and obesity [15].<br />

25<br />

2<br />

Chapter 2


Chapter 2 Glucocorticoids: metabolic side effects<br />

Figure 1. Molecular mechanisms of GR action. GCs regulate protein activity both by genomic and<br />

nongenomic pathways. In addition, various modes of transcription regulation by the GR-ligand complex<br />

have been described. Positive regulation of genes (transactivation) is mediated by binding of a ligandactivated<br />

GR homodimer to a GC response element (GRE), which is usually located in the promoter<br />

region of the target gene. An example of such regulation is the gene encoding for the gluconeogenic<br />

enzyme phosphoenolpyruvate carboxykinase (PEPCK). Ligand-activated GR homodimers may also bind<br />

to negative GREs, leading to regression of gene transcription. Moreover, inhibition of target genes may<br />

also be achieved by interaction of GR monomers with other transcription factors via protein-protein<br />

interaction (transrepression). An example of the latter includes binding of GC monomer to the p65<br />

subunit of nuclear factor κB (NF-κB), thereby preventing gene transcription. The transactivation pathway<br />

seems mainly responsible for GC-induced side effects, whereas the transrepression pathway is thought<br />

to primarily induce GC’s anti-inflammatory effects. Abbreviations: DNA: deoxyribonucleic acid; GC:<br />

glucocorticoid; GR: glucocorticoid receptor; GRE: glucocorticoid response element; mRNA: messenger<br />

ribonucleic acid; P: phosphorylated.<br />

1.1 <strong>Pharma</strong>cological modulation of glucocorticoid action<br />

Due to increased insight into the mechanisms of GC-induced dysmetabolic effects in<br />

recent years, two novel therapeutic classes are currently being explored. First, improved<br />

understanding of the molecular and cellular actions of GCs has lead to the development of<br />

compounds that may display a reduced metabolic side-effect profile compared to classic<br />

glucocorticoid receptor (GR) ligands, while preserving GC-induced anti-inflammatory effects.<br />

These so-called ‘dissociating (GR) activators’ are based on the finding, that the mechanisms by<br />

26


which GCs enhance gene transcription (‘transactivation’) differ from the modes by which GCs<br />

inhibit gene transcription (‘transrepression’) (detailed in Figure 1). GC-induced mechanisms<br />

of transrepression are considered to be responsible for GCs anti-inflammatory actions,<br />

whereas the transactivation pathways are associated with most of GC-induced side effects.<br />

Clearly, new compounds with a ‘dissociated profile’, i.e. that mainly induce transrepression<br />

with reduced transactivation activity, will have an increased therapeutic index compared<br />

to the currently available GCs. Currently, a battery of these ligands are being developed by<br />

pharmaceutical companies [16,17].<br />

Second, since in rodent models of obesity and type 2 diabetes (T2DM) pharmacological<br />

inhibition of 11βHSD1 improved glucose tolerance, insulin sensitivity and lipid profiles [18],<br />

11β-HSD type 1 inhibitors are currently being evaluated in early clinical trials. These agents<br />

are expected to provide a new treatment modality for subjects with the MetS and/or T2DM.<br />

In this review, we will describe the tissue-specific mechanisms underlying the metabolic<br />

derangements associated with GC excess, in particular GC-induced insulin resistance and<br />

beta-cell dysfunction. In addition, the role of GC metabolism in the MetS will be discussed.<br />

2. GLUCoCoR<strong>TI</strong>CoID AC<strong>TI</strong>oNS oN SKELETAL MUSCLE GLUCoSE<br />

AND PRoTEIN METABoLISM<br />

Skeletal muscle tissue plays a crucial role in glucose metabolism. It is responsible for 80% of<br />

postprandial glucose uptake from the circulation [19] and contains the body‘s largest glycogen<br />

stores. The uptake of glucose in skeletal muscle is dependent on the presence of insulin, and<br />

is therefore referred to as insulin-mediated glucose uptake (IMGU). Since impaired glucose<br />

tolerance is a very consistent finding in clinical studies after GC exposure [9,20,21], it has<br />

been assumed that GC-induced changes in metabolic functions of skeletal muscle tissue<br />

may contribute to the development of GC-induced glucometabolic abnormalities. A number<br />

of potential mechanisms through which GCs decrease IMGU in skeletal muscle tissue have<br />

been identified. GCs may induce insulin resistance by directly interfering with the insulin<br />

signaling cascade in skeletal muscle. In addition, GC-induced changes in protein [22] and<br />

lipid metabolism [23,24] may reduce insulin sensitivity of skeletal muscle tissue. The abovementioned<br />

mechanisms are further detailed below.<br />

27<br />

2<br />

Chapter 2


Chapter 2 Glucocorticoids: metabolic side effects<br />

Figure 2. Glucocorticoid-induced effects on glucose and protein metabolism in skeletal muscle<br />

tissue. Insulin stimulates glucose uptake in cells by increasing the translocation of glucose transporter<br />

(GLUT)4 from intracellular vesicles to the cell surface. Upon binding to the transmembrane insulin<br />

receptor (IR), insulin induces GLUT4 migration by activating a cascade of cytosolic proteins, including<br />

insulin receptor substrate (IRS)-1, phosphatidylinositol 3 kinase (PI3-K) and Akt/protein kinase B (PKB),<br />

although the complete cascade remains only partially understood [139]. After glucose is transported<br />

across the plasma membrane, it is further processed by either oxidative or non-oxidative pathways,<br />

the latter being associated with glycogen synthesis. Glycogen synthesis is enhanced by insulin, through<br />

inhibition of glycogen synthase kinase (GSK)-3, which inactivates glycogen synthase (GS) [139].<br />

GCs interfere at several steps in insulin signalling cascade, resulting in reduced glucose uptake and<br />

glycogen synthesis. In addition, blunted insulin signalling contributes to GC-induced protein catabolism.<br />

Abbreviations: 4E-BP1: eIF4E-binding protein 1; GLUT4: glucose transporter 4; GS: glycogen synthase;<br />

GSK-3: glycogen synthase kinase-3; IFG-1: insulin-like growth factor-1; IR: insulin receptor; IRS-1: insulin<br />

receptor substrate-1; mTOR: mammalian target of rapamycin; MuRF-1: Muscle Ring Finger-1; PI3-K:<br />

phosphatidylinositol-3 kinase; PKB: protein kinase B; PP-1: protein phosphatase-1; S6K1: protein S6<br />

kinase 1.<br />

2.1 Glucocorticoids directly inhibit insulin signaling in skeletal muscle<br />

GCs impair IMGU by directly interfering with several components of the insulin signaling<br />

cascade (Figure 2), as was extensively demonstrated both in vitro and in vivo in animals<br />

[25-29]. Since studies into the effects of GCs on the expression of the insulin receptor (IR) in<br />

skeletal muscle have rendered contradictory results [1,28], the markedly reduced IMGU seen<br />

28


after GC-treatment has been proposed as a postreceptor defect [21,30]. Accordingly, in both<br />

L6 skeletal muscle cells and in rat skeletal muscle, dexamethasone treatment decreased the<br />

expression and phosphorylation of insulin receptor substrate (IRS)-1, phosphatidylinositol<br />

3-kinase (PI3-K) and protein kinase B (PKB)/Akt [25,26,28]. In rats, this was indeed shown to<br />

result in reduced glucose transporter (GLUT4) migration to the cell surface [29]. In addition<br />

to reducing glucose uptake, GCs decreased glycogen synthesis rate in Wistar rats by lowering<br />

PKB/Akt and glycogen synthase kinase (GSK)-3 phosphorylation [27].<br />

To date, few data exist regarding the effects of GCs on insulin signaling in humans. In healthy<br />

volunteers, treated for 5 days with 4 mg dexamethasone daily [31], and in patients who were<br />

exposed to long-term, high-dose GCs following renal transplantation [32], reduced glycogen<br />

synthesis rates with concomitant decreased glycogen synthase (GS) concentration and<br />

activity in skeletal muscle biopsies were reported. Based on studies in cell lines and animals,<br />

a schematic overview of the insulin signaling pathway in skeletal muscle and the proposed<br />

sites of modulating action by GCs is depicted in Figure 2.<br />

2.2 Glucocorticoid-induced protein catabolism is linked to insulin resistance<br />

Marked skeletal muscle atrophy is a key feature of prolonged exposure to increased plasma<br />

levels of GCs, as seen in Cushing’s syndrome and during steroid therapy [7]. Also following<br />

short-term high-dose GC treatment, enhanced protein degradation, as measured by stable<br />

isotope dilution methodology, with concomitant elevating circulating amino acid levels, was<br />

reported [33]. Amino acids interfere with insulin signaling by inhibiting insulin-stimulated<br />

IRS phosphorylation and activation of PI3-K in cultured hepatoma cells and myocytes [34],<br />

and were shown to reduce IMGU and glycogen synthesis in humans [22]. Thus, the atrophyrelated<br />

decrease in total muscle area as well as the elevated circulating amino acid levels<br />

contribute to reduced IMGU in GC excess state.<br />

GCs reduce skeletal muscle mass both by decreasing the rate of protein synthesis and by<br />

increasing the rate of protein breakdown [35]. GCs inhibit protein synthesis by reducing<br />

transport of amino acids into the muscle and by abolishing the anabolic effects of insulin and<br />

insulin-like growth factor (IGF)-1. Specifically, the activation of two key proteins in the protein<br />

synthesis machinery, eIF4E-binding protein 1 (4E-BP1) and ribosomal protein S6 kinase 1<br />

(S6K1), is reduced through inhibition of PKB/Akt and mammalian target of rapamycin<br />

(mTOR) phosphorylation. GCs induce muscle proteolysis by activating a number of proteolytic<br />

systems. Most notably, various proteins of the ubiquitin-proteasome system are activated by<br />

GCs, such as Muscle Ring Finger (MuRF)-1 and Atrogin-1. The transcriptional factors FOXO 1<br />

and 3, the gene expression of which are upregulated by GCs, are thought to play a pivotal role<br />

in this pathway [35] (Figure 2).<br />

29<br />

2<br />

Chapter 2


Chapter 2 Glucocorticoids: metabolic side effects<br />

2.3 Glucocorticoid-induced dyslipidemia reduces insulin sensitivity<br />

GCs increase whole-body lipolysis [36], resulting in increased plasma levels of NEFA and TG<br />

[5,6] (further discussed under the section ‘GCs enhance whole-body lipolysis’). Augmented<br />

plasma levels of NEFA may enhance the accumulation of intramyocellular lipids (IMCL),<br />

such as fatty acyl CoA, diacylglycerol (DAG) and ceramide, which reduce glucose uptake and<br />

disposal [37]. It was proposed by Randle in 1963 [38], that intracellular lipids decrease IMGU<br />

by competing with glucose for oxidation, but more recent studies have demonstrated that<br />

IMCL rather reduce IMGU by interfering with insulin signaling. Thus, intracellular lipids may<br />

activate various serine kinases, such as c-Jun amino-terminal kinase (JNK) and IkB kinase-β<br />

(IKK-β), which phosphorylate serine sites on IRS-1, resulting in decreased insulin signaling<br />

[24].<br />

To summarize, GCs reduce insulin sensitivity and consequently IMGU in skeletal muscle<br />

tissue, not only by directly perturbing insulin signaling, but also secondary to unfavourable<br />

changes in protein and lipid metabolism.<br />

3. GLUCoCoR<strong>TI</strong>CoIDS AND HEPA<strong>TI</strong>C GLUCoSE AND LIPID<br />

METABoLISM<br />

The liver plays a key role in controlling glucose and lipid homeostasis, within a complex<br />

regulatory network of hormonal, autonomic nervous and metabolic stimuli. In the fasting<br />

state, the liver maintains euglycemia by producing glucose through both gluconeogenesis<br />

(GNG) and glycogenolysis (GGL). Insulin, in the presence of abundant glucose, is the most<br />

important hormone that suppresses endogenous glucose production (EGP) [39], whereas GCs<br />

and glucagon increase hepatic glucose output.<br />

3.1 Glucocorticoids increase endogenous glucose production<br />

Excess plasma levels of GCs may increase EGP both directly and indirectly, the latter by<br />

antagonizing insulin’s metabolic actions [1,40]. GC-induced hepatic insulin resistance<br />

results in impaired suppression of hepatic glucose production by insulin, especially in the<br />

postprandial state, as was extensively demonstrated in healthy subjects following shortterm<br />

GC exposure [21,41,42]. In addition, GCs directly increase EGP by activating a large<br />

number of genes involved in hepatic carbohydrate metabolism [43]. Accordingly, GCs were<br />

shown to augment EGP in the fasting state in healthy humans [21,42,44], although this could<br />

not be confirmed in other studies [41,45-48]. Possibly the use of different GC compounds<br />

(cortisol versus dexamethasone), different modes of administration (intravenous versus<br />

30


oral), the vulnerability of the population exposed [44] and the different stable isotope dilution<br />

protocols that were employed to quantify EGP, may explain these seemingly contrasting<br />

results. GC-induced increments in EGP in the basal state seem to be driven by increased GNG,<br />

through a number of proposed mechanisms. First, GCs were shown to induce the expression<br />

of rate limiting enzymes of GNG, including phosphoenolpyruvate carboxykinase (PEPCK)<br />

and glucose-6-phosphatase (G6Pase), both in vitro and in vivo in rats [49-51]. Indeed, the<br />

PEPCK gene contains a glucocorticoid response element (GRE) in its promoter region and is<br />

considered a key player in GC-induced hyperglycemia [43]. Second, long-term GC-exposure<br />

promotes breakdown of protein and fat stores [52], thus increasing the supply of substrates<br />

for GNG, such as alanine and glycerol, to the liver [53]. As such, increased proteolysis was<br />

shown to contribute to augmented GNG in obese nondiabetic subjects, in whom enhanced<br />

skeletal muscle GC sensitivity has been reported [54], as compared to lean controls [55].<br />

Third, GCs facilitate metabolite transport across the mitochondrial membranes in rat liver,<br />

which enhances GNG [1,53]. Fourth, GCs might potentiate the effects of other glucoregulatory<br />

hormones, such as glucagon and epinephrine, thus further enhancing EGP [52,56]. Finally,<br />

a role for the nuclear receptor peroxisome proliferator-activated receptor (PPAR)-alpha<br />

in GC-induced hepatic insulin resistance and hyperglycemia has recently been confirmed<br />

[57,58]. PPAR-α, which is predominantly expressed in the liver and is the receptor for<br />

the hypolipidemic fibrates, acts as a fatty acid sensor and is a major regulator of energy<br />

homeostasis by promoting fatty acid oxidation, GNG and ketogenesis [59]. It was elegantly<br />

demonstrated in PPAR-α knock out mice that PPAR-α expression is necessary for GC-induced<br />

increases in EGP [57], and that the autonomous nervous system plays a key role [58].<br />

The importance of the hepatic GC-GR axis in regulating EGP was additionally demonstrated<br />

in transgenic mice with a liver-specific inactivation of the GR. These mice developed<br />

hypoglycemia after prolonged fasting due to impaired activity of gluconeogenic enzymes [60].<br />

3.2. Glucocorticoids alter hepatic lipid metabolism<br />

In addition to regulating hepatic carbohydrate metabolism, insulin plays a key role in<br />

hepatic lipid metabolism. In the postprandial state, portal insulin stimulates lipogenesis and<br />

lipoprotein synthesis, whereas it suppresses very low-density lipoprotein (VLDL) secretion<br />

[61]. GCs induce hepatic insulin resistance, both by direct interference with insulin signaling<br />

and indirectly, by elevating plasma NEFA and TG supply to the liver [61]. GC-induced changes<br />

in hepatic lipid metabolism may induce hepatic steatosis and dyslipidemia in humans,<br />

although the currently available data are not conclusive.<br />

31<br />

2<br />

Chapter 2


Chapter 2 Glucocorticoids: metabolic side effects<br />

Both in vitro and in vivo in rats, GC treatment was shown to induce accumulation of<br />

intrahepatic lipids [62,63], both through increased TG synthesis and through decreased fatty<br />

acid oxidation [63]. In line with these results, mice with a liver-specific disruption of the GR<br />

had diminished hepatic TG levels [64]. In humans, one study indeed reported a prevalence<br />

of hepatic steatosis of up to 20% in patients with Cushing’s disease [65], although additional<br />

reports linking fatty liver to GC excess are limited to case reports in present literature [66,67].<br />

In addition to changing hepatic fat content, GCs may also augment hepatic VLDL secretion,<br />

resulting in increased plasma levels of TG. In studies in rodents, systemic GC excess induced<br />

an atherogenic plasma lipid profile [63]. In patients with Cushing’s disease, enhanced VLDL<br />

secretion by the liver was reported, which was normalized after correction of cortisol levels<br />

[5]. In addition, short-term treatment with prednisolone also increased VLDL concentration<br />

in healthy subjects [68], although the published results are not unequivocal [69].<br />

To summarize, increased levels of GCs in the liver contribute to GC-related dysmetabolic<br />

effects, including insulin resistance, hyperglycemia and dyslipidemia. The effects of GCs on<br />

the development of hepatic steatosis in humans warrants further investigation.<br />

4. GLUCoCoR<strong>TI</strong>CoIDS, BoDy FAT DISTRIBU<strong>TI</strong>oN AND ADIPoSE<br />

<strong>TI</strong>SSUE BIoLoGy<br />

4.1 Glucocorticoids increase body fat content and alter body fat distribution<br />

GCs play a critical role both in the regulation of adipose tissue metabolism and in the<br />

differentiation of pre-adipocytes into mature adipocytes [70]. Excess plasma levels of<br />

GCs, as seen in Cushing’s disease or during prolonged GC therapy, increase body fat mass.<br />

Remarkably, GCs specifically enhance fat deposition in the visceral compartment, while<br />

reducing peripheral fat stores [70]. It has been raised that, in peripheral fat tissue, GCs induce<br />

hormone-sensitive lipase (HSL) and reduce intravascular lipoprotein lipase (LPL) activity,<br />

resulting in augmented fat mobilization. In contrast, in visceral adipose tissue (VAT), GCs may<br />

enhance pre-adipocyte differentiation, LPL activity and TG synthesis, leading to increased<br />

adipose tissue mass [70]. However, the molecular mechanisms underlying the distinct<br />

regulation of central and peripheral adipose tissue depots by GCs remain currently largely<br />

unknown. Off note, recent research demonstrating the role of endogenous GC dysmetabolism<br />

in the development of central adiposity in ‘idiopathic’ obesity and the MetS will be addressed<br />

in a separate section.<br />

Inasmuch as increased VAT is well associated with risk for metabolic and cardiovascular<br />

disease [71], the GC-related increase in VAT may indeed translate into clinical disease [72].<br />

32


4.2. Glucocorticoids modulate adipose tissue biology<br />

4.2.1. Glucocorticoids alter the secretion of adipokines<br />

In addition to changing adipose tissue distribution, GCs also affect adipose tissue biology.<br />

In the past decades, adipose tissue has been identified as an endocrine organ, secreting<br />

numerous metabolically active hormones and peptides, collectively referred to as adipokines<br />

[73]. GCs were shown to change the secretion of a number of these adipokines in vitro and in<br />

vivo in rodents [74]. Whether the expression of the encoding genes are directly altered by GCs,<br />

or whether the changed expression may be secondary to GC-induced adipose tissue insulin<br />

resistance [75-77], is currently not clear. In 3T3-L1 adipocytes and in white adipose tissue<br />

from dexamethasone-treated mice, GCs increased mRNA and protein expression levels of<br />

resistin, an adipocytokine that impairs glucose tolerance. Furthermore, in 3T3-L1 and human<br />

adipocytes, dexamethasone decreased the expression of adiponectin, which is associated with<br />

enhanced insulin sensitivity. Conversely, GCs suppressed the pro-inflammatory adipokines<br />

interleukin (IL)-6 and tumor necrosis factor (TNF)-α in vitro, making these proteins unlikely<br />

candidates for mediating GC-induced insulin resistance. Finally, also in vitro, GCs stimulated<br />

leptin expression and secretion [78].<br />

In humans, both a two and five-day treatment with dexamethasone had no effects on plasma<br />

levels of adiponectin, resistin, TNF-α or leptin in healthy subjects, while insulin sensitivity was<br />

markedly reduced [79,80]. In addition, surgical treatment of Cushing’s disease did not alter<br />

serum adiponectin levels, despite a decrease in cortisol plasma levels and increase in insulin<br />

sensitivity [81]. Whether the expression levels of these adipokines were modulated in adipose<br />

tissue, was not determined. In contrast, a seven-day treatment with the GC prednisolone did<br />

increase the expression of leptin in a group of overweight, postmenopausal women [82].<br />

4.2.2 Glucocorticoids enhance whole-body lipolysis<br />

In addition to modifying the secretion of various adipokines, GCs also alter lipid metabolism<br />

in adipose tissue. GC-induced insulin resistance in adipose tissue may contribute, but GCs<br />

also directly alter the expression of key proteins in lipid metabolism. Thus, GCs were shown<br />

to induce the expression and activity of HSL, the enzyme responsible for the hydrolysis of TG<br />

in adipocytes [83]. Indeed, acute cortisol infusion in healthy humans lead to increased wholebody<br />

lipolysis in several studies, as measured with stable isotopes dilution techniques [36,84-<br />

86]. Although it remains to be determined which adipose tissue compartment particularly<br />

contributes to increased NEFA fluxes, one of these studies reported a decreased FFA efflux<br />

from subcutaneous abdominal tissue concurrently with increased whole-body lipolysis [85],<br />

an observation compatible with the adipose tissue distribution that is seen in patients with<br />

Cushing’s syndrome. In addition, GCs were shown to augment LPL activity in the presence<br />

33<br />

2<br />

Chapter 2


Chapter 2 Glucocorticoids: metabolic side effects<br />

of insulin [87]. Increased LPL activity and intravascular lipolysis, stimulates the uptake of<br />

NEFA and glycerol into adipose tissue. It has been proposed that GCs regulate LPL activity in<br />

a site-specific manner, i.e. increase LPL activity in VAT, but reduce LPL activity in peripheral<br />

fat stores [6,88,89], thus contributing to the Cushing phenotype. Robust evidence for this<br />

hypothesis, however, is lacking in humans.<br />

5. GLUCoCoR<strong>TI</strong>CoIDS AND INSULIN SECRE<strong>TI</strong>oN<br />

The pancreatic beta cell plays a crucial role in glucose metabolism, and in the past decades,<br />

beta-cell dysfunction has been acknowledged as the key defect underlying the development of<br />

T2DM [90]. Beta cells can adapt to changes in insulin sensitivity by varying insulin secretion,<br />

thus maintaining euglycemia. During prolonged periods of reduced insulin sensitivity, e.g. in<br />

conditions such as pregnancy and obesity, beta cells may additionally meet this increased<br />

demand by expanding their volume through both hypertrophy and hyperplasia [91]. From the<br />

above, it is clear that if beta-cell function is assessed by measuring insulin secretion, this value<br />

needs to be corrected for prevailing glucose levels and insulin sensitivity.<br />

Since GCs induce occasional hyperglycemia, it was speculated that GCs, in addition to inducing<br />

insulin resistance, might exert an inhibitory effect on beta cells. From present literature it<br />

is apparent that the effects of GCs on beta-cell function are highly dependent of duration of<br />

exposure, dosage, and susceptibility of the population exposed.<br />

5.1 Glucocorticoids impair insulin secretion in vitro<br />

Several studies have assessed the effects of GCs on insulin secretion in vitro. Rodent-derived<br />

islets decreased insulin release in response to various GC compounds, both following acute<br />

exposure (minutes) [92,93] and following more prolonged exposure (hours to days) [8,94-<br />

99]. GCs may impair the pathways of both nonmetabolizable and metabolizable insulin<br />

secretagogues, most notably glucose.<br />

Glucose uptake and its oxidation in mitochondria of beta cells results in an increase in the<br />

adenosine triphosphate (ATP)/ adenosine monophosphate (ADP) ratio, closure of ATPsensitive<br />

potassium channels, plasma membrane depolarization, increased cytoplasmatic<br />

calcium concentrations, and finally exocytosis of insulin-containing granules, through<br />

yet incompletely understood pathways. In addition, calcium fluxes activate a number<br />

of potentiating signal pathways, including protein kinase A (PKA) en protein kinase C<br />

(PKC), which amplify insulin secretion [100]. GCs were shown to impair beta-cell glucose<br />

34


metabolism by reducing the expression levels of GLUT2 [95,101] and glucokinase (GK) [102],<br />

and to increase futile glucose cycling by enhancing G6Pase activity [102,103]. Through these<br />

mechanisms, GCs may reduce glucose uptake and phosphorylation, and thereby decrease ATP<br />

synthesis and calcium influx.<br />

Figure 3. Insulin secretory process in pancreatic beta cells and the proposed modes of interference<br />

by glucocorticoids. GCs induce beta-cell dysfunction by inhibiting several pathways. Most notably, GCs<br />

impair beta-cell glucose uptake and oxidation, decrease protein kinase A and C activation, and reduce<br />

calcium fluxes by permitting repolarizing potassium currents. Abbreviations: AC: adenyl cyclase; Ach:<br />

acetylcholine; ATP: adenosine triphosphate; cAMP: cyclic adenosine monophosphate; DAG: diacylglycerol;<br />

G6P: glucose-6-phosphatase; Gi : G-coupled inhibitory protein; GC: glucocorticoid; GK: glucokinase;<br />

GLUT2: glucose transporter 2; HK: hexokinase; IP3: inositol triphosphate; K 1,5: voltage-dependent K<br />

v<br />

channel; PIP2: phosphatidylinositol biphosphate; PKA: protein kinase A; PKC: protein kinase C; PLC:<br />

phospholipase C; SGK-1: serum- and glucocorticoid inducible kinase-1.<br />

Other than impairing β-oxidation of glucose in beta-cells, additional GC-induced effects<br />

may contribute to beta-cell dysfunction. Dexamethasone was shown to augment inward,<br />

repolarizing potassium currents by upregulating K ion channels, through activation of the<br />

v<br />

serum- and glucocorticoid inducible kinase (SGK)-1 [98]. These repolarizing currents may<br />

limit calcium influx and insulin secretion. Furthermore, GCs inhibited more downstream steps<br />

in the insulin secretory pathway. In isolated rat islets, dexamethasone decreased the activation<br />

35<br />

2<br />

Chapter 2


Chapter 2 Glucocorticoids: metabolic side effects<br />

of PKC through inhibition of the DAG-phospholipase C (PLC) pathway [99]. Moreover, GCs<br />

increased the expression of a adrenergic receptors [99,104], leading to reduced cyclic<br />

2<br />

adenosine monophosphate (cAMP) levels, PKA activity and subsequent decreased insulin<br />

release. These distal sites of interference by GCs in the insulin secretory process may explain<br />

the wide range of non-glucose insulin secretagogues that are also inhibited by GCs, including<br />

ketoisocaproate [8,93], the amino acid arginine [8], the sulfonylurea drugs tolbutamide [8]<br />

and gliclazide [93], and the phorbolester PMA, which activates PKC [93,99]. In line with these<br />

results, cAMP, a potent activator of PKA, was shown to prevent GC-induced effects on insulin<br />

secretion [8]. In addition to perturbing the process of insulin secretion, GCs may also decrease<br />

insulin biosynthesis by reducing the ATP/ADP ratio [96,105] and may reduce beta-cell mass<br />

by inducing apoptosis [106].<br />

To summarize, GCs interfere with the signaling pathways of various insulin secretagogues,<br />

but the exact mechanisms through which this occurs, remain largely unknown, despite major<br />

research efforts. Figure 3 shows a schematic overview of the here described insulin secretion<br />

process and the proposed modes of interference by GCs.<br />

5.2 Glucocorticoids effects on insulin secretion ex vivo and in vivo in rodents<br />

The effects of GCs on insulin secretion have also been assessed ex vivo and in vivo in rodents,<br />

however, establishing direct effects of GCs on beta cells under these conditions may be more<br />

challenging, since systemic metabolic consequences of GC treatment (e.g. changes in levels<br />

of glucose, NEFA and other metabolites) will interfere with GC-mediated changes in beta-cell<br />

function as well.<br />

Hydrocortisone treatment acutely inhibited insulin secretion in mice, resulting in hyperglycemia<br />

following an intravenous glucose challenge [107]. However, pancreatic islets isolated from<br />

healthy rats after more prolonged treatment with dexamethasone or hydrocortisone (several<br />

days), showed both unchanged and increased glucose-stimulated insulin secretion (GSIS)<br />

[101,108], most likely to compensate for GC-induced insulin resistance. The increases in GSIS<br />

were accompanied by expansion of beta-cell volume [101,109]. However, if dexamethasone<br />

treatment induced fasting hyperglycemia in these animals, GSIS was impaired [101]. Similar<br />

results were obtained in rodent models of obesity and insulin resistance. In Zucker fatty rats<br />

(fa/fa) [101,110] and ob/ob mice [103] dexamethasone readily induced hyperglycemia and<br />

markedly reduced or completely abrogated GSIS. In rats with streptozocin-induced diabetes,<br />

dexamethasone further increased fasting hyperglycemia and diminished GSIS [111].<br />

Transgenic mice overexpressing the GR in beta cells under control of the insulin promoter<br />

were glucose intolerant, due to blunted insulin secretion as compared to wildtype mice [112].<br />

36


At older age, these mice developed manifest diabetes, which was attributed to progressive<br />

loss of beta-cell function due to higher expression of a -adrenergic receptors, while insulin<br />

2<br />

sensitivity remained intact [113].<br />

5.3 Assessment of glucocorticoid-induced beta-cell dysfunction in humans<br />

5.3.1 Acute inhibitory effects of glucocorticoids on insulin secretion in vivo<br />

Both cortisol infusion [47] and acute treatment with high-doses of oral prednisolone<br />

[114,115] acutely impaired insulin secretion in healthy volunteers. In these studies, fasting<br />

insulin levels remained unchanged following GC treatment, despite the increments in fasting<br />

glucose. Furthermore, insulin release during glucose infusion [114] and during a meal test<br />

[115] were decreased, suggesting an acute inhibitory effect on the beta-cell.<br />

5.3.2 Glucocorticoids increase insulin secretion after short-term treatment<br />

More prolonged exposure (2 to 5 days) of healthy subjects to high doses of dexamethasone<br />

or prednisolone, resulted in fasting hyperinsulinemia and increased insulin secretion<br />

during hyperglycemic clamp studies or intravenous glucose tolerance tests [9,20,44,116-<br />

118]. In these studies, healthy subjects were able to compensate for GC-induced insulin<br />

resistance. Consequently, the product of insulin secretion and insulin sensitivity, also called<br />

the disposition index, remained constant. However, in susceptible populations, including<br />

normoglycemic individuals with a reduced insulin sensitivity [9] or a low GSIS [44,117]<br />

before treatment with GCs, healthy, first-degree relatives of patients with T2DM [20] and<br />

obese women [119], this compensation failed, resulting in hyperglycemia. These studies were<br />

limited by the fact that beta-cell function was assessed by intravenous glucose tolerance tests.<br />

Although the hyperglycemic clamp is regarded as the gold standard for the quantification<br />

of insulin secretion, it represents a less physiological condition relative to tests using orally<br />

administered insulin secretagogues, since insulin secretion also depends on, among-others,<br />

non-glucose metabolic stimuli, incretins, and neuronal inputs [120]. Recently, new methods<br />

have been validated to determine various aspects of beta-cell function from a frequentlysampled<br />

OGTT or from standardized mixed-meal tests, by using modeling analysis [121].<br />

Using this approach, GCs were recently shown to induce beta-cell dysfunction in completely<br />

healthy males, indicating that the full scope of GC-induced effects on beta-cell function may as<br />

yet not be fully detailed [115].<br />

5.3.3 Chronic effects of glucocorticoid-treatment on beta-cell function<br />

The most important limitation of most studies performed in humans, however, is the short<br />

exposure (maximally up to 14 days) to GCs. In daily practice, many patients are treated with<br />

37<br />

2<br />

Chapter 2


Chapter 2 Glucocorticoids: metabolic side effects<br />

GCs for prolonged time periods, even years. However, due to ethical issues, it is not viable to<br />

chronically expose healthy volunteers to GCs in order to study the long-term effects on betacell<br />

function. Conversely, it is not feasible to study the prolonged effects of GCs on beta-cell<br />

function in patients using GCs in clinical practice. Since the indication for GC treatment often<br />

comprises diseases with a state of systemic inflammation, inflammation-induced beta-cell<br />

effects will interfere with GC-modulation of beta-cell function [122].<br />

As chronic GC-treatment is associated with the development of diabetes, GCs most likely exert<br />

inhibitory effects on the beta cell after prolonged exposure, although strong (direct) evidence<br />

is currently lacking. As described previously, GC-induced pro-apoptotic effects and reductions<br />

in insulin biosynthesis may contribute to beta-cell failure in time. In addition, chronic GCtreatment<br />

may induce beta-cell failure indirectly through elevation of plasma concentrations<br />

of TG and NEFA [5,6], which contribute to beta-cell dysfunction by mechanisms related<br />

to lipotoxicity, a process which is characterised by accumulation of NEFA in the pancreas,<br />

resulting in beta-cell dysfunction [123].<br />

Taken together, GCs acutely inhibit insulin secretion, both in vitro and in vivo. Prolonged<br />

exposure however, induces hyperinsulinemia, due to compensation for GC-induced insulin<br />

resistance. Chronic exposure likely results in progressive loss of beta-cell function in<br />

susceptible individuals.<br />

6. ENDoGENoUS GLUCoCoR<strong>TI</strong>CoID METABoLISM IN THE<br />

METABoLIC SyNDRoME<br />

Since conditions with systemic GC excess shear great similarities with the MetS, the role<br />

of (tissue-specific) GC dysmetabolism in the latter condition has been the topic of intense<br />

research in the past decade. Most notably, the 11β-HSD enzymes and their relation with the<br />

development of central adiposity and hepatic steatosis have been investigated [12,14,70,89].<br />

6.1 Altered 11β-HSD activity in adipose tissue induces central adiposity<br />

In several studies in rodent obesity models, strong evidence for disturbed GC metabolism by<br />

an altered activity of 11β-HSD1 was observed. Transgenic mice selectively overexpressing<br />

11β-HSD1 in white adipose tissue under control of the AP2 promoter, thereby increasing local<br />

cortisol concentrations, developed visceral obesity, dyslipidemia and insulin resistance [124].<br />

In contrast, 11β-HSD1 (-/-) mice gained significantly less weight in response to a high-fat diet<br />

and preferentially stored adipose tissue in peripheral fat depots [125]. In addition, transgenic<br />

38


mice with adipose tissue-specific 11β-HSD2 overexpression, resisted weight gain on high-fat<br />

diet, and displayed improved insulin sensitivity compared to wildtype mice [15]. These data<br />

support the concept that reducing GC activity in adipose tissue has favourable effects on body<br />

composition and metabolic profile. Translating these promising data to humans, however, has<br />

proven to be difficult. In obese subjects, increased 11β-HSD1mRNA and activity were shown<br />

in SCAT [126,127], rather than in VAT [128], and increased cortisol metabolite excretion was<br />

associated with total body fat content, but not specifically with VAT area [67].<br />

6.2 Glucocorticoid dysmetabolism in the liver is linked with hepatic steatosis<br />

Hepatic steatosis represents a pathophysiological hallmark of the MetS, and altered hepatic<br />

cortisol metabolism has been linked to this condition [40]. Again, particularly the enzyme<br />

11β-HSD1 has received much attention. In transgenic mice, it was elegantly demonstrated that<br />

liver-specific overexpression of 11β-HSD1 induced fatty liver by increasing the lipogenesis/<br />

lipid oxidation ratio, without increasing abdominal fat depots [129]. However, studies in<br />

humans linking 11β-HSD1 activity to hepatic fat content were less successful [67].<br />

In conclusion, involvement of GC metabolism in obesity and its metabolic consequences<br />

seems evident, although translating data from animal studies in humans has proven to be<br />

difficult. Nevertheless, given its important role in GC metabolism and disturbed activity of<br />

this enzyme in other insulin resistant states, such as polycystic ovary syndrome [130], HIVassociated<br />

lipodystrophy [131] or cortisone reductase deficiency [132], 11β-HSD1 remains a<br />

promising target for novel therapeutics, as will be discussed below.<br />

7. NoVEL THERAPEU<strong>TI</strong>CS RELATED To GLUCoCoR<strong>TI</strong>CoID<br />

METABoLISM AND AC<strong>TI</strong>oN<br />

Advances in research regarding GC metabolism and mode of action, has led to the development<br />

of two novel classes of agents, albeit it for different indications, i.e. the dissociated GR agonists<br />

and the 11β-HSD1 inhibitors.<br />

7.1 Dissociated glucocorticoid receptor agonists<br />

This new class of synthetic GR ligands is designed to specifically induce GC-induced<br />

transrepression pathways, while minimizing transactivation activity. As described earlier,<br />

and as demonstrated in figure 1, these compounds are thus expected to display the antiinflammatory<br />

properties of classic GR agonists, such as prednisolone and dexamethasone,<br />

but without inducing dysmetabolic side effects [16,17]. Both pharmaceutical companies and<br />

university groups have made great efforts to identify dissociated GR ligands with an increased<br />

39<br />

2<br />

Chapter 2


Chapter 2 Glucocorticoids: metabolic side effects<br />

therapeutic index (reviewed in [133]).<br />

The first compounds, which still had a steroidal structure, showed good dissociation in<br />

vitro, however, their beneficial profile was not sustained in vivo. Since then, several non-<br />

steroidal GR-ligands have been identified. A number of these compounds, such as AL-438, ZK<br />

216348 and BI115, indeed demonstrated an increased therapeutic index in vivo in different<br />

disease- and side effect animal models. These GR ligands were shown to be as efficient as<br />

prednisolone and dexamethasone to suppress inflammation, but they did not elevate blood<br />

glucose levels [133]. Although all dissociated GR ligands are currently still at the preclinical<br />

level of development, and many aspects of the molecular pathways that underlie GC’s actions<br />

remain to be elucidated, it is evident that these compounds may become of great value for<br />

patients who require treatment with the classic GR agonists. It will be exciting to see whether<br />

the promising results derived from in vitro assays and animal models will hold in studies in<br />

humans that are expected to be conducted in the coming years.<br />

7.2 11β-HSD type 1 inhibitors<br />

Given the role of increased GC activity in liver and adipose tissue in the development of<br />

obesity and the MetS, 11β-HSD1 inhibition has been proposed as an interesting strategy for<br />

the treatment of these conditions. Indeed, in obese rodents, pharmacological inhibition of<br />

11β-HSD1 improved glucose tolerance, insulin sensitivity and lipid profiles [15]. In humans,<br />

the first studies were performed with carbenoxolone, a nonselective 11β-HSD1 inhibitor.<br />

Although carbenoxolone decreased hepatic glucose production during a hyperinsulinemiceuglycemic<br />

clamp, it did not augment glucose disposal [70]. Several new classes of selective<br />

11β-HSD1 inhibitors are currently in development, including arylsulfonamidothiazoles [134]<br />

and perhydroquinolylbenzamides [135]. Although these agents were shown to lower blood<br />

glucose concentrations by increasing hepatic insulin sensitivity, clamp studies in obese mice<br />

demonstrated no beneficial effects on glucose disposal [136]. Most recently, new compounds<br />

with benzamide [137] and thiazole [138] structures have been published. The important<br />

challenge now is to assess whether these compounds (and other future 11β-HSD1 inhibitors),<br />

in addition to improving hepatic insulin sensitivity, exert beneficial effects on adipose tissue<br />

and whole-body insulin sensitivity in humans. Despite the limitations of the current available<br />

11β-HSD1 inhibitors, 11β-HSD1 remains an attractive target for the development of novel<br />

drugs that may be added to the present arsenal of therapeutics in the combat against the MetS<br />

pandemic.<br />

Funding: This paper was written within the framework of project T1-106 of the Dutch Top<br />

Institute <strong>Pharma</strong>.<br />

40


REFERENCES<br />

1. McMahon M, Gerich j, Rizza R. Effects of glucocorticoids on carbohydrate metabolism. Diabetes<br />

Metab Rev. 1988;4(1):17-30.<br />

2. Schacke H, Docke WD, Asadullah K. Mechanisms involved in the side effects of glucocorticoids.<br />

<strong>Pharma</strong>col Ther. 2002;96(2):23-43.<br />

3. Rockall AG, Sohaib SA, Evans D, Kaltsas G, Isidori AM, Monson jP, Besser GM, Grossman AB, Reznek<br />

RH. Computed tomography assessment of fat distribution in male and female patients with Cushing’s<br />

syndrome. Eur j Endocrinol. 2003;149(6):561-7.<br />

4. Rockall AG, Sohaib SA, Evans D, Kaltsas G, Isidori AM, Monson jP, Besser GM, Grossman AB, Reznek<br />

RH. Hepatic steatosis in Cushing’s syndrome: a radiological assessment using computed tomography.<br />

Eur j Endocrinol. 2003;149(6):543-8.<br />

5. Taskinen MR, Nikkila EA, Pelkonen R, Sane T. Plasma lipoproteins, lipolytic enzymes, and very<br />

low density lipoprotein triglyceride turnover in Cushing’s syndrome. J Clin Endocrinol Metab.<br />

1983;57(3):619-26.<br />

6. Wajchenberg BL. Subcutaneous and visceral adipose tissue: their relation to the metabolic<br />

syndrome. Endocr Rev. 2000;21(6):697-738.<br />

7. Khaleeli AA, Edwards RH, Gohil K, McPhail G, Rennie Mj, Round j, Ross Ej. Corticosteroid myopathy:<br />

a clinical and pathological study. Clin Endocrinol (Oxf). 1983;18(2):155-66.<br />

8. Lambillotte C, Gilon P, Henquin jC. Direct glucocorticoid inhibition of insulin secretion. An in vitro<br />

study of dexamethasone effects in mouse islets. j Clin Invest. 1997;99(3):414-23.<br />

9. Larsson H, Ahren B. Insulin resistant subjects lack islet adaptation to short-term dexamethasoneinduced<br />

reduction in insulin sensitivity. Diabetologia. 1999;42(8):936-43.<br />

10. Alberti KG, Zimmet P, Shaw J. The metabolic syndrome--a new worldwide definition. Lancet.<br />

2005;366(9491):1059-62.<br />

11. Reynolds RM, Walker BR, Syddall HE, Whorwood CB, Wood Pj, Phillips DI. Elevated plasma cortisol<br />

in glucose-intolerant men: differences in responses to glucose and habituation to venepuncture. j<br />

Clin Endocrinol Metab. 2001;86(3):1149-53.<br />

12. Seckl JR, Walker BR. Minireview: 11beta-hydroxysteroid dehydrogenase type 1- a tissue-specific<br />

amplifier of glucocorticoid action. Endocrinology. 2001;142(4):1371-6.<br />

13. Witchel SF, DeFranco DB. Mechanisms of disease: regulation of glucocorticoid and receptor levelsimpact<br />

on the metabolic syndrome. Nat Clin Pract Endocrinol Metab. 2006;2(11):621-31.<br />

14. Seckl jR. 11beta-hydroxysteroid dehydrogenases: changing glucocorticoid action. Curr Opin<br />

<strong>Pharma</strong>col. 2004;4(6):597-602.<br />

15. Kershaw EE, Morton NM, Dhillon H, Ramage L, Seckl JR, Flier JS. Adipocyte-specific glucocorticoid<br />

inactivation protects against diet-induced obesity. Diabetes. 2005;54(4):1023-31.<br />

41<br />

2<br />

Chapter 2


Chapter 2 Glucocorticoids: metabolic side effects<br />

16. Rosen j, Miner jN. The search for safer glucocorticoid receptor ligands. Endocr Rev. 2005;26(3):452-<br />

42<br />

64.<br />

17. Schacke H, Rehwinkel H, Asadullah K, Cato AC. Insight into the molecular mechanisms of<br />

glucocorticoid receptor action promotes identification of novel ligands with an improved therapeutic<br />

index. Exp Dermatol. 2006;15(8):565-73.<br />

18. Walker BR. Glucocorticoids and cardiovascular disease. Eur j Endocrinol. 2007;157(5):545-59.<br />

19. DeFronzo RA, jacot E, jequier E, Maeder E, Wahren j, Felber jP. The effect of insulin on the disposal<br />

of intravenous glucose. Results from indirect calorimetry and hepatic and femoral venous<br />

catheterization. Diabetes. 1981;30(12):1000-7.<br />

20. Henriksen jE, Alford F, Ward GM, Beck-Nielsen H. Risk and mechanism of dexamethasone-induced<br />

deterioration of glucose tolerance in non-diabetic first-degree relatives of NIDDM patients.<br />

Diabetologia. 1997;40(12):1439-48.<br />

21. Rizza RA, Mandarino Lj, Gerich jE. Cortisol-induced insulin resistance in man: impaired suppression<br />

of glucose production and stimulation of glucose utilization due to a postreceptor detect of insulin<br />

action. j Clin Endocrinol Metab. 1982;54(1):131-8.<br />

22. Krebs M, Krssak M, Bernroider E, Anderwald C, Brehm A, Meyerspeer M, Nowotny P, Roth E,<br />

Waldhausl W, Roden M. Mechanism of amino acid-induced skeletal muscle insulin resistance in<br />

humans. Diabetes. 2002;51(3):599-605.<br />

23. Elks ML. Fat oxidation and diabetes of obesity: the Randle hypothesis revisited. Med Hypotheses.<br />

1990;33(4):257-60.<br />

24. Perseghin G, Petersen K, Shulman GI. Cellular mechanism of insulin resistance: potential links with<br />

inflammation. Int J Obes Relat Metab Disord. 2003;27 Suppl 3:S6-11.<br />

25. Ewart HS, Somwar R, Klip A. Dexamethasone stimulates the expression of GLUT1 and GLUT4<br />

proteins via different signaling pathways in L6 skeletal muscle cells. FEBS Lett. 1998;421(2):120-4.<br />

26. Long W, Barrett Ej, Wei L, Liu Z. Adrenalectomy enhances the insulin sensitivity of muscle protein<br />

synthesis. Am j Physiol Endocrinol Metab 2003;284(1):E102-9.<br />

27. Ruzzin j, Wagman AS, jensen j. Glucocorticoid-induced insulin resistance in skeletal muscles: defects<br />

in insulin signalling and the effects of a selective glycogen synthase kinase-3 inhibitor. Diabetologia.<br />

2005;48(10):2119-30.<br />

28. Saad Mj, Folli F, Kahn jA, Kahn CR. Modulation of insulin receptor, insulin receptor substrate-1,<br />

and phosphatidylinositol 3-kinase in liver and muscle of dexamethasone-treated rats. j Clin Invest.<br />

1993;92(4):2065-72.<br />

29. Weinstein SP, Wilson CM, Pritsker A, Cushman SW. Dexamethasone inhibits insulin-stimulated<br />

recruitment of GLUT4 to the cell surface in rat skeletal muscle. Metabolism. 1998;47(1):3-6.<br />

30. Pagano G, Cavallo-Perin P, Cassader M, Bruno A, Ozzello A, Masciola P et al. An in vivo and in vitro<br />

study of the mechanism of prednisone-induced insulin resistance in healthy subjects. j Clin Invest.<br />

1983;72(5):1814-20.


31. Henriksen jE, Alford F, Vaag A, Handberg A, Beck-Nielsen H. Intracellular skeletal muscle glucose<br />

metabolism is differentially altered by dexamethasone treatment of normoglycemic relatives of type<br />

2 diabetic patients. Metabolism. 1999;48(9):1128-35.<br />

32. Ekstrand A, Schalin-jantti C, Lofman M, Parkkonen M, Widen E, Franssila-Kallunki A, Saloranta<br />

C, Koivisto, Groop L. The effect of (steroid) immunosuppression on skeletal muscle glycogen<br />

metabolism in patients after kidney transplantation. Transplantation. 1996;61(6):889-93.<br />

33. Lofberg E, Gutierrez A, Wernerman J, Anderstam B, Mitch WE, Price SR, Bergstrom J, Alvestrand<br />

A. Effects of high doses of glucocorticoids on free amino acids, ribosomes and protein turnover in<br />

human muscle. Eur j Clin Invest. 2002;32(5):345-53.<br />

34. Patti ME, Brambilla E, Luzi L, Landaker Ej, Kahn CR. Bidirectional modulation of insulin action by<br />

amino acids. j Clin Invest 1998;101(7):1519-29.<br />

35. Schakman O, Gilson H, Thissen jP. Mechanisms of glucocorticoid-induced myopathy. j Endocrinol.<br />

2008;197(1):1-10.<br />

36. Dinneen S, Alzaid A, Miles j, Rizza R. Effects of the normal nocturnal rise in cortisol on carbohydrate<br />

and fat metabolism in IDDM. Am j Physiol. 1995;268(4 Pt 1):E595-603.<br />

37. Boden G, Shulman GI. Free fatty acids in obesity and type 2 diabetes: defining their role in the<br />

development of insulin resistance and beta-cell dysfunction. Eur J Clin Invest 2002;32 Suppl 3:14-23.<br />

38. Randle Pj, Garland PB, Hales CN, Newsholme EA. The glucose fatty-acid cycle. Its role in insulin<br />

sensitivity and the metabolic disturbances of diabetes mellitus. Lancet. 1963;1(7285):785-9.<br />

39. Barthel A, Schmoll D. Novel concepts in insulin regulation of hepatic gluconeogenesis. Am j Physiol<br />

Endocrinol Metab. 2003;285(4):E685-92.<br />

40. Andrews RC, Walker BR. Glucocorticoids and insulin resistance: old hormones, new targets. Clin Sci<br />

(Lond) 1999;96(5):513-23.<br />

41. Nielsen MF, Caumo A, Chandramouli V, Schumann WC, Cobelli C, Landau BR, Vilstrup H, Rizza<br />

RA, Schmitz O. Impaired basal glucose effectiveness but unaltered fasting glucose release and<br />

gluconeogenesis during short-term hypercortisolemia in healthy subjects. Am j Physiol Endocrinol<br />

Metab. 2004;286(1):E102-E110.<br />

42. Rooney DP, Neely RD, Cullen C, Ennis CN, Sheridan B, Atkinson AB, Trimble ER, Bell PM. The effect of<br />

cortisol on glucose/glucose-6-phosphate cycle activity and insulin action. j Clin Endocrinol Metab.<br />

1993;77(5):1180-3.<br />

43. Vegiopoulos A, Herzig S. Glucocorticoids, metabolism and metabolic diseases. Mol Cell Endocrinol.<br />

2007;275(1-2):43-61.<br />

44. Wajngot A, Giacca A, Grill V, Vranic M, Efendic S. The diabetogenic effects of glucocorticoids are more<br />

pronounced in low- than in high-insulin responders. Proc Natl Acad Sci U S A. 1992;89(13):6035-9.<br />

45. Miyoshi H, Shulman GI, Peters Ej, Wolfe MH, Elahi D, Wolfe RR. Hormonal control of substrate cycling<br />

in humans. j Clin Invest. 1988;81(5):1545-55.<br />

43<br />

2<br />

Chapter 2


Chapter 2 Glucocorticoids: metabolic side effects<br />

46. Schneiter P, Tappy L. Kinetics of dexamethasone-induced alterations of glucose metabolism in<br />

healthy humans. Am j Physiol. 1998;275(5 Pt 1):E806-13.<br />

47. Shamoon H, Soman V, Sherwin RS. The influence of acute physiological increments of cortisol on<br />

fuel metabolism and insulin binding to monocytes in normal humans. j Clin Endocrinol Metab.<br />

1980;50(3):495-501.<br />

48. Wajngot A, Khan A, Giacca A, Vranic M, Efendic S. Dexamethasone increases glucose cycling, but not<br />

glucose production, in healthy subjects. Am j Physiol. 1990;259(5 Pt 1):E626-32.<br />

49. jin jY, Dubois DC, Almon RR, jusko Wj. Receptor/gene-mediated pharmacodynamic effects of<br />

methylprednisolone on phosphoenolpyruvate carboxykinase regulation in rat liver. j <strong>Pharma</strong>col Exp<br />

Ther. 2004;309(1):328-39.<br />

50. Sasaki K, Cripe TP, Koch SR, Andreone TL, Petersen DD, Beale EG et al. Multihormonal regulation of<br />

phosphoenolpyruvate carboxykinase gene transcription. The dominant role of insulin. j Biol Chem.<br />

1984;259(24):15242-51.<br />

51. Vander Kooi BT, Onuma H, Oeser JK, Svitek CA, Allen SR, Vander Kooi CW, Chazin WJ, O’Brien RM.<br />

The glucose-6-phosphatase catalytic subunit gene promoter contains both positive and negative<br />

glucocorticoid response elements. Mol Endocrinol. 2005;19(12):3001-22.<br />

52. Zimmerman T, Horber F, Rodriguez N, Schwenk WF, Haymond MW. Contribution of insulin resistance<br />

44<br />

to catabolic effect of prednisone on leucine metabolism in humans. Diabetes. 1989;38(10):1238-44.<br />

53. Kraus-Friedmann N. Hormonal regulation of hepatic gluconeogenesis. Physiol Rev. 1984;64(1):170-<br />

259.<br />

54. Whorwood CB, Donovan Sj, Flanagan D, Phillips DI, Byrne CD. Increased glucocorticoid receptor<br />

expression in human skeletal muscle cells may contribute to the pathogenesis of the metabolic<br />

syndrome. Diabetes. 2002;51(4):1066-75.<br />

55. Chevalier S, Burgess SC, Malloy CR, Gougeon R, Marliss EB, Morais jA. The greater contribution<br />

of gluconeogenesis to glucose production in obesity is related to increased whole-body protein<br />

catabolism. Diabetes. 2006;55(3):675-81.<br />

56. Dirlewanger M, Schneiter PH, Paquot N, jequier E, Rey V, Tappy L. Effects of glucocorticoids on<br />

hepatic sensitivity to insulin and glucagon in man. Clin Nutr. 2000;19(1):29-34.<br />

57. Bernal-Mizrachi C, Weng S, Feng C, Finck BN, Knutsen RH, Leone TC, Coleman T, Mecham RP, Kelly DP,<br />

Semenkovich CF. Dexamethasone induction of hypertension and diabetes is PPAR-alpha dependent<br />

in LDL receptor-null mice. Nat Med 2003;9(8):1069-75.<br />

58. Bernal-Mizrachi C, Xiaozhong L, Yin L, Knutsen RH, Howard Mj, Arends jj, Desantis P, Colemant<br />

T, Semenkovich CF. An afferent vagal nerve pathway links hepatic PPARalpha activation to<br />

glucocorticoid-induced insulin resistance and hypertension. Cell Metab. 2007;5(2):91-102.<br />

59. Van Raalte DH, Li M, Pritchard PH, Wasan KM. Peroxisome proliferator-activated receptor (PPAR)alpha:<br />

a pharmacological target with a promising future. Pharm Res. 2004;21(9):1531-8.


60. Opherk C, Tronche F, Kellendonk C, Kohlmuller D, Schulze A, Schmid W, Schutz G. Inactivation of the<br />

glucocorticoid receptor in hepatocytes leads to fasting hypoglycemia and ameliorates hyperglycemia<br />

in streptozotocin-induced diabetes mellitus. Mol Endocrinol. 2004;18(6):1346-53.<br />

61. Parekh S, Anania FA. Abnormal lipid and glucose metabolism in obesity: implications for nonalcoholic<br />

fatty liver disease. Gastroenterology. 2007;132(6):2191-207.<br />

62. Giudetti AM, Gnoni GV. Short-term effect of dexamethasone on fatty acid and cholesterol synthesis<br />

in isolated rat hepatocytes. Biochem Mol Biol Int. 1998;44(3):515-21.<br />

63. Cole TG, Wilcox HG, Heimberg M. Effects of adrenalectomy and dexamethasone on hepatic lipid<br />

metabolism. j Lipid Res. 1982;23(1):81-91.<br />

64. Shteyer E, Liao Y, Muglia Lj, Hruz PW, Rudnick DA. Disruption of hepatic adipogenesis is associated<br />

with impaired liver regeneration in mice. Hepatology. 2004;40(6):1322-32.<br />

65. Rockall AG, Sohaib SA, Evans D, Kaltsas G, Isidori AM, Monson jP, Besser GM, Grossman AB, Reznek<br />

RH. Hepatic steatosis in Cushing’s syndrome: a radiological assessment using computed tomography.<br />

Eur j Endocrinol. 2003;149(6):543-8.<br />

66. Itoh S, Igarashi M, Tsukada Y, Ichinoe A. Nonalcoholic fatty liver with alcoholic hyalin after long-term<br />

glucocorticoid therapy. Acta Hepatogastroenterol (Stuttg). 1977;24(6):415-8.<br />

67. Westerbacka j, Yki-jarvinen H, Vehkavaara S, Hakkinen AM, Andrew R, Wake Dj, Seckl jR, Walker<br />

BR. Body fat distribution and cortisol metabolism in healthy men: enhanced 5beta-reductase<br />

and lower cortisol/cortisone metabolite ratios in men with fatty liver. j Clin Endocrinol Metab.<br />

2003;88(10):4924-31.<br />

68. Ettinger WH, jr., Hazzard WR. Prednisone increases very low density lipoprotein and high density<br />

lipoprotein in healthy men. Metabolism. 1988;37(11):1055-8.<br />

69. Taskinen MR, Kuusi T, Yki-jarvinen H, Nikkila EA. Short-term effects of prednisone on serum lipids<br />

and high density lipoprotein subfractions in normolipidemic healthy men. j Clin Endocrinol Metab.<br />

1988;67(2):291-9.<br />

70. Morton NM, Seckl jR. 11beta-hydroxysteroid dehydrogenase type 1 and obesity. Front Horm Res.<br />

2008;36:146-64.<br />

71. Bjorntorp P. Visceral fat accumulation: the missing link between psychosocial factors and<br />

cardiovascular disease? j Intern Med. 1991;230(3):195-201.<br />

72. Whitworth jA, Williamson PM, Mangos G, Kelly jj. Cardiovascular consequences of cortisol excess.<br />

Vasc Health Risk Manag. 2005;1(4):291-9.<br />

73. Kershaw EE, Flier jS. Adipose tissue as an endocrine organ. j Clin Endocrinol Metab. 2004;89(6):2548-<br />

56.<br />

74. Fasshauer M, Paschke R. Regulation of adipocytokines and insulin resistance. Diabetologia.<br />

2003;46(12):1594-603.<br />

45<br />

2<br />

Chapter 2


Chapter 2 Glucocorticoids: metabolic side effects<br />

75. Lundgren M, Buren j, Ruge T, Myrnas T, Eriksson jW. Glucocorticoids down-regulate glucose uptake<br />

46<br />

capacity and insulin-signaling proteins in omental but not subcutaneous human adipocytes. j Clin<br />

Endocrinol Metab. 2004;89(6):2989-97.<br />

76. Turnbow MA, Keller SR, Rice KM, Garner CW. Dexamethasone down-regulation of insulin receptor<br />

substrate-1 in 3T3-L1 adipocytes. j Biol Chem 1994;269(4):2516-20.<br />

77. Sakoda H, Ogihara T, Anai M, Funaki M, Inukai K, Katagiri H, Fukushima Y, Onishi Y, Ono H, Fujishiro M,<br />

Kikushi M, Oka Y, Asano T. Dexamethasone-induced insulin resistance in 3T3-L1 adipocytes is due to<br />

inhibition of glucose transport rather than insulin signal transduction. Diabetes. 2000;49(10):1700-<br />

8.<br />

78. Fasshauer M, Paschke R. Regulation of adipocytokines and insulin resistance. Diabetologia.<br />

2003;46(12):1594-603.<br />

79. Patel jV, Cummings DE, Girod jP, Mascarenhas AV, Hughes EA, Gupta M, Zavanone M, Beck-Peccoz P,<br />

Spada A. Role of metabolically active hormones in the insulin resistance associated with short-term<br />

glucocorticoid treatment. j Negat Results Biomed. 2006;11(5):14.<br />

80. Lewandowski KC, Szosland K, Lewinski A. Short-term dexamethasone administration does not alter<br />

serum adiponectin or resistin concentrations in overweight and obese subjects despite an increase<br />

in insulin resistance. Clin Endocrinol (Oxf). 2006;65(4):551-2.<br />

81. Libe R, Morpurgo PS, Cappiello V, Maffini A, Bondioni S, Locatelli M et al. Ghrelin and adiponectin in<br />

patients with Cushing’s disease before and after successful transsphenoidal surgery. Clin Endocrinol<br />

(Oxf). 2005;62(1):30-6.<br />

82. Udden j, Bjorntorp P, Arner P, Barkeling B, Meurling L, Rossner S. Effects of glucocorticoids on leptin<br />

levels and eating behaviour in women. j Intern Med. 2003;253(2):225-31.<br />

83. Slavin BG, Ong jM, Kern PA. Hormonal regulation of hormone-sensitive lipase activity and mRNA<br />

levels in isolated rat adipocytes. j Lipid Res. 1994;35(9):1535-41.<br />

84. Divertie GD, jensen MD, Miles jM. Stimulation of lipolysis in humans by physiological<br />

hypercortisolemia. Diabetes. 1991;40(10):1228-32.<br />

85. Samra jS, Clark ML, Humphreys SM, MacDonald IA, Bannister PA, Frayn KN. Effects of physiological<br />

hypercortisolemia on the regulation of lipolysis in subcutaneous adipose tissue. j Clin Endocrinol<br />

Metab. 1998;83(2):626-31.<br />

86. Djurhuus CB, Gravholt CH, Nielsen S, Mengel A, Christiansen jS, Schmitz OE, Moller N. Effects of<br />

cortisol on lipolysis and regional interstitial glycerol levels in humans. Am j Physiol Endocrinol<br />

Metab. 2002;283(1):E172-7.<br />

87. Ottosson M, Vikman-Adolfsson K, Enerback S, Olivecrona G, Bjorntorp P. The effects of cortisol<br />

on the regulation of lipoprotein lipase activity in human adipose tissue. j Clin Endocrinol Metab.<br />

1994;79(3):820-5.<br />

88. Arner P, Lithell H, Wahrenberg H, Bronnegard M. Expression of lipoprotein lipase in different human<br />

subcutaneous adipose tissue regions. j Lipid Res. 1991;32(3):423-9.


89. Seckl jR, Morton NM, Chapman KE, Walker BR. Glucocorticoids and 11beta-hydroxysteroid<br />

dehydrogenase in adipose tissue. Recent Prog Horm Res. 2004;59:359-93.<br />

90. Kahn SE. The relative contributions of insulin resistance and beta-cell dysfunction to the<br />

pathophysiology of Type 2 diabetes. Diabetologia. 2003;46(1):3-19.<br />

91. Weyer C, Bogardus C, Mott DM, Pratley RE. The natural history of insulin secretory dysfunction and<br />

insulin resistance in the pathogenesis of type 2 diabetes mellitus. j Clin Invest. 1999;104(6):787-94.<br />

92. Barseghian G, Levine R. Effect of corticosterone on insulin and glucagon secretion by the isolated<br />

perfused rat pancreas. Endocrinology. 1980;106(2):547-52.<br />

93. Billaudel B, Mathias PC, Sutter BC, Malaisse WJ. Inhibition by corticosterone of calcium inflow and<br />

insulin release in rat pancreatic islets. j Endocrinol. 1984;100(2):227-33.<br />

94. Brunstedt j, Nielsen jH. Direct long-term effect of hydrocortisone on insulin and glucagon release<br />

from mouse pancreatic islets in tissue culture. Acta Endocrinol (Copenh). 1981;96(4):498-504.<br />

95. Gremlich S, Roduit R, Thorens B. Dexamethasone induces posttranslational degradation of GLUT2<br />

and inhibition of insulin secretion in isolated pancreatic beta cells. Comparison with the effects of<br />

fatty acids. j Biol Chem. 1997;272(6):3216-22.<br />

96. jeong IK, Oh SH, Kim Bj, Chung jH, Min YK, Lee MS, Lee MK, Kim KW. The effects of dexamethasone<br />

on insulin release and biosynthesis are dependent on the dose and duration of treatment. Diabetes<br />

Res Clin Pract. 2001;51(3):163-71.<br />

97. Pierluissi j, Navas FO, Ashcroft Sj. Effect of adrenal steroids on insulin release from cultured rat islets<br />

of Langerhans. Diabetologia. 1986;29(2):119-21.<br />

98. Ullrich S, Berchtold S, Ranta F, Seebohm G, Henke G, Lupescu A, Mack AF, Chao CM, Su j, Nitschke<br />

R, Alexander D, Friedrich B, Wulff P, Kuld D, Lang F. Serum- and glucocorticoid-inducible kinase 1<br />

(SGK1) mediates glucocorticoid-induced inhibition of insulin secretion. Diabetes. 2005;54(4):1090-<br />

9.<br />

99. Zawalich WS, Tesz Gj, Yamazaki H, Zawalich KC, Philbrick W. Dexamethasone suppresses<br />

phospholipase C activation and insulin secretion from isolated rat islets. Metabolism. 2006;55(1):35-<br />

42.<br />

100. Nesher R, Anteby E, Yedovizky M, Warwar N, Kaiser N, Cerasi E. Beta-cell protein kinases and the<br />

dynamics of the insulin response to glucose. Diabetes. 2002;51 Suppl 1:S68-73.<br />

101. Ogawa A, johnson jH, Ohneda M, McAllister CT, Inman L, Alam T, Unger RH. Roles of insulin resistance<br />

and beta-cell dysfunction in dexamethasone-induced diabetes. j Clin Invest. 1992;90(2):497-504.<br />

102. Borboni P, Porzio O, Magnaterra R, Fusco A, Sesti G, Lauro R, Marlier LN. Quantitative analysis of<br />

pancreatic glucokinase gene expression in cultured beta cells by competitive polymerase chain<br />

reaction. Mol Cell Endocrinol. 1996;117(2):175-81.<br />

103. Khan A, Ostenson CG, Berggren PO, Efendic S. Glucocorticoid increases glucose cycling and inhibits<br />

insulin release in pancreatic islets of ob/ob mice. Am j Physiol. 1992;263(4 Pt 1):E663-6.<br />

47<br />

2<br />

Chapter 2


Chapter 2 Glucocorticoids: metabolic side effects<br />

104. Hamamdzic D, Duzic E, Sherlock jD, Lanier SM. Regulation of alpha 2-adrenergic receptor expression<br />

48<br />

and signaling in pancreatic beta-cells. Am J Physiol 1995;269(1Pt 1):E162-71.<br />

105. Philippe j, Missotten M. Dexamethasone inhibits insulin biosynthesis by destabilizing insulin<br />

messenger ribonucleic acid in hamster insulinoma cells. Endocrinology. 1990;127(4):1640-5.<br />

106. Ranta F, Avram D, Berchtold S, Dufer M, Drews G, Lang F, Ullrich S. Dexamethasone induces cell death<br />

in insulin-secreting cells, an effect reversed by exendin-4. Diabetes. 2006;55(5):1380-90.<br />

107. Longano CA, Fletcher HP. Insulin release after acute hydrocortisone treatment in mice. Metabolism.<br />

1983;32(6):603-8.<br />

108. Wang ZL, Bennet WM, Wang RM, Ghatei MA, Bloom SR. Evidence of a paracrine role of neuropeptide-Y<br />

in the regulation of insulin release from pancreatic islets of normal and dexamethasone-treated rats.<br />

Endocrinology. 1994;135(1):200-6.<br />

109. Mulder H, Ahren B, Stridsberg M, Sundler F. Non-parallelism of islet amyloid polypeptide (amylin)<br />

and insulin gene expression in rats islets following dexamethasone treatment. Diabetologia.<br />

1995;38(4):395-402.<br />

110. Ohneda M, johnson jH, Inman LR, Unger RH. GLUT-2 function in glucose-unresponsive beta cells of<br />

dexamethasone-induced diabetes in rats. j Clin Invest. 1993;92(4):1950-6.<br />

111. Grill V, Rundfeldt M. Abnormalities of insulin responses after ambient and previous exposure to<br />

glucose in streptozocin-diabetic and dexamethasone-treated rats. Role of hyperglycemia and<br />

increased B-cell demands. Diabetes. 1986;35(1):44-51.<br />

112. Delaunay F, Khan A, Cintra A, Davani B, Ling ZC, Andersson A, Ostenson CG, Gustafsson j, Efendic S,<br />

Okret S. Pancreatic beta cells are important targets for the diabetogenic effects of glucocorticoids. j<br />

Clin Invest. 1997;100(8):2094-8.<br />

113. Davani B, Portwood N, Bryzgalova G, Reimer MK, Heiden T, Ostenson CG, Okret S, Ahren B, Efendic<br />

S, Khan A. Aged transgenic mice with increased glucocorticoid sensitivity in pancreatic beta-cells<br />

develop diabetes. Diabetes. 2004;53 Suppl 1:S51-9.<br />

114. Kalhan SC, Adam PA. Inhibitory effect of prednisone on insulin secretion in man: model for<br />

duplication of blood glucose concentration. j Clin Endocrinol Metab. 1975;41(3):600-10.<br />

115. Van Raalte DH, Mari A, Bunck MC, Nofrate V, Dokter W, Nassander U et al. Acute and 2-week exposure<br />

to prednisolone impair different aspects of beta-cell function in healthy men. Eur j Endocrinol.<br />

2010;162(4):729-35.<br />

116. Beard jC, Halter jB, Best jD, Pfeifer MA, Porte D, jr. Dexamethasone-induced insulin resistance<br />

enhances B cell responsiveness to glucose level in normal men. Am j Physiol. 1984;247(5 Pt<br />

1):E592-6.<br />

117. Grill V, Pigon j, Hartling SG, Binder C, Efendic S. Effects of dexamethasone on glucose-induced insulin<br />

and proinsulin release in low and high insulin responders. Metabolism. 1990;39(3):251-8.


118. Matsumoto K, Yamasaki H, Akazawa S, Sakamaki H, Ishibashi M, Abiru N, Uotani S, Matsuo H,<br />

Yamaguchi Y, Tokuyama K, Nagataki S. High-dose but not low-dose dexamethasone impairs glucose<br />

tolerance by inducing compensatory failure of pancreatic beta-cells in normal men. j Clin Endocrinol<br />

Metab. 1996;81(7):2621-6.<br />

119. Besse C, Nicod N, Tappy L. Changes in insulin secretion and glucose metabolism induced by<br />

dexamethasone in lean and obese females. Obes Res. 2005;13(2):306-11.<br />

120. Cobelli C, Toffolo GM, Dalla Man C, Campioni M, Denti P, Caumo A, Butler P, Rizza R. Assessment of<br />

beta-cell function in humans, simultaneously with insulin sensitivity and hepatic extraction, from<br />

intravenous and oral glucose tests. Am j Physiol Endocrinol Metab. 2007;293(1):E1-15.<br />

121. Mari A, Schmitz O, Gastaldelli A, Oestergaard T, Nyholm B, Ferrannini E. Meal and oral glucose tests<br />

for assessment of beta -cell function: modeling analysis in normal subjects. Am j Physiol Endocrinol<br />

Metab. 2002;283(6):E1159-66.<br />

122. Dessein PH, joffe BI. Insulin resistance and impaired beta cell function in rheumatoid arthritis.<br />

Arthritis Rheum. 2006;54(9):2765-75.<br />

123. Stumvoll M, Goldstein Bj, van Haeften TW. Type 2 diabetes: principles of pathogenesis and therapy.<br />

Lancet. 2005;365(9467):1333-46.<br />

124. Masuzaki H, Paterson j, Shinyama H, Morton NM, Mullins jj, Seckl jR, Flier jS. A transgenic model of<br />

visceral obesity and the metabolic syndrome. Science. 2001;294(5549):2166-70.<br />

125. Morton NM, Paterson jM, Masuzaki H, Holmes MC, Staels B, Fievet C et al. Novel adipose tissuemediated<br />

resistance to diet-induced visceral obesity in 11 beta-hydroxysteroid dehydrogenase type<br />

1-deficient mice. Diabetes. 2004;53(4):931-8.<br />

126. Paulmyer-Lacroix O, Boullu S, Oliver C, Alessi MC, Grino M. Expression of the mRNA coding for<br />

11beta-hydroxysteroid dehydrogenase type 1 in adipose tissue from obese patients: an in situ<br />

hybridization study. j Clin Endocrinol Metab. 2002;87(6):2701-5.<br />

127. Rask E, Olsson T, Soderberg S, Andrew R, Livingstone DE, Johnson O, Walker BR. Tissue-specific<br />

dysregulation of cortisol metabolism in human obesity. j Clin Endocrinol Metab. 2001;86(3):1418-<br />

21.<br />

128. Tomlinson jW, Sinha B, Bujalska I, Hewison M, Stewart PM. Expression of 11beta-hydroxysteroid<br />

dehydrogenase type 1 in adipose tissue is not increased in human obesity. j Clin Endocrinol Metab.<br />

2002;87(12):5630-5.<br />

129. Paterson jM, Morton NM, Fievet C, Kenyon Cj, Holmes MC, Staels B, Seckl j, Mullins jj. Metabolic<br />

syndrome without obesity: Hepatic overexpression of 11beta-hydroxysteroid dehydrogenase type<br />

1 in transgenic mice. Proc Natl Acad Sci U S A. 2004;101(18):7088-93.<br />

130. Tsilchorozidou T, Honour jW, Conway GS. Altered cortisol metabolism in polycystic ovary syndrome:<br />

insulin enhances 5alpha-reduction but not the elevated adrenal steroid production rates. j Clin<br />

Endocrinol Metab. 2003;88(12):5907-13.<br />

49<br />

2<br />

Chapter 2


Chapter 2 Glucocorticoids: metabolic side effects<br />

131. Sutinen j, Kannisto K, Korsheninnikova E, Nyman T, Ehrenborg E, Andrew R, Wake Dj, Hamsten A,<br />

Walker BR, Yki-jarvinen H. In the lipodystrophy associated with highly active antiretroviral therapy,<br />

pseudo-Cushing’s syndrome is associated with increased regeneration of cortisol by 11betahydroxysteroid<br />

dehydrogenase type 1 in adipose tissue. Diabetologia. 2004;47(10):1668-71.<br />

132. Draper N, Walker EA, Bujalska Ij, Tomlinson jW, Chalder SM, Arlt W, Lavery GG, Bedendo O,<br />

Ray DW, Laing I, Malunowicz E, White PC, Hewison M, Mason Pj, Connell jM, Shackleton CH,<br />

Stewart PM. Mutations in the genes encoding 11beta-hydroxysteroid dehydrogenase type 1 and<br />

hexose-6-phosphate dehydrogenase interact to cause cortisone reductase deficiency. Nat Genet<br />

2003;34(4):434-9.<br />

133. Schacke H, Berger M, Rehwinkel H, Asadullah K. Selective glucocorticoid receptor agonists (SEGRAs):<br />

novel ligands with an improved therapeutic index. Mol Cell Endocrinol. 2007;275(1-2):109-17.<br />

134. Barf T, Vallgarda j, Emond R, Haggstrom C, Kurz G, Nygren A et al. Arylsulfonamidothiazoles as a<br />

new class of potential antidiabetic drugs. Discovery of potent and selective inhibitors of the 11betahydroxysteroid<br />

dehydrogenase type 1. j Med Chem. 2002;45(18):3813-5.<br />

135. Coppola GM, Kukkola Pj, Stanton jL, Neubert AD, Marcopulos N, Bilci NA, Wang H, Tomaselli HC,<br />

Tan j, Aicher TD, Knorr DC, jeng AY, Dardik B, Chatelain RE. Perhydroquinolylbenzamides as novel<br />

inhibitors of 11beta-hydroxysteroid dehydrogenase type 1. j Med Chem. 2005;48(21):6696-712.<br />

136. Alberts P, Nilsson C, Selen G, Engblom LO, Edling NH, Norling S, Klingstrom G, Larsson C, Forsgren<br />

M, Ashkzari M, Nilsson CE, Fiedler M, Bergqvist E, Ohman B, Bjorkstrand E, Abrahmsen LB. Selective<br />

inhibition of 11 beta-hydroxysteroid dehydrogenase type 1 improves hepatic insulin sensitivity in<br />

hyperglycemic mice strains. Endocrinology. 2003;144(11):4755-62.<br />

137. julian LD, Wang Z, Bostick T, Caille S, Choi R, DeGraffenreid M, Di Y, He X, Hungate RW, jaen jC, liu<br />

j, Monshouwer M, McMinn D, Rew Y, Sudom A, Sun D, Tu H, Ursu S, Walker N, Yan X, Ye Q, Powers<br />

jP. Discovery of novel, potent benzamide inhibitors of 11beta-hydroxysteroid dehydrogenase<br />

type 1 (11beta-HSD1) exhibiting oral activity in an enzyme inhibition ex vivo model. j Med Chem.<br />

2008;51(13):3953-60.<br />

138. johansson L, Fotsch C, Bartberger MD, Castro VM, Chen M, Emery M, Gustafsson S, Hale C, Hickman<br />

D, Homan E, jordan SR, Komorowski R, Li A, McRae K, Moniz G, Matsumoto G, Orihuela C, Palm G,<br />

Veniant M, Wang M, Williams M, Zhang j. 2-amino-1,3-thiazol-4(5H)-ones as potent and selective<br />

11beta-hydroxysteroid dehydrogenase type 1 inhibitors: enzyme-ligand co-crystal structure and<br />

demonstration of pharmacodynamic effects in C57Bl/6 mice. j Med Chem. 2008;51(10):2933-43.<br />

139. Saltiel AR, Kahn CR. Insulin signalling and the regulation of glucose and lipid metabolism. Nature.<br />

2001;414(6865):799-806.<br />

50


PART<br />

Glucocorticoids and islet-cell fuction<br />

II


Chapter 3<br />

Prednisolone-Induced Beta-Cell Dysfunction<br />

is Associated with Impaired Endoplasmic<br />

Reticulum Homeostasis in INS-1E Cells<br />

M.M.L. Linssen, D.H. van Raalte, E.J.M. Toonen, W. Alkema, G.C. van<br />

der Zon, W.H. Dokter, M. Diamant, B. Guigas and D. M. Ouwens<br />

Cellular Signalling. 2011;23(11):1708-15


Chapter 3 Glucocorticoids induce beta-cell ER stress<br />

56<br />

ABSTRACT<br />

Glucocorticoids (GCs), such as prednisolone, are widely prescribed anti-inflammatory<br />

drugs, but their use may induce glucose intolerance and diabetes. GC-induced betacell<br />

dysfunction contributes to these diabetogenic effects through mechanisms that<br />

remain to be elucidated. In this study, we hypothesized that activation of the unfolded<br />

protein response (UPR) following endoplasmic reticulum (ER) stress could be one of the<br />

underlying mechanisms involved in GC-induced beta-cell dysfunction. We report here that<br />

prednisolone did not affect basal insulin release, but time-dependently inhibited glucosestimulated<br />

insulin secretion in INS-1E cells. Prednisolone treatment also decreased both<br />

PDX1 and insulin expression, leading to a marked reduction in cellular insulin content.<br />

These prednisolone-induced detrimental effects were found to be prevented by prior<br />

treatment with the glucocorticoid receptor (GR) antagonist RU486 and associated with<br />

activation of two of the three branches of the UPR. Indeed, prednisolone induced a GRmediated<br />

activation of both ATF6 (activating transcription factor 6) and IRE1 (inositolrequiring<br />

kinase 1)/XBP1 (X-box-binding protein 1) pathways, but was found to reduce<br />

the phosphorylation of PERK (protein kinase RNA-like ER kinase) and its downstream<br />

substrate eIF2α (eukaryotic initiation factor 2α). These modulations of ER stress pathways<br />

were accompanied by upregulation of calpain 10 and increased cleaved caspase 3,<br />

indicating that long-term exposure to prednisolone ultimately promotes apoptosis. Taken<br />

together, our data suggest that the inhibition of insulin biosynthesis by prednisolone in the<br />

insulin-secreting INS-1E cells results, at least in part, from a GR-mediated impairment in ER<br />

homeostasis, which may lead to apoptotic cell death.


INTRoDUC<strong>TI</strong>oN<br />

Glucocorticoids (GCs), such as prednisolone, are potent and widely prescribed antiinflammatory<br />

drugs. However, their use is frequently associated with the development of<br />

adverse metabolic effects, and may lead to the development of steroid diabetes in up to 20-<br />

50% of patients [1, 2]. GC-induced hyperglycemia has been classically attributed to progressive<br />

development of insulin resistance in peripheral tissues [1, 2], but recent studies also point<br />

toward involvement of beta-cell dysfunction [3]. A clinical study performed in healthy<br />

males recently showed that both acute and chronic treatment with prednisolone attenuated<br />

pancreatic insulin secretion during standardized meal tests by impairing multiple aspects<br />

of beta-cell function [3]. In vitro studies have also shown that GCs lower glucose-stimulated<br />

insulin secretion (GSIS) in beta-cell lines and isolated pancreatic islets by various putative<br />

mechanisms involving either downregulation of GLUT2, inhibition of phospholipase C/<br />

protein kinase C signaling, modulation of the voltage-gated K(+) channels activity, alterations<br />

in intracellular Ca2+ homeostasis and/or decreased calcium efficacy on the insulin secretory<br />

process (see [2] for recent review). In addition, GCs may also reduce pancreatic beta-cell mass<br />

and affect insulin biosynthesis, at least in part by inducing apoptotic cell death [4-6].<br />

Defects in endoplasmic reticulum (ER) homeostasis have been proposed to underlie beta-cell<br />

dysfunction and impairment in insulin secretion in patients with type 2 diabetes, Wollcot-<br />

Rallison and Wolfram syndrome [7-9]. Three proteins associated with the ER-membrane,<br />

activating transcription factor 6 (ATF6), inositol-requiring enzyme 1α (IRE1α), and PKRlike<br />

eukaryotic initiation factor 2α kinase (PERK) monitor the activity of ER [8]. In absence<br />

of stress, these sensors of ER homeostasis are held in an inactive state through interaction<br />

with the ER-chaperone immunoglobulin binding protein (BIP) [8]. Conditions that affect ER<br />

homeostasis in beta cells, such as increased unfolded/misfolded proteins, Ca2+ depletion or<br />

enhanced insulin biosynthesis, result in release of BIP from the sensor proteins and activation<br />

of the unfolded protein response (UPR) [7, 8].<br />

Activation of ATF6, the first branch of the UPR, requires its translocation from the ER to<br />

the Golgi apparatus where ATF6 is cleaved into an active nuclear transcription factor that<br />

can regulate the expression of ER genes, such as X-box binding protein-1 (XBP1) [8]. The<br />

second pathway involved in the UPR leads to activation of the endoribonuclease IRE1α and<br />

subsequent cleavage of XBP1 mRNA into XBP1s, an alternative spliced form. XBS1s alone, or<br />

in conjunction with ATF6, regulates the synthesis of ER chaperones, and proteins involved in<br />

both ER biogenesis and ER-associated protein degradation (ERAD) [7, 8]. Finally, activation of<br />

PERK, which constitutes the third branch of the UPR, occurs through autophosphorylation of<br />

57<br />

3<br />

Chapter 3


Chapter 3 Glucocorticoids induce beta-cell ER stress<br />

its kinase domain and results in phosphorylation of its substrate eukaryotic initiation factor 2α<br />

(eIF2α). This leads to inhibition of global protein synthesis in addition to increased synthesis<br />

of ATF4 through alternative translation [10]. ATF4 does not only regulate the expression of<br />

ER-chaperones in order to restore ER homeostasis, but can also increase the expression of<br />

genes involved in apoptosis, like C/EBP-homologous protein (CHOP) and tribbles homolog<br />

3 (TRIB3) [8]. Mutations that functionally impair the activity of the PERK/eIF2α-pathway<br />

cause neonatal diabetes in humans (Wollcot-Rallison syndrome) and mouse models [11-13],<br />

further illustrating the importance of this pathway for beta cell function.<br />

Given the critical role of the UPR in beta-cell homeostasis, the aim of this study was to<br />

determine whether ER stress could be one of the underlying mechanisms involved in<br />

prednisolone-induced beta-cell dysfunction.<br />

MATERIAL AND METHoDS<br />

Cell culture: Rat insulinoma-derived insulin-secreting INS-1E cells (kind gift from Pr. P.<br />

Maechler, [14]) were cultured in a humidified atmosphere containing 5% CO in RPMI 1640<br />

2<br />

medium (Invitrogen, Breda, The Netherlands) supplemented with 10 mM HEPES, pH7.4,<br />

5% (v/v) heat-inactivated fetal calf serum, 2 mM glutamine, 1 mM sodium pyruvate, 50 mM<br />

2-mercaptoethanol, 100 units/ml penicillin, and 100 mg/ml streptomycin (Invitrogen, Breda,<br />

The Netherlands) as described previously [15]. For experiments, INS-1E cells were seeded<br />

at 2x105 cells/1 ml in Falcon 24-well plates (insulin secretion), or 9x105 cells/2 ml in Falcon<br />

6-well plates (western blotting, gene expression), and used 4 days later, with one medium<br />

change on day 3. When indicated, prednisolone, the glucocorticoid receptor antagonist RU486<br />

(mifepristone) and/or their vehicle (DMSO, 0.5% v/v) were added to the medium.<br />

Insulin secretion: Insulin secretion in response to glucose was performed as described<br />

earlier [15]. Briefly, cells were incubated with prednisolone or vehicle (0.5% DMSO) for<br />

various time points. Then, the cells were maintained for 2 h in glucose-free culture medium<br />

containing prednisolone or vehicle (0.5% DMSO), followed by two washes and a 30 min at<br />

37oC incubation in glucose-free Krebs–Ringer bicarbonate HEPES buffer (KRBH: 135 mM<br />

NaCl, 3.6 mM KCl, 5 mM NaHCO3, 0.5 mM NaH2PO4, 0.5 mM MgCl2, 1.5 mM CaCl2, 0.1%<br />

BSA and 10 mM HEPES, pH 7.4) in the presence or absence of DMSO or prednisolone.<br />

Next, cells were washed with glucose-free KRBH and incubated for 30 min in KRBH with<br />

or without prednisolone and secretagogues, as indicated. Then, plates were placed on ice<br />

and the supernatants were collected for determination of insulin secretion. Cellular insulin<br />

58


content was measured in acid–ethanol extracts [15]. Insulin concentrations were measured<br />

in supernatants and acid–ethanol extracts using a rat/mouse ELISA kit (Millipore, Nuclilab,<br />

Ede, The Netherlands).<br />

Protein analysis: INS-1E cells were incubated with vehicle or prednisolone as indicated.<br />

Then, cells were washed twice with phosphate buffered saline (PBS) and lysed in a buffer<br />

containing 10% (w/v) glycerol, 3% (w/v) SDS and 100 mM Tris–HCl (pH 6.8). Lysates were<br />

immediately boiled for 5 min and centrifugated (13,200 rpm; 2 min). Protein content of the<br />

supernatant was determined using a bicinchoninic acid protein assay kit (Pierce, Rockford,<br />

USA). Proteins were separated by SDS-PAGE followed by transfer to a polyvinylidene<br />

difluoride (PVDF) membrane. Membranes were blocked for 1 h at room temperature in TBST<br />

buffer (10 mM Tris–HCl (pH 7.4), 150 mM NaCl, and 0.1% (v/v) Tween 20 containing 5%<br />

(w/v) fat free milk) and incubated overnight with primary antibodies (see Supplementary<br />

Table 1). The membranes were then washed in TBST buffer and incubated with horseradish<br />

peroxidase-conjugated secondary antibodies for 1 h at room temperature. After washing,<br />

blots were developed using enhanced chemiluminescence and quantified by densitometry<br />

analysis using Image j software (NIH, Bethesda, USA).<br />

Gene expression studies: INS-1E cells were lysed in RLT-buffer (Qiagen, Hilden, Germany)<br />

for isolation of total RNA using the RNeasy system including on-column DNAse I treatment<br />

(Qiagen, Hilden, Germany). For PCR analysis, total RNA (1.5 µg) was reverse transcribed using<br />

a Superscript first strand synthesis kit (Invitrogen, Breda, The Netherlands) and quantitative<br />

real-time PCR was performed with SYBR Green on a StepOne Plus Real-time PCR system<br />

(Applied Biosystems, Foster City, CA, USA). All the primers sets used were designed for<br />

spanning an exon (if any) and have an efficiency of ~100+/-5% (Supplementary Table 2). Every<br />

sample was analyzed in duplicate and mRNA expression was normalized to hypoxanthineguanine<br />

phosphoribosyltransferase (HPRT) mRNA content, and expressed as arbitrary<br />

units. For whole genome expression profiling, total RNA (200 ng) was amplified, labeled and<br />

fragmented using the GeneChip 3’ IVT Express Kit (Affymetrix, Santa Clara, CA) according<br />

to the manufacturer’s instructions. Fragmented amplified RNA (10 µg) was applied to the<br />

Rat Genome 230 2.0 Array and hybridized for 16 hours at 45°C in a GeneChip Hybridization<br />

Oven 640 (Affymetrix). Following hybridization, the arrays were washed and stained with a<br />

GeneChip Fluidics Station 450 (Affymetrix) using the Affymetrix Hybridization Wash Stain<br />

(HWS) kit. The arrays were laser scanned with a GeneChip Scanner 3000 7G (Affymetrix,<br />

Santa Clara, CA). Data was saved as raw image file and quantified using Affymetrix GeneChip<br />

Command Console v 1.0 (Affymetrix).<br />

59<br />

3<br />

Chapter 3


Chapter 3 Glucocorticoids induce beta-cell ER stress<br />

Data analysis: All data are presented as means ± SEM. Statistical analysis was performed in<br />

SPSS version 17.0 (SPSS Inc., Chicago, IL, USA). Differences among groups were determined<br />

using independent t-tests or by ANOVA followed by Bonferroni correction for multiple<br />

comparisons. P2 and p-value


Figure 2. Effects of prednisolone on PDX1 and insulin expression in INS-1E cells. INS-1E cells were<br />

incubated with vehicle (0.5% v/v DMSO) or 700 nM prednisolone for the indicated times. A. Representative<br />

immunoblots for PDX1 and insulin protein expression are shown. Insulin receptor β-subunit (IRβ)<br />

expression was used as loading control. B-C. Densitometric quantification of immunoblots was performed<br />

and the results expressed in % of those from vehicle (DMSO)-treated cells at the same time-point. D-F.<br />

PDX1, insulin 1 and 2 mRNA expressions were determined using real time PCR, normalized for HPRT<br />

expression using the ∆Ct-method and expressed in arbitrary units. All the results are expressed as means<br />

± SEM (n=4). *, p


Chapter 3 Glucocorticoids induce beta-cell ER stress<br />

with the reduction in insulin content measured by ELISA, the insulin protein level was also<br />

significantly reduced 12 h following prednisolone addition when compared to vehicle-treated<br />

cells (Fig. 2a/c). The reductions in PDX1 and insulin protein expression induced by<br />

prednisolone were preceded by rapid decline in PDX1, insulin 1 and insulin 2 mRNA levels,<br />

which already became significant 1 h after addition of the drug (Fig. 2d-f). All these<br />

prednisolone-induced decrease in PDX1 and insulin mRNA and protein levels were prevented<br />

by prior treatment with RU486 (Fig. 3).<br />

Figure 4. Effects of prednisolone and RU486 on ATF6-mediated XBP1 mRNA expression in INS-<br />

1E cells. INS-1E cells were incubated with vehicle (0.5% v/v DMSO) or 700 nM prednisolone for the<br />

indicated times (A) or for 20h in the presence (black bars) or absence (open bars) of 1 µM of RU486 as<br />

indicated (B). A-B. XBP1 mRNA expression was determined using real time PCR, normalized for HPRT<br />

expression using the ∆Ct-method and expressed in arbitrary units. All the results are expressed as means<br />

± SEM (n=4). *, p


Figure 5. Effects of prednisolone and RU486 on IRE1α/XBP1s pathway in INS-1E cells. INS-1E<br />

cells were incubated with vehicle (0.5% v/v DMSO) or 700 nM prednisolone for the indicated times<br />

(A,C,E) or for 20h in the presence (black bars) or absence (open bars) of 1 µM of RU486 (B,D,F). A,E.<br />

Time-dependent effects of prednisolone were determined on IRE1α and XBP1s by immunoblots and<br />

densitometric quantifications. The results are expressed in % of those from vehicle (DMSO)-treated<br />

cells at the same time-point. C. Time-dependent effect of prednisolone on XBP1s mRNA expression was<br />

determined using real time PCR, normalized for HPRT expression using the ∆Ct-method and expressed in<br />

arbitrary unit. G. Representative immunoblots for IRE1α, XBP1s and IRβ protein expression are shown.<br />

B,F. Densitometric quantifications of IRE1α and XBP1s immunoblots were performed and the results<br />

expressed in arbitrary units. D. XBP1s mRNA expression was determined and expressed in arbitrary<br />

units. All the results are expressed as means ± SEM (n=4). *, p


Chapter 3 Glucocorticoids induce beta-cell ER stress<br />

reduced 8-12 h following the addition of prednisolone (Fig. 6a-b). This prednisolone-induced<br />

dephosphorylation of PERK and eIF2α was prevented by pre-treatment with RU486 (Fig. 6ce).<br />

The prednisolone-induced inactivation of the PERK/eIF2α pathway was accompanied by<br />

decrease in mRNA levels of CHOP and TRIB3 downstream target genes (data not shown).<br />

Figure 6. Effects of prednisolone and RU486 on PERK/eIF2α pathway in INS-1E cells. INS-1E cells<br />

were incubated with vehicle (0.5% v/v DMSO) or 700 nM prednisolone for the indicated times (A,C) or for<br />

20h in the presence (black bars) or absence (open bars) of 1 µM of RU486 (B,D,E). A,C. Time-dependent<br />

effects of prednisolone were determined on pPERK-Thr980, peIF2α-Ser51 and IRβ by immunoblots and<br />

densitometric quantifications. The results are expressed in % of those from vehicle (DMSO)-treated<br />

cells at the same time-point. E. Representative immunoblots for pPERK-Thr980, peIF2α-Ser51 and IRβ<br />

protein expression are shown. B,D. Densitometric quantifications of pPERK-Thr980 and peIF2α-Ser51<br />

immunoblots were performed and the results expressed in arbitrary units. All the results are expressed<br />

as means ± SEM (n=4). *, p


Prednisolone increases expression of distal markers of ER-stress pathways and induces<br />

apoptosis: Prednisolone did not affect mRNA or protein expression of the ER-chaperone BIP<br />

(data not shown). However, using microarray profiling analysis, we found that prednisolone<br />

induced a time-dependent upregulation of the ER-associated protein degradation (ERAD)<br />

components EDEM1 and MAN1A1 (Fig. 7a-b). Furthermore, we also observed a significant<br />

increase in calpain 10 mRNA expression within 4 h after the addition of prednisolone to INS-<br />

1E cells (Fig. 7c). This protease is involved in the ER-stress-induced apoptosis via delayed<br />

activation of the caspases 3 and 12 [17]. Accordingly, a significant GR-mediated increase in<br />

cleaved caspase-3 was found in INS-1E cells treated for 20h with prednisolone (Fig. 7d-e).<br />

Figure 7. Effects of prednisolone on distal markers of ER stress and apoptosis in INS-1E cells. INS-<br />

1E cells were incubated with vehicle (0.5% v/v DMSO) or 700 nM prednisolone for the indicated times<br />

(A-D) or for 20h in the presence (black bars) or absence (open bars) of 1 µM of RU486 (E-F). A-C. Timedependent<br />

effect of prednisolone on EDEM1, MAN1A1 and calpain 10 mRNA expression was determined<br />

using Affymetrix profiling. D. Time-dependent effect of prednisolone was determined on cleaved caspase<br />

3 by immunoblots and densitometric quantification was performed. The results are expressed in % of<br />

those from vehicle (DMSO)-treated cells at the same time-point. E. Representative immunoblots for<br />

cleaved caspase 3 and IRβ protein expression are shown. F. Densitometric quantification of cleaved<br />

caspase 3 immunoblots was performed and the results expressed in arbitrary units. All the results are<br />

expressed as means ± SEM (n=4). *, p


Chapter 3 Glucocorticoids induce beta-cell ER stress<br />

of prednisolone on beta-cell dysfunction are tightly associated with atypical GR-mediated<br />

modulation of the ER stress-induced UPR, as illustrated by concomitant activation of ATF6<br />

and IRE1α and inactivation of PERK/eIF2α signaling pathways.<br />

In response to perturbations of ER homeostasis in beta cells, a coordinated program is<br />

activated leading to upregulation of genes enhancing the ER protein processing capacity,<br />

such as protein folding, trafficking, and degradation [7-9]. ATF6-dependent transcriptional<br />

induction is regulated by ER stress-induced trafficking of ATF6 from the ER to the Golgi where<br />

the protein is cleaved by proteases to release a cytosolic active fragment. Although we were<br />

not able to detect an effect of prednisolone on ATF6 cleavage, the GR-mediated upregulation<br />

of the well-characterized ATF6 downstream target gene XBP1 suggests that this pathway<br />

is activated. Of note, prednisolone had no significant effect on XBP1 protein expression<br />

(data not shown), suggesting that the XBP1 mRNA is rapidly cleaved into XBP1s by IRE1α.<br />

Interestingly, nutrient or chemical ER stress-mediated activation of ATF6 has been shown<br />

to impair insulin expression and secretion in INS-1E cells, at least in part via suppression of<br />

PDX1 expression [18]. In addition to mitigating ER stress by increasing the transcription of<br />

key proteins involved in the regulation of ER folding capacity, IRE1α also lowers ER protein<br />

load by degrading mRNA of ER-targeted proteins [19]. In beta cells, IRE1α was shown to<br />

be directly involved in the degradation of insulin mRNA in response to ER stress [20-22].<br />

Furthermore, accumulation of the transcription factor XBP1s has also been linked to betacell<br />

dysfunction by reducing both insulin and PDX1 expression, and promoting apoptotic<br />

beta-cell death [23]. An additional explanation for the rapid decrease in insulin mRNA level<br />

induced by prednisolone could be the presence of a GC responsive element in the promoter<br />

region of the gene, which acts as a negative regulatory site [24, 25]. However, although this<br />

could contribute to the rapid downregulation of insulin expression, it can not explain the<br />

similar rapid decline in PDX1 mRNA observed. Interestingly, in our experimental conditions,<br />

the prednisolone-induced reduction of PDX1 and insulin mRNA levels preceded the onset of<br />

the ER-stress response. Therefore, it is possible that the ER-stress response is triggered by<br />

an increased demand for insulin synthesis caused by the initial decline in insulin and PDX1<br />

mRNA levels. Furthermore, PDX1 deficiency also enhances beta-cell susceptibility to ER<br />

stress-associated apoptosis [26].<br />

Another cellular strategy to alleviate ER stress is the reduction in protein influx by reducing<br />

protein translation, a process which is classically mediated by activation of PERK and<br />

subsequent phosphorylation of its downstream target eIF2α [7-9]. One of our most puzzling<br />

results is that prednisolone, by contrast to its activating effect on ATF6 and IRE1α pathways,<br />

decreases the phosphorylation of PERK and eIF2α, suggesting a GR-mediated increase in the<br />

66


activity of protein phosphatase(s) involved in the dephosphorylation of these key proteins.<br />

Although we did not find any change in protein phosphate 1 expression (data not shown),<br />

which has been implicated in the dephosphorylation of eIF2α [7-9], we cannot exclude more<br />

subtle enzymatic regulation in response to prednisolone and/or involvement of upstream<br />

phosphatases targeting PERK. It should however be mentioned that the exact mechanism(s)<br />

by which the PERK/eIF2α pathway is regulated during ER stress is still not entirely clear.<br />

For example, similar counter-intuitive observations were recently made in activated B cells<br />

where phosphorylation of the PERK/eIF2α pathway was reduced during ER stress and did<br />

not correlate with cellular protein synthesis rate [27]. In addition, loss of PERK function has<br />

also been reported not to affect protein synthesis but rather to impair ER-to-Golgi protein<br />

trafficking and proteasomal degradation [28].<br />

In relation to signaling pathways attenuating protein translation, crosstalk between GR and the<br />

nutritional sensor mammalian of rapamycin (mTOR) has been recently highlighted in muscle,<br />

identifying Regulated in Development and DNA damage responses 1 (REDD1), an upstream<br />

negative regulator of mTOR, as a direct GR-targeted gene [29]. Interestingly, in our conditions,<br />

prednisolone induced significant upregulation of REDD1 and concomitantly reduced mTORC1<br />

activity in INS-1E cells (MML/DMO, unpublished data), suggesting that modulation of this<br />

pathway could contribute to the GR-mediated reduction in insulin biosynthesis by decreasing<br />

global protein synthesis. Further experiments are still required to clarify this point.<br />

One of the limitations of our findings could eventually be the concentration of prednisolone<br />

used in the present study (700 nM), which is ~3-4 fold higher than the plasma therapeutic level<br />

reported in humans treated with the drug [30]. However, significant effects of prednisolone<br />

on insulin secretion/biosynthesis and ER stress signaling pathways are already present<br />

at lower concentrations (data not shown). To note, similar time- and dose-dependent GRmediated<br />

effects were also observed in INS-1E cells treated with dexamethasone, another GC<br />

compound (MML/ DMO, unpublished data).<br />

In a recent clinical study, we have reported that treatment with the glucagon-like peptide-1<br />

(GLP-1) receptor agonist exenatide (exendin-4) prevented prednisolone-induced beta-cell<br />

dysfunction in healthy men [31]. Although the underlying molecular mechanism(s) remain(s)<br />

to be elucidated, it is striking that exendin-4 was previously shown to decrease geneticallyor<br />

pharmacologically-induced ER stress and to improve beta-cell function and survival in<br />

mice, isolated islets and INS-1E cells [32, 33]. In addition, exendin-4 prevented GC-induced<br />

apoptosis in INS-1E cells [6]. Taken together, it is therefore tempting to suggest that the<br />

beneficial effect of exenatide evidenced on beta cell functions in humans could be ascribed,<br />

67<br />

3<br />

Chapter 3


Chapter 3 Glucocorticoids induce beta-cell ER stress<br />

at least in part, to a reduction in prednisolone-induced pancreatic ER stress [31]. Further in<br />

vitro studies are required to investigate whether exendin-4 is able to prevent prednisoloneinduced<br />

ER stress and beta-cell dysfunction.<br />

In conclusion, we report here that prednisolone increases ER stress in INS-1E cells, promoting<br />

activation of selective UPR signaling pathways which leads to decreased insulin expression.<br />

Taken together, our data suggest that the inhibition of GSIS could be ascribed, at least in<br />

part, to a prednisolone-induced decrease in insulin biosynthesis resulting from GR-mediated<br />

impairment in ER homeostasis and apoptotic cell death.<br />

Funding: This work was supported by Dutch <strong>TI</strong> <strong>Pharma</strong> grants (project T1-106, to ML, DHR,<br />

EjT, WHD, MD and DMO; project T2-105, to BG and DMO), the Federal Ministry of Health, the<br />

Ministry of Innovation, Science, Research and Technology of the German State of North-Rhine<br />

Westphalia (DMO), and the EU European Cooperation in the field of Scientific and Technical<br />

Research (COST) Action BM0602 (DMO).<br />

68


REFERENCES<br />

1. Clore jN, Thurby-Hay L. Glucocorticoid-induced hyperglycemia. Endocr Pract. 2009;15(5):469-74.<br />

2. van Raalte DH, Ouwens DM, Diamant M. Novel insights into glucocorticoid-mediated diabetogenic<br />

effects: towards expansion of therapeutic options? Eur j Clin Invest. 2009;39(2):81-93.<br />

3. van Raalte DH, Nofrate V, Bunck MC, van Iersel T, Elassaiss Schaap j, Nassander UK, Heine Rj, Mari<br />

A, Dokter WH, Diamant M. Acute and 2-week exposure to prednisolone impair different aspects of<br />

beta-cell function in healthy men. Eur j Endocrinol. 2010;162(4):729-35.<br />

4. jeong IK, Oh SH, Kim Bj, Chung jH, Min YK, Lee MS, Lee MK, Kim KW. The effects of dexamethasone<br />

on insulin release and biosynthesis are dependent on the dose and duration of treatment. Diabetes<br />

Res Clin Pract. 2001;51(3):163-71.<br />

5. Philippe j, Missotten M. Dexamethasone inhibits insulin biosynthesis by destabilizing insulin<br />

messenger ribonucleic acid in hamster insulinoma cells. Endocrinology. 1990;127(4):1640-5.<br />

6. Ranta F, Avram D, Berchtold S, Dufer M, Drews G, Lang F, Ullrich S. Dexamethasone induces cell death<br />

in insulin-secreting cells, an effect reversed by exendin-4. Diabetes. 2006;55(5):1380-90.<br />

7. Eizirik DL, Cardozo AK, Cnop M. The role for endoplasmic reticulum stress in diabetes mellitus.<br />

Endocr Rev. 2008;29(1):42-61.<br />

8. Hotamisligil GS. Endoplasmic reticulum stress and the inflammatory basis of metabolic disease. Cell.<br />

2010;140(6):900-17.<br />

9. Scheuner D, Kaufman Rj. The unfolded protein response: a pathway that links insulin demand with<br />

beta-cell failure and diabetes. Endocr Rev. 2008;29(3):317-33.<br />

10. Harding HP, Zhang Y, Ron D. Protein translation and folding are coupled by an endoplasmicreticulum-resident<br />

kinase. Nature. 1999;397(6716):271-4.<br />

11. Delepine M, Nicolino M, Barrett T, Golamaully M, Lathrop GM, julier C. EIF2AK3, encoding translation<br />

initiation factor 2-alpha kinase 3, is mutated in patients with Wolcott-Rallison syndrome. Nat Genet.<br />

2000;25(4):406-9.<br />

12. Scheuner D, Song B, McEwen E, Liu C, Laybutt R, Gillespie P, Saunders T, Bonner-Weir S, Kaufman Rj.<br />

Translational control is required for the unfolded protein response and in vivo glucose homeostasis.<br />

Mol Cell. 2001;7(6):1165-76.<br />

13. Zhang W, Feng D, Li Y, Iida K, McGrath B, Cavener DR. PERK EIF2AK3 control of pancreatic beta<br />

cell differentiation and proliferation is required for postnatal glucose homeostasis. Cell Metab.<br />

2006;4(6):491-7.<br />

14. Merglen A, Theander S, Rubi B, Chaffard G, Wollheim CB, Maechler P. Glucose sensitivity and<br />

metabolism-secretion coupling studied during two-year continuous culture in INS-1E insulinoma<br />

cells. Endocrinology. 2004;145(2):667-78.<br />

69<br />

3<br />

Chapter 3


Chapter 3 Glucocorticoids induce beta-cell ER stress<br />

15. de Leeuw van Weenen jE, Parlevliet ET, Maechler P, Havekes LM, Romijn jA, Ouwens DM, Pijl H,<br />

70<br />

Guigas B. The dopamine receptor D2 agonist bromocriptine inhibits glucose-stimulated insulin<br />

secretion by direct activation of the alpha2-adrenergic receptors in beta cells. Biochem <strong>Pharma</strong>col.<br />

2010;79(12):1827-36.<br />

16. Chen X, Shen j, Prywes R. The luminal domain of ATF6 senses endoplasmic reticulum (ER) stress<br />

and causes translocation of ATF6 from the ER to the Golgi. j Biol Chem. 2002;277(15):13045-52.<br />

17. Nakagawa T, Yuan j. Cross-talk between two cysteine protease families. Activation of caspase-12 by<br />

calpain in apoptosis. j Cell Biol. 2000;150(4):887-94.<br />

18. Seo HY, Kim YD, Lee KM, Min AK, Kim MK, Kim HS, Won KC, Park jY, Lee KU, Choi HS, Park KG, Lee<br />

IK. Endoplasmic reticulum stress-induced activation of activating transcription factor 6 decreases<br />

insulin gene expression via up-regulation of orphan nuclear receptor small heterodimer partner.<br />

Endocrinology. 2008;149(8):3832-41.<br />

19. Hollien j, Weissman jS. Decay of endoplasmic reticulum-localized mRNAs during the unfolded<br />

protein response. Science. 2006;313(5783):104-7.<br />

20. Lipson KL, Fonseca SG, Ishigaki S, Nguyen LX, Foss E, Bortell R, Rossini AA, Urano F. Regulation of<br />

insulin biosynthesis in pancreatic beta cells by an endoplasmic reticulum-resident protein kinase<br />

IRE1. Cell Metab. 2006;4(3):245-54.<br />

21. Lipson KL, Ghosh R, Urano F. The role of IRE1alpha in the degradation of insulin mRNA in pancreatic<br />

beta-cells. PLoS One. 2008;3(2):e1648.<br />

22. Pirot P, Naamane N, Libert F, Magnusson NE, Orntoft TF, Cardozo AK, Eizirik DL. Global profiling<br />

of genes modified by endoplasmic reticulum stress in pancreatic beta cells reveals the early<br />

degradation of insulin mRNAs. Diabetologia. 2007;50(5):1006-14.<br />

23. Nader N, Ng SS, Lambrou GI, Pervanidou P, Wang Y, Chrousos GP, Kino T. AMPK regulates metabolic<br />

actions of glucocorticoids by phosphorylating the glucocorticoid receptor through p38 MAPK. Mol<br />

Endocrinol. 2010;24(9):1748-64.<br />

24. Goodman PA, Medina-Martinez O, Fernandez-Mejia C. Identification of the human insulin<br />

negative regulatory element as a negative glucocorticoid response element. Mol Cell Endocrinol.<br />

1996;120(2):139-46.<br />

25. Lee jK, Tsai SY. Multiple hormone response elements can confer glucocorticoid regulation on the<br />

human insulin receptor gene. Mol Endocrinol. 1994;8(5):625-34.<br />

26. Sachdeva MM, Claiborn KC, Khoo C, Yang j, Groff DN, Mirmira RG, Stoffers DA. Pdx1 (MODY4) regulates<br />

pancreatic beta cell susceptibility to ER stress. Proc Natl Acad Sci U S A. 2009;106(45):19090-5.<br />

27. Goldfinger M, Shmuel M, Benhamron S, Tirosh B. Protein synthesis in plasma cells is regulated<br />

by crosstalk between endoplasmic reticulum stress and mTOR signaling. Eur j Immunol.<br />

2011;41(2):491-502.<br />

28. Gupta S, McGrath B, Cavener DR. PERK (EIF2AK3) regulates proinsulin trafficking and quality<br />

control in the secretory pathway. Diabetes. 2010;59(8):1937-47.


29. Shimizu N, Yoshikawa N, Ito N, Maruyama T, Suzuki Y, Takeda S, Nakae j, Tagata Y, Nishitani S,<br />

Takehana K, Sano M, Fukuda K, Suematsu M, Morimoto C, Tanaka H. Crosstalk between Glucocorticoid<br />

Receptor and Nutritional Sensor mTOR in Skeletal Muscle. Cell Metab. 2011;13(2):170-82.<br />

30. Xu j, Winkler j, Derendorf H. A pharmacokinetic/pharmacodynamic approach to predict total<br />

prednisolone concentrations in human plasma. j <strong>Pharma</strong>cokinet <strong>Pharma</strong>codyn. 2007;34(3):355-72.<br />

31. van Raalte DH, van Genugten RE, Linssen MM, Ouwens DM, Diamant M. Glucagon-Like Peptide-1<br />

Receptor Agonist Treatment Prevents Glucocorticoid-Induced Glucose Intolerance and Islet-Cell<br />

Dysfunction in Humans. Diabetes Care. 2011.<br />

32. Cunha DA, Ladriere L, Ortis F, Igoillo-Esteve M, Gurzov EN, Lupi R, Marchetti P, Eizirik DL, Cnop M.<br />

Glucagon-like peptide-1 agonists protect pancreatic beta-cells from lipotoxic endoplasmic reticulum<br />

stress through upregulation of BiP and junB. Diabetes. 2009;58(12):2851-62.<br />

33. Yusta B, Baggio LL, Estall jL, Koehler jA, Holland DP, Li H, Pipeleers D, Ling Z, Drucker Dj. GLP-1<br />

receptor activation improves beta cell function and survival following induction of endoplasmic<br />

reticulum stress. Cell Metab. 2006;4(5):391-406.<br />

71<br />

3<br />

Chapter 3


Chapter 3 Glucocorticoids induce beta-cell ER stress<br />

SUPPLEMENTARy FILES<br />

Supplementary Table 1. List of Antibodies<br />

Primary antibody Supplier Reference Dilution<br />

cleaved caspase 3 R&D systems Mab835 1:1000<br />

Insulin Santa Cruz Biotechnology sc-9168 1:1000<br />

Insulin receptor β-subunit Santa Cruz Biotechnology sc-711 1:1000<br />

IRE1α Cell Signaling Technology 3294 1:1000<br />

PDX1 Santa Cruz Biotechnology sc-14664 1:1000<br />

peIF2α-S51 Cell Signaling Technology 9721 1:1000<br />

pPERK-T980 Santa Cruz Biotechnology 32577 1:1000<br />

XBP1s Biolegend 619502 1:1000<br />

XBP1t Abcam ab37151 1:1000<br />

Supplementary Table 2. Primer sequences<br />

Gene Accession no Forward primer Reverse primer<br />

HPRT NM012583 5’-ggtccttttcaccagcaagct-3’ 5’-tgacactggcaaaacaatgca-3’<br />

Insulin 1 NM019129 5’-gcagaagcgtggcattgtg-3’ 5’-ggtggactcsagttgcagtagttctc-3’<br />

Insulin 2 NM019130 5’-gctggccctgctcatcct-3’ 5’-ccaccaagtgagaaccacaaag-3’<br />

PDX1 NM022852 5’-ggtatagccagcgagatgct-3’ 5’-tcagttgggagcctgattct-3’<br />

XBP1s NM001004210 5’-gagtccgcagcaggtg-3’ 5’-gcgtcagaatccatggga-3’<br />

XBP1t NM001004210 5’-gagcagcaagtggtggattt-3’ 5’-tctcaatcacaagcccatga-3’<br />

72


Chapter 4<br />

Acute and 2-Week Exposure to Prednisolone<br />

Impair Different Aspects of Beta-Cell Function<br />

in Healthy Men<br />

D.H. van Raalte, V. Nofrate, M.C. Bunck, T. van Iersel, J. Elassaiss<br />

Schaap, U.K Nässander, R. J. Heine, A. Mari, W.H.A. Dokter and M.<br />

Diamant<br />

Eur J Endocrinol. 2010;162(4):729-35


Chapter 4 Glucocorticoids impair beta-cell function in humans<br />

76<br />

ABSTRACT<br />

objective. Glucocorticoids (GCs), such as prednisolone, are associated with adverse<br />

metabolic effects, including glucose intolerance and diabetes. In contrast to the well-known<br />

GC-induced insulin resistance, effects of GCs on beta-cell function are less well established.<br />

We assessed the acute and short-term effects of prednisolone treatment on beta-cell<br />

function in healthy men.<br />

Research Design and Methods. A randomized, double blind, placebo-controlled trial,<br />

consisting of two protocols was conducted. In protocol 1 (n=6), placebo and a single dose<br />

of 75 mg prednisolone were administered. In protocol 2 (n=23), participants received 30<br />

mg prednisolone daily or placebo for 15 days. Both empirical and model-based parameters<br />

of beta-cell function were calculated from glucose, insulin and C-peptide concentrations<br />

obtained during standardized meal tests before and during prednisolone treatment<br />

(protocols 1 and 2), and one day after cessation of treatment (protocol 2).<br />

Results: 75 mg prednisolone acutely increased the area under the postprandial glucose<br />

curve (AUC ) (P=0.005) and inhibited several parameters of beta-cell function, including<br />

gluc<br />

AUC /AUC ratio (P=0.004), insulinogenic index (P=0.007), glucose sensitivity (P=0.02)<br />

c-pep gluc<br />

and potentiation factor ratio (PFR) (P=0.04). A 15-day treatment with prednisolone<br />

increased AUC (P


Glucocorticoids (GCs), such as prednisolone, are very efficacious and frequently prescribed<br />

anti-inflammatory drugs. Unfortunately, supraphysiological levels of GCs induce adverse<br />

metabolic effects, including glucose intolerance and diabetes [1]. Steroid diabetes may<br />

develop in up to 20-50% of patients with excessive plasma GC levels [2]. GCs are well known<br />

to reduce insulin sensitivity, resulting in increased hepatic glucose production and decreased<br />

peripheral glucose disposal [3]. The role of beta-cell dysfunction in GC-related diabetogenic<br />

effects is less clear. GCs impaired insulin secretion in rodent-derived islets in vitro [4]. In<br />

vivo in both rodents [5] and humans [6], a single day of GC administration impaired insulin<br />

secretion, resulting in hyperglycemia. More prolonged exposure to GCs, on the other hand,<br />

induced fasting hyperinsulinemia and increased insulin secretion in both wild type rodents<br />

[7] and healthy humans [8-12], most likely to compensate for impaired insulin sensitivity. In<br />

rodent models of obesity [13, 14] and in susceptible humans, however, this compensation<br />

failed. These ‘at risk’ populations included normoglycemic individuals with reduced insulin<br />

sensitivity or low glucose-stimulated insulin secretion (GSIS) before GC treatment [9, 11, 12]<br />

and normoglycemic, first-degree relatives of patients with type 2 diabetes mellitus [10]. It<br />

was concluded that GCs may only induce beta-cell dysfunction in vulnerable populations.<br />

The above-mentioned studies, however, have a limitation. Beta-cell function was assessed<br />

by tests using intravenous glucose loads, such as the intravenous glucose tolerance test<br />

(IVGTT) or the hyperglycemic clamp. As the magnitude of the insulin response under normal<br />

conditions also depends on other factors, such as non-glucose substrates [15, 16], incretins<br />

[17] and neurotransmitters [18], the hyperglycemic clamp may represent a less physiological<br />

condition relative to tests using orally administered insulin secretagogues.<br />

More recently, various parameters of beta-cell function have been calculated by modeling<br />

glucose and C-peptide plasma concentrations during standardized meal tests [19]. This<br />

approach enables the assessment of various aspects of beta-cell function under daily life<br />

conditions and also allows to evaluate the separate roles of insulin secretion and insulin<br />

sensitivity on glucose control in a single test [19].<br />

The aim of the present study was to assess the effects of both acute and short-term exposure<br />

to a widely used GC, i.e. prednisolone, on various aspects of beta-cell function in healthy men.<br />

77<br />

4<br />

Chapter 4


Chapter 4 Glucocorticoids impair beta-cell function in humans<br />

RESEARCH DESIGN AND METHoDS<br />

Study design: The study was a single-center, double blind, randomized, placebo-controlled<br />

study, consisting of two distinct parts.<br />

Protocol 1: Acute Study: In order to assess the acute effects of prednisolone treatment, eligible<br />

participants (n=6) ingested a placebo tablet on day 0 at 08:00 AM and a 75 mg prednisolone<br />

capsule at 08:00 AM on day 1. No study medication was given on day 2. Standardized meal<br />

tests were performed on days 0, 1 and 2 at 10:00 AM.<br />

Protocol 2: Two-week study: The effects of short-term treatment with prednisolone on beta-<br />

cell function were assessed in different subjects. Participants (n=23) were randomly assigned<br />

to a treatment with either 30 mg prednisolone once daily (n=12) or placebo (n=11) for a period<br />

of 15 days (medication was taken in the morning). Standardized meal tests were performed<br />

at day 0 and at day 15 at 10.00 AM. Placebo was administered as a subject-blinded treatment<br />

on day 0 at 08:00 AM (baseline). On day 15, study medication was also administered at 08:00<br />

AM.<br />

Study medication: Prednisolone tablets were obtained from Pfizer AB (Sollentuna, Sweden),<br />

placebo tablets were provided by Schering-Plough (Oss, The Netherlands). The tablets were<br />

encapsulated in order to allow the treatment to be blinded.<br />

Study population: Both protocols enrolled healthy male volunteers (age range 20 and<br />

45 years; body mass index (BMI) 22-30 kg/m 2 ). Health status was confirmed by medical<br />

history taking, physical and laboratory examinations, and ECG and vital signs recordings.<br />

Furthermore, normal glucose metabolism was verified by a 75 g 2-hour oral glucose tolerance<br />

test (OGTT). Participants were excluded if they had a clinically relevant history or presence<br />

of a medical disorder known to affect the investigational parameters, if they were taking<br />

medication, except for incidental analgesics, or if they had a first-degree relative with type 2<br />

diabetes mellitus.<br />

Study assessments: Screening assessments were performed within a three-week period prior<br />

to inclusion. Subjects were admitted to the clinical research unit at Xendo Drug Development<br />

(Groningen, The Netherlands) at 10:00 PM. The following day at 10:00 AM, subjects<br />

consumed a standardized 4-hour meal test (35 g proteins, 39 g fat and 75 g carbohydrates)<br />

after an overnight fast of minimal 12 hours. Samples for determination of glucose, insulin<br />

and C-peptide were obtained at times 0, 5, 10, 20, 30, 60, 90, 120, 180, and 240 min, with the<br />

78


meal beginning immediately after the time 0 sample and consumed within 15 min. Insulin<br />

and C-peptide were measured by Xendo Drug Development using immuno assays (Mercodia,<br />

Uppsala, Sweden).<br />

Data analysis: Area under the 4-hr postprandial glucose (AUC ) and C-peptide (AUC gluc c-<br />

) curves were determined by using the trapezoidal rule. Measures of insulin sensitivity,<br />

pep<br />

including homeostatic model assessment (HOMA-IR) [20] and oral glucose insulin sensitivity<br />

(OGIS) [21] were calculated. Empirical measures of beta-cell function, including AUC / c-pep<br />

AUC ratio and the insulinogenic index (IGI) (insulin -insulin )/(gluc -gluc ) [22]<br />

gluc t=30 t=0 t=30 t=0<br />

were calculated.<br />

Modeling analysis of beta-cell function: Pancreatic beta-cell function was assessed with a<br />

model that describes the relationship between insulin secretion and glucose concentration,<br />

which has been described in detail previously [19]. The model expresses insulin secretion (in<br />

pmol/min per m2 of body surface area) as the sum of two components. The first component<br />

describes the dose-response relation between insulin secretion and glucose concentrations<br />

during the meal test. Three parameters are obtained from this dose-response relation. First,<br />

the sensitivity of the beta cell to changes in plasma glucose levels, called glucose sensitivity.<br />

It is derived from the mean slope of the dose-response curve. Second, the fasting secretory<br />

tone is calculated from the dose-response curve. This represents fasting insulin secretion<br />

rates at a fixed glucose concentration of 4.5 mM (approximately the mean fasting glucose<br />

concentration in the groups). The third parameter is a potentiation factor, which may<br />

account for several potentiating signals to the beta cell (e.g. non-glucose metabolites, incretin<br />

hormones and neural factors) or amplifying pathways within the beta cell [23], although its<br />

exact physiological basis warrants further investigation. The excursion of the potentiation<br />

factor was quantified using a ratio between mean values at times 160-180 and 0–20 min, and<br />

is called the potentiation factor ratio (PFR). The second component of the model describes<br />

the insulin response to the rate of change of glucose concentration. This component is termed<br />

rate sensitivity, which is related to early insulin release [19].<br />

The model parameters were estimated from glucose and C-peptide concentration by<br />

regularized least squares, as described previously. Regularization involves the choice of<br />

smoothing factors that were selected to obtain glucose and C-peptide model residuals with<br />

standard deviations close to the expected measurement error (1% for glucose and 5% for<br />

C-peptide). Insulin secretion rates (ISR) were calculated from the model every 5 min [24].<br />

Estimation of the individual model parameters was performed blinded to the randomization<br />

of patients to treatment.<br />

79<br />

4<br />

Chapter 4


Chapter 4 Glucocorticoids impair beta-cell function in humans<br />

Statistical analysis: Data are presented as mean values ± standard error of the mean (SEM)<br />

or, in case of skewed distribution, as median (interquartile range). In the acute study, outcome<br />

variables were log-transformed and differences were tested by analysis of variance (ANOVA)<br />

with Bonferroni post-hoc test. For the two-week study, between-group treatment effects were<br />

tested with Mann-Whitney U. All statistical analyses were run on SPSS for Windows version<br />

15.0 (SPSS, Chicago, IL, USA). A P < 0.05 was considered statistically significant.<br />

Ethics and good clinical practice: All participants provided written informed consent. The<br />

study was approved by an independent ethics committee and the study was conducted in<br />

accordance with the Declaration of Helsinki, using good clinical practice.<br />

RESULTS<br />

Acute study<br />

Fasting metabolic parameters and glucose- and C-peptide profiles during meal tests:<br />

Six healthy participants were included in this protocol (age: 24.3 ±1.5 years, BMI: 24.2 ± 0.9<br />

kg/m2 ). High single-dose prednisolone treatment did not change fasting plasma glucose (FPG)<br />

or insulin (FPI), but decreased OGIS (P=0.009) (Table 1). During the meal test, prednisolone<br />

augmented AUC (P=0.005), while C-peptide secretion remained unchanged (Figure<br />

gluc<br />

1, panels A and B; Table 1). One day following discontinuation of prednisolone treatment,<br />

glucose levels during the meal test normalized, but AUC was increased (P=0.002).<br />

c-pep<br />

Figure 1. 75 mg Prednisolone acutely increased postprandial glucose concentrations (panel A), which<br />

was not accompanied by increased AUCc-pep (panel B). After discontinuation of prednisolone treatment,<br />

glucose concentrations normalized during the meal test, but C-peptide levels were increased. Solid line<br />

with black squares represents day 0 (placebo), dotted line with black circles denotes day 1 (prednisolone<br />

75 mg), and intersected line with white circles is day 2 (no treatment). Date are mean ± SEM.<br />

80


Table 1 (Acute study). Subject characteristics following administration of placebo (day 0) and 75 mg<br />

prednisolone (day 1), and after cessation of treatment (day 2). Values are median (interquartile range).<br />

P1 indicates day 0 versus day 1; P2 denotes day 1 versus day 2.<br />

Fasting plasma glucose<br />

(mmol/l)<br />

Fasting plasma insulin<br />

(pmol/l)<br />

HOMA-IR<br />

(no dimension)<br />

OGIS<br />

(ml/min.m2 )<br />

AUC gluc<br />

(mmol/L.240min)<br />

AUC cpep<br />

(nmol/L.240min)<br />

Insulinogenic Index<br />

(no dimension)<br />

AUC /AUC x 1000<br />

ISR gluc<br />

(no dimension)<br />

P1 P2<br />

Day 0 4.6 (4.1-4.8) NS 0.037<br />

Day 1 4.8 (4.3-5.2)<br />

Day 2 4.2 (3.8-4.3)<br />

Day 0 22 (21-27) NS NS<br />

Day 1 26 (22-31)<br />

Day 2 29 (22-33)<br />

Day 0 0.5 (0.5-0.6) NS NS<br />

Day 1 0.6 (0.5-0.7)<br />

Day 2 0.6 (0.5-0.7)<br />

Day 0 459 (424-509) 0.009 0.003<br />

Day 1 387 (341-418)<br />

Day 2 490 (470-503)<br />

Day 0 1210 (966-1295) 0.005 0.017<br />

Day 1 1565 (1393-1778)<br />

Day 2 1264 (1033-1309)<br />

Day 0 37 (32-41) NS 0.003<br />

Day 1 34 (30-40)<br />

Day 2 50 (48-57)<br />

Day 0 173 (131-303) 0.007 0.04<br />

Day 1 74 (55-94)<br />

Day 2 174 (96-217)<br />

Day 0 30 (26-36) 0.004


Chapter 4 Glucocorticoids impair beta-cell function in humans<br />

Figure 2. 75 mg Prednisolone reduced all model-derived parameters of beta-cell function, of which<br />

glucose sensitivity and potentiation factor ratio reached statistical significance. Graphs are Box-and-<br />

Whisker plots with median and interquartile range. Panel A: glucose sensitivity of the beta cell; panel<br />

B: basal secretory tone; panel C: potentiation factor ratio; panel D: rate sensitivity. * P


Figure 3. A two-week treatment with prednisolone increased area under the postprandial glucose curve<br />

(panel A: placebo; panel B: prednisolone), despite increased C-peptide levels (panel C: placebo; panel D:<br />

prednisolone). Solid line with black squares represents day 0, dotted line with white squares represents<br />

day 15. Date are mean ± SEM.<br />

Figure 4. A 15-day treatment with prednisolone decreased the basal secretory tone and potentiation<br />

factor ratio (panel B and C). Glucose sensitivity and rate sensitivity were not affected (panel A and D).<br />

Graphs are Box-and-Whisker plots with median and interquartile range. * P


Chapter 4 Glucocorticoids impair beta-cell function in humans<br />

Table 2 (Two-week study). Subject characteristics before and during 15 day-treatment with 30 mg<br />

prednisolone daily (n =12) or placebo (n =11). Values are median (interquartile range).<br />

Fasting plasma glucose<br />

(mmol/l)<br />

Fasting plasma insulin<br />

(pmol/l)<br />

HOMA-IR<br />

(no dimension)<br />

OGIS<br />

(ml/min.m2 )<br />

AUC gluc<br />

(mmol/L.240min)<br />

AUC cpep<br />

(nmol/L.240min)<br />

Insulinogenic Index<br />

(no dimension)<br />

AUC /AUC x 1000<br />

ISR gluc<br />

(no dimension)<br />

84<br />

Pre-treatment On-treatment P<br />

PLB 4.6 (4.5-4.9) 4.4 (3.9-4.8) 0.023<br />

30 4.3 (3.9-5.2) 5.0 (4.4-5.1)<br />

PLB 22 (21-36) 27 (22-37) NS<br />

30 28 (22-39) 57 (39-68)<br />

PLB 0.5 (0.4-0.8) 0.6 (0.5-0.7) 0.06<br />

30 0.6 (0.5-0.8) 1.2 (0.8-1.5)<br />

PLB 463 (441-524) 496 (444-517) 0.031<br />

30 457 (427-511) 404 (374-415)<br />

PLB 1173 (1038-1241) 1147 (1097-1224) 0.001<br />

30 1224 (1070-1297) 1466 (1389-1532)<br />

PLB 292 (223-316) 260 (213-288) 0.05<br />

30 278 (256-364) 334 (320-391)<br />

PLB 127 (117-160) 115 (87-182) NS<br />

30 139 (78-163) 155 (79-161)<br />

PLB 34 (28-43) 31 (25-37) NS<br />

30 34 (30-38) 31 (30-38)<br />

Abbreviations. 30: prednisolone 30 mg daily; AUCgluc: area under postprandial glucose curve; AUCc-pep:<br />

area under postprandial C-peptide curve; HOMA-IR: homeostatic model assessment insulin resistance;<br />

IQR: interquartile range; OGIS: oral glucose insulin sensitivity; PLB: placebo.<br />

Comparisons between subjects of both protocols: No statistical significant differences<br />

were measured between the subjects of the acute and two-week protocol, regarding age, BMI,<br />

physical activity and metabolic parameters at baseline.<br />

Experience of prednisolone-related symptoms: Subjects treated with prednisolone did<br />

not report increased physical complaints as compared to placebo. The completion rate of<br />

both studies was 100%, none of the participants withdrew due to side effects of prednisolone<br />

exposure.<br />

DISCUSSIoN<br />

GCs are well known to perturb glucose metabolism in humans, which most often is attributed<br />

to reduction of insulin sensitivity. It is far less clear to what extent beta-cell dysfunction<br />

contributes to the diabetogenic effects of GC therapy. In this study, we demonstrate that


prednisolone, as the most widely prescribed oral GC worldwide, in addition to reducing<br />

insulin sensitivity, impairs beta-cell function in healthy men, both following acute and twoweek<br />

exposure. In contrast to previous studies, we assessed the effects of GCs on beta-cell<br />

function under daily life conditions, i.e. during standardized meal tests [19].<br />

A single dose of 75 mg prednisolone acutely increased AUC gluc , while C-peptide secretion<br />

failed to respond (Figure 1, panels A and B). These data confirm and expand previous studies<br />

assessing the acute effects of GCs in both rodents [5] and humans [6], in which a single-day<br />

treatment with hydrocortisone [5] or prednisolone [6] prevented adequate response of beta<br />

cells to hyperglycemia. In line with these observations, Plat and colleagues demonstrated that<br />

elevation of morning cortisol levels acutely impairs insulin secretion [25]. In our study, all<br />

measured beta-cell function parameters declined by 25-50%, including measures for earlyand<br />

late phase insulin secretion. In vitro studies in rodent islet cells have revealed several<br />

mechanisms by which GCs acutely interfere with insulin secretory pathways. First, GCs reduce<br />

the uptake and oxidation of several metabolites including glucose. Moreover, GCs augment<br />

outward potassium currents, which, by reducing cell membrane depolarization, limit calcium<br />

influx. In addition, GCs may reduce insulin secretion by decreasing the efficacy of calcium on<br />

the secretory process. Finally, GCs were shown to reduce insulin secretion induced by the<br />

parasympathetic nervous system [4, 26].<br />

On day 2, beta-cell function appeared to have recovered from the acute effects of prednisolone,<br />

since fasting insulin secretion and insulin secretion during the standardized meal test were<br />

increased. This may indicate delayed compensation for the insulin resistance on day 1, but<br />

the increased insulin secretion could also serve to correct subtle disturbances in glucose<br />

homeostasis, although surrogate markers for insulin sensitivity were not decreased on day 2.<br />

In contrast to the acute study, subjects treated with prednisolone 30 mg daily for 15 days,<br />

increased C-peptide secretion during prednisolone treatment (Figure 3). Despite this<br />

enhanced secretion, however, fasting glucose and postprandial AUC exceeded baseline<br />

gluc<br />

levels, indicating a relative hypoinsulinemia and accordingly a decline in beta-cell function.<br />

This relative hypoinsulinemia under fasting conditions becomes evident in our beta-cell<br />

model parameter ‘insulin secretory tone at a glucose level of 4.5 mM’, in which ISR are directly<br />

related to glucose plasma concentrations. This parameter was significantly reduced following<br />

15-days of prednisolone exposure. In addition to insufficient basal secretory tone, PFR was<br />

also reduced by a two-week prednisolone treatment.<br />

Previous studies in which subjects were exposed for multiple days to GCs, also reported<br />

increased insulin secretion, as assessed by hyperglycemic clamps [8, 9, 11, 12] and IVGTTs<br />

85<br />

4<br />

Chapter 4


Chapter 4 Glucocorticoids impair beta-cell function in humans<br />

[10]. However, the studies that accounted for GC-induced reductions in insulin sensitivity<br />

by calculating the disposition index [11] or by using minimal model analysis [10], reported<br />

adequate compensation of insulin resistance in healthy subjects through sufficiently<br />

augmented insulin secretion. Only subjects with (subtle) glucometabolic abnormalities prior<br />

to GC treatment were unable to fully compensate for GC-induced insulin resistance [9-12].<br />

The most important difference between the above-mentioned studies and our study is that we<br />

used an oral stimulation test to assess various aspects of beta-cell function. Our experimental<br />

design may be more physiological compared to tests using intravenous glucose, since the<br />

former comprises the contribution of multiple factors, including incretins [17], non-glucose<br />

metabolic stimuli, such as non-esterified fatty acids (NEFA) [15] or amino acids [16], and the<br />

autonomic nervous system [18], all of which together account for a substantial proportion<br />

of the normal meal-related insulin response. These non-glucose insulin secretagogues are<br />

included as the ‘potentiation factor’ in our beta-cell model, which was significantly impaired<br />

by prednisolone, both following acute and short-term exposure. Our findings illustrate that<br />

the harmful effects of GCs on beta-cell function may only become fully apparent when using<br />

an oral stimulation test, which more comprehensively tests the role of the intestinal-islet axis<br />

on glucose homeostasis. To identify the specific non-glucose stimuli for insulin secretion that<br />

are impaired by GC treatment will require additional investigation.<br />

It is important to note that the meal-induced insulin response during prednisolone treatment<br />

was markedly different in the acute protocol as compared to the two-week study. We propose<br />

that GCs induce an acute inhibitory effect on beta-cell function, as extensively demonstrated<br />

in both in vitro and in vivo experiments, but that beta-cell function partly recovers following<br />

more prolonged exposure. In the latter situation, GC-induced insulin resistance may oppose<br />

the direct effect of GCs on the beta cell by enhancing insulin secretion. It should however<br />

also be stressed that the dosages used in the two protocols were different, which could have<br />

contributed to the observed difference in insulin responses. It is well known that the effects of<br />

GCs on glucose metabolism are highly dependent on the administered dose [27].<br />

We conclude that GCs impair several aspects of beta-cell function, both following acute and<br />

short-term treatment, in healthy normoglycemic men, when measured under physiological,<br />

daily life conditions. These data indicate that GC-induced beta-cell dysfunction, in addition to<br />

insulin resistance, contributes to the development of steroid diabetes.<br />

Funding: This paper was written within the framework of project T1-106 of the Dutch Top<br />

Institute <strong>Pharma</strong>.<br />

86


REFERENCES<br />

1. Schacke H, Docke WD, Asadullah K. Mechanisms involved in the side effects of glucocorticoids.<br />

<strong>Pharma</strong>col Ther. 2002;96(1):23-43.<br />

2. Onwubalili jK, Obineche EN. High incidence of post-transplant diabetes mellitus in a single-centre<br />

study. Nephrol Dial Transplant. 1992;7(4):346-9.<br />

3. McMahon M, Gerich j, Rizza R. Effects of glucocorticoids on carbohydrate metabolism. Diabetes<br />

Metab Rev. 1988;4(1):17-30.<br />

4. Lambillotte C, Gilon P, Henquin jC. Direct glucocorticoid inhibition of insulin secretion. An in vitro<br />

study of dexamethasone effects in mouse islets. j Clin Invest. 1997;99(3):414-23.<br />

5. Longano CA, Fletcher HP. Insulin release after acute hydrocortisone treatment in mice. Metabolism.<br />

1983;32(6):603-8.<br />

6. Kalhan SC, Adam PA. Inhibitory effect of prednisone on insulin secretion in man: model for<br />

duplication of blood glucose concentration. j Clin Endocrinol Metab. 1975;41(3):600-10.<br />

7. Ogawa A, johnson jH, Ohneda M, McAllister CT, Inman L, Alam T, Unger RH. Roles of insulin resistance<br />

and beta-cell dysfunction in dexamethasone-induced diabetes. j Clin Invest. 1992;90(2):497-504.<br />

8. Beard jC, Halter jB, Best jD, Pfeifer MA, Porte D, jr. Dexamethasone-induced insulin resistance<br />

enhances B cell responsiveness to glucose level in normal men. Am j Physiol. 1984;247(5 Pt<br />

1):E592-6.<br />

9. Grill V, Pigon j, Hartling SG, Binder C, Efendic S. Effects of dexamethasone on glucose-induced insulin<br />

and proinsulin release in low and high insulin responders. Metabolism. 1990;39(3):251-8.<br />

10. Henriksen jE, Alford F, Ward GM, Beck-Nielsen H. Risk and mechanism of dexamethasone-induced<br />

deterioration of glucose tolerance in non-diabetic first-degree relatives of NIDDM patients.<br />

Diabetologia. 1997;40(12):1439-48.<br />

11. Larsson H, Ahren B. Insulin resistant subjects lack islet adaptation to short-term dexamethasoneinduced<br />

reduction in insulin sensitivity. Diabetologia. 1999;42(8):936-43.<br />

12. Wajngot A, Giacca A, Grill V, Vranic M, Efendic S. The diabetogenic effects of glucocorticoids are more<br />

pronounced in low- than in high-insulin responders. Proc Natl Acad Sci U S A. 1992;89(13):6035-9.<br />

13. Khan A, Ostenson CG, Berggren PO, Efendic S. Glucocorticoid increases glucose cycling and inhibits<br />

insulin release in pancreatic islets of ob/ob mice. Am j Physiol. 1992;263(4 Pt 1):E663-6.<br />

14. Ohneda M, johnson jH, Inman LR, Unger RH. GLUT-2 function in glucose-unresponsive beta cells of<br />

dexamethasone-induced diabetes in rats. j Clin Invest. 1993;92(4):1950-6.<br />

15. Boden G. Role of fatty acids in the pathogenesis of insulin resistance and NIDDM. Diabetes.<br />

1997;46(1):3-10.<br />

87<br />

4<br />

Chapter 4


Chapter 4 Glucocorticoids impair beta-cell function in humans<br />

16. Newsholme P, Bender K, Kiely A, Brennan L. Amino acid metabolism, insulin secretion and diabetes.<br />

Biochem Soc Trans. 2007;35(Pt 5):1180-6.<br />

17. Nauck MA, Homberger E, Siegel EG, Allen RC, Eaton RP, Ebert R, Creutzfeldt W. Incretin effects of<br />

increasing glucose loads in man calculated from venous insulin and C-peptide responses. j Clin<br />

Endocrinol Metab. 1986;63(2):492-8.<br />

18. Romijn jA, Corssmit EP, Havekes LM, Pijl H. Gut-brain axis. Curr Opin Clin Nutr Metab Care.<br />

2008;11(4):518-21.<br />

19. Mari A, Tura A, Gastaldelli A, Ferrannini E. Assessing insulin secretion by modeling in multiple-meal<br />

tests: role of potentiation. Diabetes. 2002;51 Suppl 1:S221-6.<br />

20. Levy jC, Matthews DR, Hermans MP. Correct homeostasis model assessment (HOMA) evaluation<br />

uses the computer program. Diabetes Care. 1998;21(12):2191-2.<br />

21. Mari A, Pacini G, Murphy E, Ludvik B, Nolan jj. A model-based method for assessing insulin sensitivity<br />

from the oral glucose tolerance test. Diabetes Care. 2001;24(3):539-48.<br />

22. Seltzer HS, Allen EW, Herron AL, jr., Brennan MT. Insulin secretion in response to glycemic stimulus:<br />

relation of delayed initial release to carbohydrate intolerance in mild diabetes mellitus. j Clin Invest.<br />

1967;46(3):323-35.<br />

23. Henquin jC. Regulation of insulin secretion: a matter of phase control and amplitude modulation.<br />

Diabetologia. 2009;52(5):739-51.<br />

24. Van Cauter E, Mestrez F, Sturis j, Polonsky KS. Estimation of insulin secretion rates from C-peptide<br />

levels. Comparison of individual and standard kinetic parameters for C-peptide clearance. Diabetes.<br />

1992;41(3):368-77.<br />

25. Plat L, Byrne MM, Sturis j, Polonsky KS, Mockel j, Fery F, Van Cauter E. Effects of morning cortisol<br />

elevation on insulin secretion and glucose regulation in humans. Am j Physiol. 1996;270(1 Pt<br />

1):E36-42.<br />

26. van Raalte DH, Ouwens DM, Diamant M. Novel insights into glucocorticoid-mediated diabetogenic<br />

effects: towards expansion of therapeutic options? Eur j Clin Invest. 2009;39(2):81-93.<br />

27. jeong IK, Oh SH, Kim Bj, Chung jH, Min YK, Lee MS, Lee MK, Kim KW. The effects of dexamethasone<br />

on insulin release and biosynthesis are dependent on the dose and duration of treatment. Diabetes<br />

Res Clin Pract. 2001;51(3):163-71.<br />

88


Chapter 5<br />

Islet-cell Dysfunction Induced by Glucocorticoid<br />

Treatment: Potential Role for Altered<br />

Sympathovagal Balance?<br />

D.H. van Raalte, K.A.A. Kwa, R.E. van Genugten, M.E. Tushuizen,<br />

J.J. Holst, C.F. Deacon, J.M. Karemaker, R.J. Heine, A. Mari and M.<br />

Diamant.<br />

Metabolism, in revision


Chapter 5 Glucocorticoids, islet-cell function & ANS<br />

92<br />

ABSTRACT<br />

objectives: To assess the effects of low- and high-dose glucocorticoid (GC) treatment on<br />

pancreatic islet-cell function in healthy males and to study the potential influence of altered<br />

autonomous nervous system (ANS) balance and meal-related incretin responses.<br />

Research design and methods: In a randomized, placebo-controlled, double-blind,<br />

dose-response intervention study, 32 males (age: 21±2 years; BMI: 21.9±1.7 kg/m2 ) were<br />

allocated to prednisolone 7.5 mg once daily (n=12), prednisolone 30 mg once daily (n=12),<br />

or placebo (n=8) for two weeks using block randomization. Main outcome measures were:<br />

1) C-peptide secretion during a hyperglycemic clamp with additional arginine stimulation<br />

2) fasting and postprandial glucagon levels 3) parasympathetic activity and sympathovagal<br />

balance and 4) postprandial incretin responses.<br />

Results: Prednisolone treatment did not significantly affect first- and second-phase<br />

glucose-stimulated C-peptide secretion, however, C-peptide secretion following arginine<br />

stimulation on top of hyperglycemia (ASI-iAUC ) was dose-dependently reduced: -2.8<br />

CP<br />

(-5.2;0.2) and -3.1 (-8.8; -1.0) nmol/L.min for prednisolone 7.5 mg and prednisolone 30<br />

mg, respectively (P=0.035 vs. placebo). Fasting glucagon levels and postprandial glucagon<br />

levels were only increased by prednisolone 30 mg. Measures of the ANS and incretin<br />

responses were not significantly altered by treatment. However, changes in parasympathic<br />

activity associated with fasting plasma glucose levels (r=-0.407; P=0.03) and tended<br />

to associate with fasting glucagon concentrations (r=-0.337; P=0.07). The change in<br />

sympathovagal balance was inversely related to ASI-iAUC (r=-0.365; P=0.05). No side<br />

CP<br />

effects of prednisolone treatment were reported.<br />

Conclusions: Impairments in islet-cell function contribute to prednisolone-induced<br />

glucose intolerance in healthy men. Altered sympathovagal balance may contribute to these<br />

effects.<br />

Clinical Trial Registration Number: ISRCTN78149983


Glucocorticoids (GCs) are the cornerstone in the treatment of numerous diseases due to their<br />

potent anti-inflammatory and immunosuppressive actions [1]. However, pharmacological GC<br />

levels also induce adverse effects on glucose metabolism [1, 2]. In population-based studies,<br />

GC therapy was associated with incident diabetes [3]. Classically, the association between GCs<br />

and diabetes has been attributed to GC-induced insulin resistance [4].<br />

The extent to which pancreatic islet-cell dysfunction, and particularly beta-cell dysfunction,<br />

contributes to the diabetogenic effects of GCs is less well-known. In vitro, GCs were shown<br />

to decrease insulin secretion and insulin synthesis [2]. In addition to reducing glucosestimulated<br />

insulin secretion (GSIS), GCs impaired the in vitro effects of nonmetabolizable<br />

insulin secretagogues, including arginine and acetylcholine, suggesting that the site of action<br />

of GCs is in the end of the insulin secretory process [5]. Transgenic mice with a beta-cell<br />

specific overexpression of the glucocorticoid receptor (GR) develop diabetes due to beta-cell<br />

failure, in the presence of increased α-2 adrenergic activity [6].<br />

In humans, a single high-dose of prednisolone was shown to impair both first-phase glucosestimulated<br />

and arginine-stimulated C-peptide secretion during a hyperglycemic clamp [7] and<br />

glucose sensitivity of the beta cell during a meal challenge test [8]. (Sub)acute, GC-exposure<br />

(a 2 day-treatment), however, generated seemingly opposing results. A number of studies<br />

reported increased insulin secretion during a hyperglycemic clamp or intravenous glucose<br />

tolerance test following this treatment duration with high-dose GCs [9-13]. This most likely<br />

can be attributed to reduced insulin sensitivity. Only one of these studies measured insulin<br />

sensitivity with the hyperinsulinemic-euglycemic clamp [11], allowing adjustment prevailing<br />

insulin sensitivity. This study indicated impaired compensation in several subjects.<br />

In addition to beta-cell function, GCs may also affect pancreatic alpha-cell function. Two<br />

studies showed increased glucagon secretion during high-dose GC treatment [14, 15].<br />

However, there are no data available regarding the effects of more prolonged GC treatment on<br />

islet-cell function in humans. Also, the dose-dependency of these effects is largely unknown.<br />

Finally, mechanisms that could contribute to GC-induced effects on islet cell function in<br />

humans have, to our knowledge, not been investigated. Given the preclinical data [5, 6],<br />

alterations in the autonomic nervous system (ANS) balance could be implicated in GC-induced<br />

islet effects. Whereas parasympathetic branches of the ANS are well-known to stimulate<br />

insulin secretion via acetylcholine signaling [16], sympathetic fibers decrease insulin release<br />

via catecholamine-related pathways [17] and stimulate glucagon release [18]. Also, it is at<br />

present unclear whether the incretin hormones, important regulators of postprandial isletcell<br />

function, may be involved in GC-induced islet effects.<br />

93<br />

5<br />

Chapter 5


Chapter 5 Glucocorticoids, islet-cell function & ANS<br />

Therefore, in the present study, we assessed the effects of a prolonged treatment, i.e. two<br />

weeks, with low or high-dose prednisolone, the most commonly prescribed GC, on islet-cell<br />

function in healthy normoglycemic males and cardiovascular ANS balance and meal-related<br />

incretin responses.<br />

RESEARCH DESIGN AND METHoDS<br />

Participants: Thirty-two healthy Caucasian males were recruited by local advertisement.<br />

Inclusion criteria included: age 18-35 years, body mass index (BMI) 20.0-25.0 kg/m2 , good<br />

physical health (determined by medical history, physical examination and screening blood<br />

tests) and normoglycemia as defined by fasting plasma glucose (FPG) < 5.6 mmol/L and<br />

2-h glucose < 7.8 mmol/L following a 75g oral glucose tolerance test (OGTT), performed at<br />

screening visit. Exclusion criteria were the presence of any disease, use of any medication,<br />

first-degree relative with type 2 diabetes, smoking, shift work, a history of GC use, excessive<br />

sport activities (i.e. > two times/week) and recent changes in weight or physical activity.<br />

The study was approved by an independent ethics committee and the study was conducted<br />

in accordance with the Declaration of Helsinki. All participants provided written informed<br />

consent before participation.<br />

Study design: The study was a randomized, placebo-controlled, double blind, doseresponse<br />

intervention study. Following assessment of eligibility and baseline measurements,<br />

participants were randomized to receive either prednisolone 30 mg once daily (n=12),<br />

prednisolone 7.5 mg once daily (n=12) or placebo (n=8) treatment for a period of 14 days using<br />

block randomization, as carried out by the Department of Experimental <strong>Pharma</strong>cology of the<br />

VU University Medical Center. An outline of the study design is presented in Supplementary<br />

Figure 1. At baseline and day 13 of treatment, insulin sensitivity and beta-cell function were<br />

measured in a combined euglycemic-hyperglycemic clamp procedure (Supplementary Figure<br />

2A). At baseline and day 14 of treatment, a standardized consecutive meal challenge test was<br />

performed and cardiovascular ANS function was measured in the fasted state (Supplementary<br />

Figure 2B). All measurements were conducted following a 12-h overnight fast with the<br />

subjects in the semi-supine position. Subjects refrained from drinking alcohol for a period of<br />

24h before the study days and did not perform strenuous exercise for a period of 48h before<br />

the study days. During all visits, including a follow-up visit at day 7 of treatment, safety and<br />

tolerability were assessed. A patient flow diagram is shown in Supplementary Figure 3.<br />

94


Hyperinsulinemic-euglycemic clamp and hyperglycemic clamp: After an overnight fast,<br />

participants were admitted to the clinical research unit at 7.30 AM. An indwelling cannula<br />

was inserted into an antecubital vein for infusion of glucose and insulin. To obtain arterialized<br />

venous blood samples, a retrograde cannula was inserted in a contralateral wrist vein and<br />

the hand placed in a heated box, maintained at 50°C. A primed, continuous (40 mU/m2 .min)<br />

insulin infusion (Actrapid; Novo Nordisk, Bagsvaerd, Denmark) was given for 120 min;<br />

plasma glucose was kept at 5 mmol/L by a variable infusion of 20% glucose, as described<br />

previously [19]. The hyperglycemic clamp was started 60 min after cessation of exogenous<br />

insulin infusion. Plasma glucose concentration was then raised to 10 mmol/L by a body<br />

weight-adjusted intravenous bolus of 20% glucose and a variable 20% glucose infusion was<br />

adjusted to maintain the targeted glucose level. After 80 min hyperglycemia, an intravenous<br />

bolus of 5 g arginine (dissolved in 50 mL NaCl) was given over 45 seconds, and the glucose<br />

level was maintained at 10 mmol/L for an additional 30 min (Supplementary Figure 2A).<br />

Standardized consecutive meal challenge: After an overnight fast, participants were<br />

admitted to the clinical research unit at 7.30 AM. An indwelling cannula was inserted to allow<br />

blood sampling during the test. Two consecutive identical meals were served as breakfast<br />

at 09.00 AM and as lunch at 1.00 PM, each containing 905 kcal (50 g fat, 75 g carbohydrates,<br />

35 g protein). Samples for determination of glucose, insulin, C-peptide, glucagon, glucagonlike<br />

peptide (GLP)-1 and glucose-dependent insulinotropic polypeptide (GIP) were obtained<br />

at times 0, 5, 10, 20, 30, 60, 90, 120, 150, 180, 210 and 240 min following each meal, with<br />

the meal beginning immediately after the time 0 sample and being consumed within 15 min<br />

(Supplementary Figure 2B).<br />

Heart rate variability: Prior to consumption of the first meal, continuous finger arterial<br />

blood pressure (BP) and heart rate (HR) were recorded (Portapres, FMS, Amsterdam, The<br />

Netherlands) in the supine position for 30 min on a beat-to-beat basis as described previously<br />

[20]. From the arterial pressure signal, interbeat interval was derived (Beatscope software1.1,<br />

FMS, Amsterdam, The Netherlands). Power spectral analysis was assessed by discrete Fourier<br />

transform as described previously [21]. The low-frequency band (LF; 0.04-0.15 Hz) and the<br />

high frequency band (HF; 0.15-0.4 Hz) were selected. LF and HF bands were expressed in<br />

normalized units (LF and HF , respectively), where HF represents parasympathetic<br />

norm norm norm<br />

activity. In addition, the LF/HF ratio was computed as a measure of sympathovagal balance<br />

[21].<br />

Study medication: Prednisolone tablets were purchased from Pfizer AB (Sollentuna,<br />

Sweden) and placebo tablets were obtained from Xendo Drug Development (Groningen, The<br />

95<br />

5<br />

Chapter 5


Chapter 5 Glucocorticoids, islet-cell function & ANS<br />

Netherlands). Tablets were capsulated in order to allow the treatment to be blinded [8]. Study<br />

medication was taken at 08.00 AM during the two-week treatment except for day 13 and<br />

day 14, when it was ingested at 06.00 AM. Patients kept a diary in which the exact time of<br />

medication intake during the study was registered.<br />

Analytical determinations: Blood glucose concentrations were measured using an YSI 2300<br />

STAT Plus analyzer (YSI, Yellow Springs, OH). Insulin and C-peptide levels were determined<br />

using an immunometric assay (Advia, Centaur, Siemens Medical Solutions Diagnostics, USA).<br />

Glucagon concentrations were determined by radioimmuno assay (Linco Research, St. Louis,<br />

USA). Total GLP-1 levels were measured using a C-terminally directed radioimmunoassay<br />

(antiserum no. 89390) as described previously [22]. Total GIP was analyzed using a newly<br />

developed assay, employing a C-terminally directed antiserum (code no 80867) raised in<br />

rabbits immunized with a C-terminal fragment of GIP [GIP (28-42)] conjugated to keyhole<br />

limpet haemocyanin via its N-terminus. This assay has broadly the same specificity and<br />

characteristics as the previously published assay using antiserum R65 [23], recognizing<br />

equally both intact GIP (1-42) and the primary metabolite, GIP (3-42).<br />

Data analyses: From the hyperglycemic clamp the beta-cell function parameters first-phase<br />

(min 0-10) and second-phase (min 10-80) incremental area under the C-peptide curve<br />

(iAUC ), as well as arginine-stimulated C-peptide secretion (ASI- iAUC ) (min 80-110) were<br />

CP CP<br />

calculated using the trapezoid method (nmol/L.min). From the hyperinsulinemic-euglycemic<br />

clamp whole-body insulin sensitivity was quantified by the M-value (mg/kg.min), calculated<br />

between min 90-120 during steady-state insulin concentrations as described previously [19].<br />

During the double meal challenge absolute area under the curves (AUCs) for glucose, insulin,<br />

C-peptide, glucagon, GLP-1 and GIP were calculated using the trapezoid method.<br />

Beta-cell function during the meal challenge test: Beta-cell function during the standardized<br />

double meal challenge test was assessed by mathematical modeling which was described in<br />

detail previously [24]. The model describes the relationship between insulin secretion and<br />

glucose concentration as the sum of two components. The first component represents the<br />

dependence of insulin secretion on absolute glucose concentrations at any time point and<br />

is characterized by a dose-response function relating the two variables. The characteristic<br />

parameter of the dose response is its mean slope, denoted here as glucose sensitivity. The<br />

dose response is modulated by both glucose-mediated and non-glucose-mediated factors<br />

(i.e. non-glucose substrates, gastrointestinal hormones, and neurotransmitters), which are<br />

collectively modeled as a potentiation factor. The excursion of the potentiation factor was<br />

quantified using a ratio between mean values at times 160-180 min and 0–20 min, and is<br />

96


called the potentiation factor ratio (PFR). In addition, the fasting secretory tone is calculated<br />

from the dose-response curve as insulin secretion at the glucose concentration of 4.5 mmol/L.<br />

The second component of the model describes the insulin response to the rate of change of<br />

glucose concentration. This component is termed rate sensitivity, which is related to early<br />

insulin release [24].<br />

Statistical analyses: Data are presented as mean values ± standard deviation (SD), or as<br />

median (interquartile range) in case of skewed distribution. Non-parametric analysis was<br />

chosen due to uneven sample size over the groups, the relatively small N, and unequal variances<br />

that were observed for some parameters. Between-group comparisons of baseline values<br />

were performed using Kruskal-Wallis test. For treatment-induced effects, absolute changes<br />

from baseline were calculated (on-treatment value minus pre-treatment value) and were<br />

compared by Kruskal-Wallis test with trend analysis (‘the Jonckheere-Terpstra test’). Only in<br />

case of a significant finding, prednisolone 7.5 mg and prednisolone 30 mg were compared<br />

against placebo by posthoc testing, using the Mann-Whitney U test. To correct for multiple<br />

testing, Bonferroni correction was applied. Correlations between the various parameters<br />

were assessed with Pearson’s correlations. All statistical analyses were run on SPSS version<br />

15 for Windows (Chicago, IL, USA). A P < 0.05 was considered statistically significant.<br />

RESULTS<br />

Anthropometric characteristics: No significant differences in subject characteristics were<br />

observed among the groups at baseline (Table 1). Body weight was not altered by prednisolonetreatment<br />

irrespective of the dose (Supplementary Table 1). Systolic blood pressure (6±1.2<br />

mmHg increase; P=0.006), but not diastolic blood pressure was raised by prednisolone 30<br />

mg. Prednisolone 7.5 mg did not affect blood pressure (Supplementary Table 1).<br />

Fasting glucose and hormone levels: A dose-dependent rise in fasting plasma glucose<br />

levels was observed by prednisolone treatment relative to placebo (P=0.04) (Table 2).<br />

Fasting plasma insulin levels were significantly increased (P=0.008) in the prednisolone 30<br />

mg arm. The Prednisolone-induced increase in fasting insulin levels was due to increased<br />

basal secretion, not altered clearance, since fasting C-peptide levels were similarly enhanced<br />

(P=0.001) (Table 2).<br />

97<br />

5<br />

Chapter 5


Chapter 5 Glucocorticoids, islet-cell function & ANS<br />

Table 1. Subject characteristics at inclusion<br />

Placebo Prednisolone<br />

7.5 mg 30 mg P<br />

N 8 12 12<br />

Age (y) 21±3 21±2 21±2 0.670<br />

Weight (kg) 75±8 75±6 73±10 0.886<br />

Height (cm) 186±7 184±6 185±5 0.718<br />

BMI (kg m-2 ) 21.6±1.5 22.1±1.5 21.4±2.2 0.656<br />

Systolic blood pressure (mmHg) 126±7 124±9 120±9 0.284<br />

Diastolic blood pressure (mmHg) 79±8 79±6 78±10 0.872<br />

Fasting plasma glucose (mmol/l) 4.4±0.3 4.4±0.2 4.5±0.2 0.852<br />

2-hr glucose OGTT (mmol/l) 3.7±1.0 3.9±0.9 3.6±0.8 0.782<br />

Data are mean±SD. Significance was tested by Kruskal-Wallis. No statistically significant differences were<br />

observed between the groups at baseline. 2-hr glucose OGTT denotes plasma glucose concentrations 2 h<br />

after ingestion of 75 g glucose during an oral glucose tolerance test.<br />

Table 2. Fasting metabolic parameters before and on day 14 of treatment with placebo, prednisolone 7.5<br />

mg once daily and prednisolone 30 mg once daily.<br />

Fasting plasma glucose<br />

(mmol/l)<br />

Fasting plasma insulin<br />

(pmol/l)<br />

Fasting C-peptide<br />

(nmol/l)<br />

Fasting glucagon<br />

(pmol/l)<br />

98<br />

Pre-treatment On-treatment P1 P2 P3<br />

PLB 4.4±0.3 4.5±0.3<br />

7.5 4.4±0.2 4.7±0.3<br />

0.04 0.418 0.09<br />

30 4.5±0.2 4.9±0.4<br />

PLB 31 (28-40) 33 (28-39)<br />

7.5 36 (29-47) 36 (27-65) 0.001 1.0 0.008<br />

30 32 (26-44) 56 (41-72)<br />

PLB 0.4±0.1 0.4±0.1<br />

7.5 0.4±0.1 0.5±0.2<br />

0.001 0.918 0.002<br />

30 0.4±0.1 0.6±0.2<br />

PLB 12±4 10±3<br />

7.5 12±3 13±3<br />

0.002 0.152 0.014<br />

30 12±3 15±3<br />

Data are mean±SD or median (interquartile range). Between-group changes from baseline were tested<br />

by Kruskal-Wallis with trend analysis (P1). In case of a significant finding, posthoc testing (by Mann-<br />

Whitney U) was done (P2 and P3). Treatment groups: PLB=placebo; 7.5=prednisolone 7.5 mg daily;<br />

30=prednisolone 30 mg daily.<br />

Hyperglycemic clamp: Prednisolone treatment tended to increase first- and second-phase<br />

glucose-stimulated C-peptide secretion (Figure 1A-B). In multivariate analysis however,<br />

this trend was no longer observed when the M-value was added in the model (1st-phase


iAUC: β=0.126; P=0.638; R 2 =0.119 and 2 nd -phase iAUC: β=0.051; P=0.847; R 2 =0.152).<br />

C-peptide secretion following arginine stimulation on top of hyperglycemia (ASI-iAUC CP )<br />

was significantly and dose-dependently reduced by prednisolone treatment: -2.7 (-5.2;-0.3)<br />

nmol/L.min for prednisolone 7.5 mg and -3.0 (-7.6;-0.2) ) nmol/L.min for prednisolone 30 mg<br />

(P=0.035) (Figure 1C). In multivariate analysis, this relation was independent of changes in<br />

insulin sensitivity. The results from the hyperglycemic clamp were similar when insulin iAUCs<br />

were calculated (data not shown).<br />

Euglycemic clamp: Prednisolone treatment dose-dependently decreased insulin sensitivity<br />

(M-value) as compared to placebo: mean differences: -2.1±0.8 mg kg-1 min-1 for prednisolone 7.5<br />

mg and -4.5±0.7 mg kg-1 min-1 for prednisolone 30 mg (Figure 2). Insulin levels reached steady<br />

state during min 90-120 of the euglycemic clamp at 490 ± 77 pmol/L prior to treatment, and<br />

were not altered during the on-treatment clamps (Supplementary Table 2). Adjustment of the<br />

M-value by insulin levels during the steady-state part of the clamp (M/I) did not affect the results.<br />

Figure 1. The effects of prednisolone treatment on first-phase- (A) and second-phase (B) glucosestimulated,<br />

and arginine-stimulated C-peptide secretion (C) during the hyperglycemic clamp.<br />

Prednisolone 30 mg tended to increase first-and second-phase C-peptide secretion. However, both<br />

dosages of prednisolone significantly decreased C-peptide secretion following arginine administration on<br />

top of hyperglycemia. Box-and-Whisker plots (min-max) are shown. White bars: before treatment; black<br />

bar: on treatment. Between-group changes from baseline were tested by Kruskal-Wallis test with trend<br />

analysis (indicated by top line). Posthoc tests were done by Mann-Whitney U with Bonferroni correction<br />

for multiple testing (indicated by line with brackets). *P


Chapter 5 Glucocorticoids, islet-cell function & ANS<br />

sensitivity (Figure 4A-B). PFR was significantly decreased by prednisolone 30 mg, but not by<br />

prednisolone 7.5 mg (Figure 4C). Basal insulin secretion at a fixed glucose level of 4.5 mmol/L<br />

tended to decrease following prednisolone treatment (P=0.052) (Figure 4D).<br />

Figure 2. Prednisolone treatment dose-dependently decreased insulin sensitivity during the<br />

hyperinsulinemic-euglycemic clamp. Box-and-Whisker plots with the absolute change from baseline are<br />

shown. Between-group changes from baseline were tested by Kruskal-Wallis test with trend analysis<br />

(indicated by top line). Posthoc tests were done by Mann-Whitney U with Bonferroni correction for<br />

multiple testing (indicated by line with brackets). ***P


Figure 3. Postprandial responses at baseline and at 14 day of intervention. Panel A: Glucose concentrations.<br />

Panel B: Insulin concentrations. Panel C: C-peptide concentrations. Panel D: Glucagon-like peptide (GLP)-<br />

1 concentrations. Panel E: Glucose-dependent insulinotropic polypeptide (GIP) concentrations. Panel F:<br />

Glucagon concentrations. Straight line with black squares denotes pre-treatment. Dotted line with open<br />

circles represents on-treatment. Box-and-Whisker plots with absolute change in area under the curve<br />

(AUC) from baseline are shown. Between-group changes from baseline were tested by Kruskal-Wallis test<br />

with trend analysis (indicated by top line). Posthoc tests were done by Mann-Whitney U with Bonferroni<br />

correction for multiple testing (indicated by line with brackets). ***P


Chapter 5 Glucocorticoids, islet-cell function & ANS<br />

Figure 4. Results from the modeling analysis of beta-cell function from glucose and C-peptide levels<br />

during 2 consecutive standardized meals given as breakfast (t=0) and as lunch (t=240 min). Glucose<br />

sensitivity of the beta cell (A) and rate sensitivity (B) were not altered by study medication. Potentiation<br />

factor ratio between T=160-180 min and T=0-20 min was significantly decreased by prednisolone 30 mg<br />

(C). Finally, prednisolone treatment non-significantly reduced fasting insulin secretion rates at a fixed<br />

glucose level of 4.5 mmol/L. Box-and-Whisker plots (min-max) are shown. White bars: before treatment;<br />

black bar: on treatment. Between-group changes from baseline were tested by Kruskal-Wallis test with<br />

trend analysis (indicated by top line). Posthoc tests were done by Mann-Whitney U with Bonferroni<br />

correction for multiple testing (indicated by line with brackets). **P


Safety and tolerability: One subject in the prednisolone 30 mg group complained of<br />

sleeplessness, which was mild and transient of nature. Otherwise, no side effects were<br />

reported in any of the treatment arms.<br />

DISCUSSIoN<br />

The principal findings of the present study are that a two-week treatment with high-dose<br />

prednisolone impaired various parameters of pancreatic islet-cell function, and that<br />

altered ANS balance may be involved in these changes, although ANS balance itself was<br />

not significantly altered. Furthermore in this study, we observed a clear dose-dependency<br />

of prednisolone-induced effects. However, probably due to lack of power, the effects of lowdose<br />

prednisolone treatment did not reach statistical significance. At increased levels, GCs<br />

induce glucose intolerance and diabetes in susceptible individuals, which has classically been<br />

attributed to GC-induced insulin resistance (4). Indeed, our study confirmed the presence<br />

of GC-induced insulin resistance: prednisolone treatment dose-dependently reduced clampmeasured<br />

M-value.<br />

Figure 6. The relation between treatment-induced changes in insulin sensitivity and 1st phase glucosestimulated<br />

C-peptide secretion for placebo (A), prednisolone 7.5 mg daily (B) and prednisolone 30 mg<br />

daily (C). As demonstrated in (C), during treatment with prednisolone 30 mg, C-peptide secretion is<br />

enhanced to compensate for reduced insulin sensitivity. Black squares: pre-treatment. Open circles: ontreatment.<br />

Grey triangle: mean pre-treatment. Open triangle: mean on-treatment.<br />

The effects of short-term GC treatment on islet-cell function, and particularly beta-cell<br />

function, have been under debate. The observation that 2 days GC exposure induced fasting<br />

hyperinsulinemia and increased insulin secretion in response to oral and intravenous glucose<br />

loads, may suggest a lowering of insulin sensitivity by GCs [9-13]. In the present study, we<br />

similarly observed elevated fasting insulin levels, postprandial hyperinsulinemia and a<br />

tendency towards increased first- and second-phase glucose-stimulated C-peptide secretion.<br />

Using multivariate analysis, we demonstrated in this study that increased 1st and 2nd phase<br />

103<br />

5<br />

Chapter 5


Chapter 5 Glucocorticoids, islet-cell function & ANS<br />

C-peptide secretion was driven by treatment-related reduction in insulin sensitivity, showing<br />

adequate compensation for prednisolone-induced insulin resistance (Figure 6), and did not<br />

indicate improved beta-cell function per se. Interestingly, prednisolone treatment reduced<br />

arginine-stimulated C-peptide secretion, a measure of insulin secretory capacity, in a dosedependent<br />

matter.<br />

A similar pattern was observed during the meal challenge test. Modeling analysis of glucose<br />

and C-peptide concentrations, revealed decreased glucose-adjusted insulin secretion rates<br />

following prednisolone treatment, also at fasting glucose levels (Supplementary Figure 4;<br />

Figure 4D for insulin secretion at 4.5 mmol/L glucose). While prednisolone treatment did<br />

not affect glucose sensitivity of the beta-cell or rate sensitivity, prednisolone 30 mg markedly<br />

decreased the potentiation factor ratio, a finding that confirms data from a previous study<br />

published by our group [8]. This represents insulin secretion that is not primarily related<br />

to plasma glucose levels and may include secretion induced by non-glucose secretagogues,<br />

incretin hormones, and neuronal factors [25, 26].<br />

From both the hyperglycemic clamp test and the meal test we conclude that prednisolone<br />

induces beta-cell dysfunction, although this effect is detectable on specific beta-cell function<br />

parameters that seem to involve potentiation phenomena. Both arginine-induced secretion<br />

at 10mmol/L glucose [27] and potentiation factor ratio [24, 26] during the meal are<br />

dependent on potentiation. On the other hand, the enhancement of first- and second phase<br />

insulin secretion as well as postprandial insulin release is mainly mediated by an increase in<br />

glucose levels and a compensatory response to prednisolone-induced insulin resistance. In<br />

addition to impairments in beta-cell function, we observed GC-induced increased fasting and<br />

postprandial glucagon levels during high-dose prednisolone treatment. Thus, prednisolone<br />

treatment altered islet-cell functional balance.<br />

Additionally, we evaluated mechanisms possibly underlying the effects of GCs on islet-cell<br />

function. First, we evaluated the role of the ANS system. Whereas catecholamines released<br />

by the sympathetic nervous system inhibit insulin secretion via a -adrenergic receptor<br />

2<br />

(AR) signaling and stimulate glucagon release, acetylcholine released by parasympathetic<br />

nerves stimulates insulin release via protein kinase C-related pathways [2]. In addition,<br />

parasympathetic nerve fibers increase islet blood flow, thus facilitating increased insulin<br />

secretion [28]. In beta-cell lines, GCs decreased the efficacy of acetylcholine to release<br />

insulin [5], and also upregulated expression and signaling of a -ARs [29]. Transgenic mice<br />

2<br />

with beta-cell specific overexpression of the GR were shown to develop diabetes through<br />

beta-cell failure consequential to increased a -AR expression. We observed a tendency<br />

2<br />

104


towards withdrawal of vagal activity following high-dose prednisolone treatment, which<br />

was negatively associated with fasting glucose and glucagon levels. In addition, alterations<br />

in sympathovagal balance, expressed as LF/HF ratio, were negatively related to changes in<br />

C-peptide secretion in response to arginine. The latter finding is partly in line with a previous<br />

study, in which interruption of cholinergic transmission by trimethaphan impaired argininestimulated<br />

insulin secretion during treatment with dexamethasone [30]. However, in that<br />

acute study, a 2-day treatment with high-dose dexamethasone, increased arginine-stimulated<br />

secretion, whereas we observed a decline in this beta-cell function parameter. Differences in<br />

treatment duration and study population (in that study, highly insulin sensitivity participants,<br />

as determined by hyperinsulinemic-euglycemic clamp prior to inclusion, were studied) may<br />

have contributed to these seemingly opposing results. In addition, dexamethasone is a pure<br />

GR agonist, whereas prednisolone also activates the mineralocorticoid receptor. Various GC<br />

compounds, depending on their receptor specificity, may have a different effect on measures<br />

of cardiovascular autonomic function and variables such as blood pressure and heart rate,<br />

making the compounds difficult to compare [31, 32].<br />

Second, we measured incretin responses following prednisolone treatment. The incretin<br />

hormones GLP-1 and GIP substantially contribute to glucose tolerance and we hypothesized<br />

that prednisolone treatment would reduce incretin levels [33]. However, we did not observe<br />

declined plasma levels of GLP-1 and GIP following prednisolone treatment, confirming<br />

a recently published study, in which high-dose prednisolone treatment combined with a<br />

hypercaloric diet and physical inactivity did not decrease postprandial levels of GLP-1 and<br />

GIP [14]. However, in the same study, prednisolone treatment was shown to reduce the<br />

insulinotropic effects of GLP-1 and GIP. Thus, an impaired incretin effect at the level of the<br />

beta cell may in fact contribute to GC-induced hyperglycemia. Future in vitro studies will need<br />

to address this hypothesis.<br />

The observations done in the present study may have important implications for the treatment<br />

of GC-induced glucose intolerance and diabetes. Our data indicate that pharmacological<br />

measures that aim to improve pancreatic islet-cell function may particularly be effective in<br />

restoring glucose tolerance during prednisolone treatment. In line with this hypothesis, we<br />

have recently shown that the acute effects of high-dose prednisolone treatment on glucose<br />

tolerance and islet-cell function in healthy humans could be prevented by concomitant<br />

treatment with the GLP-1 receptor agonist exenatide [7]. Further studies in relevant<br />

populations should explore the full potential of incretin-based therapies to prevent GCinduced<br />

glucose intolerance.<br />

105<br />

5<br />

Chapter 5


Chapter 5 Glucocorticoids, islet-cell function & ANS<br />

We conclude that, in addition to reducing insulin sensitivity, prolonged prednisolone treatment<br />

dose-dependently impairs islet-cell function in healthy males. Our data furthermore suggest<br />

that changes in ANS balance may contribute to these GC-related changes.<br />

Funding: This paper was written within the framework of the Dutch Top Institute <strong>Pharma</strong><br />

T1.106. Drs. Renate van Genugten is supported by the EFSD/MSD Clinical Research<br />

Programme 2008. The authors thank dr. M.W. Heymans of the department of Epidemiology<br />

and Biostatistics of the VU University Medical Center for his help with the statistical analyses.<br />

106


REFERENCES<br />

1. Schacke H, Docke WD, Asadullah K. Mechanisms involved in the side effects of glucocorticoids.<br />

<strong>Pharma</strong>col Ther. 2002;96(1):23-43.<br />

2. van Raalte DH, Ouwens DM, Diamant M. Novel insights into glucocorticoid-mediated diabetogenic<br />

effects: towards expansion of therapeutic options? Eur j Clin Invest. 2009;39(2):81-93.<br />

3. Gulliford MC, Charlton j, Latinovic R. Risk of diabetes associated with prescribed glucocorticoids in<br />

a large population. Diabetes Care. 2006;29(12):2728-9.<br />

4. McMahon M, Gerich j, Rizza R. Effects of glucocorticoids on carbohydrate metabolism. Diabetes<br />

Metab Rev. 1988;4(1):17-30.<br />

5. Lambillotte C, Gilon P, Henquin jC. Direct glucocorticoid inhibition of insulin secretion. An in vitro<br />

study of dexamethasone effects in mouse islets. j Clin Invest. 1997;99(3):414-23.<br />

6. Davani B, Portwood N, Bryzgalova G, Reimer MK, Heiden T, Ostenson CG, Okret S, Ahren B, Efendic<br />

S, Khan A. Aged transgenic mice with increased glucocorticoid sensitivity in pancreatic beta-cells<br />

develop diabetes. Diabetes. 2004;53 Suppl 1:S51-9.<br />

7. van Raalte DH, van Genugten RE, Linssen MM, Ouwens DM, Diamant M. Glucagon-like peptide-1<br />

receptor agonist treatment prevents glucocorticoid-induced glucose intolerance and islet-cell<br />

dysfunction in humans. Diabetes Care. 2011;34(2):412-7.<br />

8. van Raalte DH, Nofrate V, Bunck MC, van Iersel T, Elassaiss Schaap j, Nassander UK, Heine Rj, Mari<br />

A, Dokter WH, Diamant M. Acute and 2-week exposure to prednisolone impair different aspects of<br />

beta-cell function in healthy men. Eur j Endocrinol. 2010;162(4):729-35.<br />

9. Beard jC, Halter jB, Best jD, Pfeifer MA, Porte D, jr. Dexamethasone-induced insulin resistance<br />

enhances B cell responsiveness to glucose level in normal men. Am j Physiol. 1984;247(5 Pt<br />

1):E592-6.<br />

10. Henriksen jE, Alford F, Ward GM, Beck-Nielsen H. Risk and mechanism of dexamethasone-induced<br />

deterioration of glucose tolerance in non-diabetic first-degree relatives of NIDDM patients.<br />

Diabetologia. 1997;40(12):1439-48.<br />

11. Larsson H, Ahren B. Insulin resistant subjects lack islet adaptation to short-term dexamethasoneinduced<br />

reduction in insulin sensitivity. Diabetologia. 1999;42(8):936-43.<br />

12. Matsumoto K, Yamasaki H, Akazawa S, Sakamaki H, Ishibashi M, Abiru N, Uotani S, Matsuo H,<br />

Yamaguchi Y, Tokuyama K, Nagataki S. High-dose but not low-dose dexamethasone impairs glucose<br />

tolerance by inducing compensatory failure of pancreatic beta-cells in normal men. j Clin Endocrinol<br />

Metab. 1996;81(7):2621-6.<br />

13. Wajngot A, Giacca A, Grill V, Vranic M, Efendic S. The diabetogenic effects of glucocorticoids are more<br />

pronounced in low- than in high-insulin responders. Proc Natl Acad Sci U S A. 1992;89(13):6035-9.<br />

107<br />

5<br />

Chapter 5


Chapter 5 Glucocorticoids, islet-cell function & ANS<br />

14. Hansen KB, Vilsboll T, Bagger jI, Holst jj, Knop FK. Increased postprandial GIP and glucagon<br />

responses, but unaltered GLP-1 response after intervention with steroid hormone, relative physical<br />

inactivity, and high-calorie diet in healthy subjects. j Clin Endocrinol Metab. 2011;96(2):447-53.<br />

15. Wise JK, Hendler R, Felig P. Influence of glucocorticoids on glucagon secretion and plasma amino<br />

acid concentrations in man. j Clin Invest. 1973;52(11):2774-82.<br />

16. Gilon P, Henquin JC. Mechanisms and physiological significance of the cholinergic control of<br />

pancreatic beta-cell function. Endocr Rev. 2001;22(5):565-604.<br />

17. Campfield LA, Smith FJ. Neural control of insulin secretion: interaction of norepinephrine and<br />

acetylcholine. Am j Physiol. 1983;244(5):R629-34.<br />

18. Ahren B. Autonomic regulation of islet hormone secretion--implications for health and disease.<br />

Diabetologia. 2000;43(4):393-410.<br />

19. DeFronzo RA, Tobin jD, Andres R. Glucose clamp technique: a method for quantifying insulin<br />

secretion and resistance. Am j Physiol. 1979;237(3):E214-23.<br />

20. Imholz BP, Wieling W, van Montfrans GA, Wesseling KH. Fifteen years experience with finger arterial<br />

pressure monitoring: assessment of the technology. Cardiovasc Res. 1998;38(3):605-16.<br />

21. Heart rate variability. Standards of measurement, physiological interpretation, and clinical use.<br />

Task Force of the European Society of Cardiology and the North American Society of Pacing and<br />

Electrophysiology. Eur Heart j. 1996;17(3):354-81.<br />

22. Orskov C, Rabenhoj L, Wettergren A, Kofod H, Holst jj. Tissue and plasma concentrations of amidated<br />

and glycine-extended glucagon-like peptide I in humans. Diabetes. 1994;43(4):535-9.<br />

23. Deacon CF, Nauck MA, Meier j, Hucking K, Holst jj. Degradation of endogenous and exogenous gastric<br />

inhibitory polypeptide in healthy and in type 2 diabetic subjects as revealed using a new assay for<br />

the intact peptide. j Clin Endocrinol Metab. 2000;85(10):3575-81.<br />

24. Mari A, Tura A, Gastaldelli A, Ferrannini E. Assessing insulin secretion by modeling in multiple-meal<br />

tests: role of potentiation. Diabetes. 2002;51 Suppl 1:S221-6.<br />

25. Henquin jC. Regulation of insulin secretion: a matter of phase control and amplitude modulation.<br />

Diabetologia. 2009;52(5):739-51.<br />

26. Bonuccelli S, Muscelli E, Gastaldelli A, Barsotti E, Astiarraga BD, Holst jj, Mari A, Ferrannini E.<br />

Improved tolerance to sequential glucose loading (Staub-Traugott effect): size and mechanisms. Am<br />

j Physiol Endocrinol Metab. 2009;297(2):E532-7.<br />

27. Taborsky Gj, jr., Smith PH, Halter jB, Porte D, jr. Glucose infusion potentiates the acute insulin response<br />

to nonglucose stimuli during the infusion of somatostatin. Endocrinology. 1979;105(5):1215-20.<br />

28. Atef N, Laury MC, N’Guyen JM, Mokhtar N, Ktorza A, Penicaud L. Increased pancreatic islet blood<br />

flow in 48-hour glucose-infused rats: involvement of central and autonomic nervous systems.<br />

Endocrinology. 1997;138(5):1836-40.<br />

108


29. Hamamdzic D, Duzic E, Sherlock jD, Lanier SM. Regulation of alpha 2-adrenergic receptor expression<br />

and signaling in pancreatic beta-cells. Am j Physiol. 1995;269(1 Pt 1):E162-71.<br />

30. Ahren B. Evidence that autonomic mechanisms contribute to the adaptive increase in insulin secretion<br />

during dexamethasone-induced insulin resistance in humans. Diabetologia. 2008;51(6):1018-24.<br />

31. Dodt C, Keyser B, Molle M, Fehm HL, Elam M. Acute suppression of muscle sympathetic nerve activity<br />

by hydrocortisone in humans. Hypertension. 2000;35(3):758-63.<br />

32. Dodt C, Wallin G, Fehm HL, Elam M. The stress hormone adrenocorticotropin enhances sympathetic<br />

outflow to the muscle vascular bed in humans. J Hypertens. 1998;16(2):195-201.<br />

33. Zhang R, Packard BA, Tauchi M, D’Alessio DA, Herman JP. Glucocorticoid regulation of preproglucagon<br />

transcription and RNA stability during stress. Proc Natl Acad Sci U S A. 2009;106(14):5913-8.<br />

109<br />

5<br />

Chapter 5


Chapter 5 Glucocorticoids, islet-cell function & ANS<br />

SUPPLEMENTARy FIGURES<br />

Supplementary Figure 1. Study Design.<br />

Day<br />

110<br />

-14<br />

S<br />

Baseline<br />

PLB, PRED 7.5 mg or PRED 30 mg QD<br />

-2 -1 1<br />

7<br />

13 14<br />

C M T<br />

V C M<br />

Offtreatment<br />

S: Screening M: Meal Test C: Clamp Test T: start treatment V: Follow-up visits<br />

Supplementary Figure 2A. Combined Hyperinsulinemic-Euglycemic and Hyperglycemic Clamp<br />

Procedure.<br />

Insulin 40 mU/m 2.min<br />

Variable glucose 20% infusion<br />

Plasma Glucose: 5.0 mM 10.0 mM<br />

0 90 120 150 0 10<br />

80 110<br />

M-value -30<br />

Time (min)<br />

Glucose bolus<br />

Supplementary Figure 2B. Double Standardized Meal Challenge Test.<br />

- 60<br />

HRV<br />

studies<br />

Time (min)<br />

5g Arginine<br />

0 60 120 180 240 300 360 420 480<br />

Meal<br />

75 g carbs<br />

50 g fat<br />

34 g protein<br />

Meal<br />

75 g carbs<br />

50 g fat<br />

34 g protein<br />

28<br />

V


Supplementary Figure 3. Patient flow diagram. 100% of the randomized participants completed the<br />

study.<br />

Supplementary Figure 4. Dose-response curve for insulin secretion and glucose levels during the meal<br />

challenge test. The dose-response curve is shifted downwards by both PRED dosages. Panel A: Placebo.<br />

Panel B: Prednisolone 7.5 mg daily. Panel C: Prednisolone 30 mg daily. Straight line with black squares<br />

denotes pre-treatment. Dotted line with open circles represents on-treatment.<br />

111<br />

5<br />

Chapter 5


Chapter 5 Glucocorticoids, islet-cell function & ANS<br />

Supplementary Table 1. Body weight and blood pressure before and during two-week treatment with<br />

placebo, prednisolone 7.5 mg once daily, or prednisolone 30 mg once daily.<br />

Weight<br />

(kg)<br />

BMI<br />

(kg/m2 )<br />

Systolic Blood Pressure<br />

(mmHg)<br />

Diastolic Blood Pressure<br />

(mmHg)<br />

112<br />

Pre-treatment On-treatment P1 P2 P3<br />

PLB 75±8 76±7<br />

7.5 75±6 75±6<br />

0.539 NA NA<br />

30 73±10 74±10<br />

PLB 21.6±1.5 21.7±1.3<br />

7.5 22.1±1.5 22.1±1.7 0.614 NA NA<br />

30 21.4±2.2 21.5±2.4<br />

PLB 126±7 123±9<br />

7.5 124±9 124±11<br />

0.002 0.710 0.006<br />

30 120±9 124±8<br />

PLB 79±8 76±7<br />

7.5 79±6 76±6<br />

0.042 1.0 0.164<br />

30 78±10 80±9<br />

Mean±SD or median (interquartile range) are provided. Between-group changes from baseline were<br />

tested by Kruskal-Wallis with trend analysis (P1). In case of a significant finding, posthoc testing (by<br />

Mann-Whitney U) was done (P2 and P3). Treatment groups: PLB=placebo; 7.5=prednisolone 7.5 mg<br />

daily; 30=prednisolone 30 mg daily.<br />

Supplementary Table 2. Clamp insulin levels before and during two-week treatment with placebo,<br />

prednisolone 7.5 mg once daily, or prednisolone 30 mg once daily.<br />

Clamp Insulin levels<br />

(pmol/l)<br />

Pre-treatment On-treatment P<br />

PLB 441±81 412±49<br />

7.5 488±61 453±69<br />

NS<br />

30 504±78 460±81<br />

Mean±SD or median (interquartile range) are provided. Between-group changes from baseline were<br />

tested by Kruskal-Wallis with trend analysis (P). Treatment groups: PLB=placebo; 7.5=prednisolone 7.5<br />

mg daily; 30=prednisolone 30 mg daily.


Chapter 6<br />

Glucocorticoid Receptor Gene Polymorphisms<br />

are Associated with Reduced First-phase<br />

Glucose-Stimulated Insulin Secretion and<br />

Disposition Index in Women, but not in Men.<br />

D.H. van Raalte*, N. van Leeuwen*, A.M. Simonis-Bik, G. Nijpels,<br />

T.W. van Haeften, S.A. Schafer, D.I. Boomsma, M.H.H. Kramer,<br />

R.J. Heine, J.A. Maassen, H. Staiger, F. Machicao, H.U. Häring, P.E.<br />

Slagboom, G. Willemsen, E.J. de Geus, J.M. Dekker, A. Fritsche, E.<br />

M. Eekhoff, M. Diamant, L.M. ‘t Hart.<br />

*Both authors contributed equally<br />

Diabetic Medicine. 2012; 29(8):e211-e216


Chapter 6 Glucocorticoid receptor SNPs & beta-cell function<br />

ABSTRACT<br />

Background and aim: Glucocorticoids (GCs) are efficacious anti-inflammatory agents,<br />

but in susceptible individuals, these drugs may induce glucose intolerance and diabetes by<br />

affecting beta-cell function and insulin sensitivity. We assessed whether polymorphisms in<br />

the glucocorticoid receptor (GR) gene NR3C1 associate with measures of beta-cell function<br />

and insulin sensitivity derived from hyperglycemic clamps in subjects with normal or<br />

impaired glucose tolerance.<br />

Methods and study design: A cross-sectional cohort study was conducted in four academic<br />

medical centers in the Netherlands and Germany. Four hundred and forty-nine volunteers<br />

(188 males; 261 females) were recruited with normal glucose tolerance (n=261) and<br />

impaired glucose tolerance (n=188). From 2-h hyperglycemic clamps, first- and secondphase<br />

glucose-stimulated insulin secretion (GSIS )as well as insulin sensitivity index and<br />

disposition index were calculated. All participants were genotyped for the functional<br />

NR3C1 polymorphisms N363S (rs6195), BclI (rs41423247), ER22/23EK (rs6189/6190),<br />

9β A/G (rs6198) and ThtIIII (rs10052957). Associations between these polymorphisms<br />

and beta-cell function parameters were assessed.<br />

Results: In women, but not in men, the N363S polymorphism was associated with reduced<br />

disposition index (P=1.06 10-4 ). Also only in women, the ER22/23EK polymorphism was<br />

associated with reduced first-phase GSIS (P=0.011) and disposition index (P=0.003). The<br />

other SNPs were not associated with beta-cell function. Finally, none of the polymorphisms<br />

was related to insulin sensitivity.<br />

Conclusion: The N363S and ER22/23EK polymorphisms of the NR3C1 gene are negatively<br />

associated with parameters of beta-cell function in women, but not in men.<br />

116


Excess glucocorticoid (GC) levels induce glucose intolerance [1, 2] and are associated with<br />

incident diabetes [3]. In addition to GC-induced insulin resistance [4], GC-induced beta-cell<br />

dysfunction is a hallmark of GC-associated adverse metabolic effects [2, 5-7]. GCs exert many<br />

effects by binding to its cytosolic glucocorticoid receptor (GR), following which the ligandactivated<br />

GR translocates to the nucleus where it regulates target gene transcriptional activity.<br />

Considerable variability exists in the sensitivity to GCs across individuals, a phenomenon that<br />

was linked to functional polymorphisms in the GR gene (NR3C1) [8-10]. As such, the NR3C1<br />

variants ER22/23EK (two Single Nucleotide Polymorphisms (SNPs) that are in complete<br />

linkage disequilibrium) [11] and 9β A/G [12] may induce relative GC resistance, whereas the<br />

NR3C1 gene variants BclI C/G [13] and N363S [14] are associated with enhanced GC sensitivity.<br />

The effects of the TthIIII polymorphism are currently less clear [15]. Importantly, several of<br />

these SNPs have been linked to metabolic parameters. In some studies, GC resistance was<br />

associated with insulin sensitivity, increased lean body mass and reduced waist circumference<br />

[11, 16, 17]. In contrast, GC sensitivity may be associated with a less favorable metabolic<br />

profile [18, 19]. Importantly, gender-specific effects have frequently been observed [8-10,<br />

17, 19], e.g. the ER22/23EK variant was associated with beneficial body composition, muscle<br />

strength and metabolic profile in men, but not in women [16].<br />

It is currently unknown whether these NR3C1 gene polymorphisms affect beta-cell function.<br />

Interestingly, mice overexpressing the GR in beta cells become diabetic because of beta-cell<br />

failure [20]. We hypothesized that alterations in GC sensitivity due to SNPs in the NR3C1<br />

gene could relate to beta-cell function. This hypothesis was addressed for the first time in the<br />

present study, where 449 subjects were genotyped for NR3C1 polymorphisms and beta-cell<br />

function was measured by the gold-standard hyperglycemic clamp.<br />

Research design and methods<br />

Cohorts: 449 Caucasian subjects were recruited from three independent studies from the<br />

Netherlands and one from Germany [21-25]. Characteristics and inclusion criteria of the<br />

separate cohorts are provided in supplemental Table 1 and 2.<br />

Hyperglycemic clamp procedure: All participants underwent a hyperglycemic clamp at 10<br />

mmol/l glucose for at least two hours as described previously [21, 22, 24, 25]. First-phase<br />

glucose-stimulated insulin secretion (GSIS) was computed as the sum of the insulin levels<br />

during the first 10 minutes of the clamp. Second-phase GSIS was determined as the mean<br />

insulin level during the last 40 min of the second hour of the clamp (80-120 min). The insulin<br />

sensitivity index (ISI) was defined as the glucose infusion rate (M, mmol/min.kg) divided by<br />

the plasma insulin concentration (I, pmol/l) during the last 40 min of the clamp (M/I, mmol/<br />

117<br />

6<br />

Chapter 6


Chapter 6 Glucocorticoid receptor SNPs & beta-cell function<br />

min.kg.pmol/l), which was shown to correlate well with insulin sensitivity measured by the<br />

hyperinsulinemic-euglycemic clamp [26]. Insulin secretion adjusted for insulin sensitivity<br />

was expressed as the disposition index (DI), calculated by multiplying first-phase GSIS and<br />

ISI [27].<br />

Genotyping: Based on the available literature, 5 SNPs were genotyped: TthIIII (rs10052957),<br />

ER22/23EK (rs6189/6190), N363S (rs6195), BclI site (rs41423247) and 9β A/G (rs6198),<br />

using the Sequenom platform (Sequenom, San Diego, California, USA). The genotyping success<br />

rate was above 98% for all SNPs and samples measured in duplicate (~5%) were in complete<br />

concordance. The SNPs did not deviate from Hardy-Weinberg equilibrium (HWE, Haploview<br />

program (MIT, Harvard Broad Institute, Cambridge, MA, USA)). Individual haplotypes were<br />

constructed using SNPHAP (http://www-gene.cimr.cam.ac.uk/clayton/software/snphap.<br />

txt).<br />

Statistics: The effect of the SNPs on beta-cell function was examined with linear regression<br />

assuming an additive model. To take into account the family relatedness, empirical standard<br />

errors were used (using the generalized estimating equations). For the monozygotic twins we<br />

computed the mean of the beta-cell measures and included only these mean measures in the<br />

analysis. The data from the non-identical twins were both used. The analyses of both first- and<br />

second-phase GSIS were adjusted for age, BMI, study centre, glucose tolerance status (NGT/<br />

IGT) and ISI. For the analysis of ISI and DI, ISI was removed from the covariates. All outcomes<br />

were log transformed prior to analysis. Due to the fact that NR3C1 polymorphisms have<br />

previously been shown to display gender-specific effects, male and females participants were<br />

analyzed separately [8-10]. All data are given as estimated mean (95%CI). After Bonferroni<br />

correction for multiple testing, results were regarded significant at a level of P


Figure 1. Schematic overview of the glucocorticoid receptor (GR) gene, NR3C1, polymorphisms and<br />

haplotypes. The location of each SNP in the NR3C1 gene is indicated by a black arrow. Minor allele<br />

frequencies of the SNPs are indicated below the SNP name. Haplotype alleles are indicated with black<br />

lines containing the nucleic acid for each SNP. Haplotype allele frequencies are displayed left of the<br />

haplotype allele and haplotype alleles are numbered in order of decreasing frequency.<br />

Associations with beta-cell function: The N363S variant was associated with reduced<br />

disposition index (P=1.06 10-4 ) and showed a trend towards reduced first-phase insulin<br />

secretion in women (Table 1). Also in women only, the ER22/23EK polymorphism was<br />

associated with reduced first-phase GSIS (P=0.011) and disposition index (P=0.003). No<br />

other associations were observed between NR3C1 SNPs and measures of beta-cell function,<br />

nor in men, nor in women. Similar results were obtained for the associations between NR3C1<br />

haplotypes and beta-cell function parameters (Supplementary Table 3). The results of the<br />

analyses were not different when NGT and IGT subjects were analyzed separately (data not<br />

shown).<br />

Associations with insulin sensitivity: None of the NR3C1 SNPs or haplotypes was significantly<br />

associated with insulin sensitivity (Table 1 and Supplementary Table 3 respectively).<br />

119<br />

6<br />

Chapter 6


Chapter 6 Glucocorticoid receptor SNPs & beta-cell function<br />

Table 1. Insulin response according to NR3C1 SNP in women and men.<br />

120<br />

Women Men<br />

DI<br />

(mmol/min.<br />

kg)<br />

ISI<br />

(mmol/min.<br />

kg.pmol/l)<br />

2 nd -phase<br />

GSIS (pmol/l)<br />

1 st -phase<br />

GSIS (pmol/l)<br />

n<br />

DI<br />

(mmol/min.<br />

kg)<br />

ISI<br />

(mmol/min.<br />

kg.pmol/l)<br />

Second-phase<br />

GSIS (pmol/l)<br />

First-phase<br />

GSIS (pmol/l)<br />

Genotype n<br />

BclI (rs41423247)<br />

CC 110 713 (656-766) 244 (225-264) 0.14 (0.12-0.15) 100 (91-109) 74 730 (643-828 262 (239-288) 0.15 (0.13-0.17) 107 (94-123)<br />

CG 98 773 (712-839) 248 (229-269) 0.14 (0.13-0.16) 109 (100-118) 82 665 (594-745) 236 (217-257) 0.16 (0.14-0.18) 103 (91-117)<br />

GG 30 837 (688-1017) 270 (233-312) 0.15 (0.12-0.19) 116(95-143) 13 710 (518-972) 258 (195-341) 0.12(0.08-0.17) 70 (70-119)<br />

β (SE) 0.035 (0.020) 0.018 (0.017) 0.018 (0.024) 0.035 (0.021) -0.021 (0.029) -0.023 (0.023) -0.013 (0.031) -0.027 (0.028)<br />

P 0.084 0.292 0.448 0.100 0.462 0.314 0.683 0.349<br />

9β<br />

(rs6198)<br />

AA 156 733 (685-784) 244 (229-260) 0.15 (0.13-0.16) 104 (98-112) 111 687 (622-759) 250 (231-271) 0.15 (0.13-0.17) 102 (92-114)<br />

AG 76 804 (723-893) 256 (233-281) 0.13 (0.11-0.15) 108 (96-121) 51 679 (577-798) 244 (216-275) 0.15 (0.13-0.18) 103 (87-122)<br />

GG 8 758 (503-1144) 313 (210-468) 0.11 (0.07-0.17) 91 (59-122) 7 930 (722- 240 (207-280) 0.17 (0.11-0.27) 142 (94-216)<br />

1197)<br />

β (SE) 0.028 (0.026) 0.033 (0.024) -0.051 (0.030) -0.003 (0.028) 0.022 (0.030) -0.010 (0.020) 0.011 (0.034) 0.030 (0.034)<br />

P 0.280 0.174 0.085 0.911 0.457 0.628 0.753 0.386<br />

TthIIII (rs10052957)<br />

CC 117 740 (686-798) 244 (226-264) 0.14(0.13-0.16) 104 (97-113) 71 698 (619-787) 217 (239-286) 0.15 (0.13-0.17) 102 (94-123)<br />

CT 99 787 (719-862) 260 (240-283) 0.14 (0.12-0.16) 109 (98-120) 77 686 (607-776) 243 (219-268) 0.15 (0.13-0.17) 103 (91-117)<br />

TT 23 715 (569-897) 228(193-271) 0.14 (0.10-0.16) 97 (78-121) 19 719 (553-934) 218 (184-259) 0.16 (0.12-0.22) 106 (70-119)<br />

β (SE) 0.005 (0.021) 0.001 (0.018) -0.018 (0.024) -0.004 (0.022) 0.002 (0.027) -0.037 (0.019) 0.011 (0.030) 0.007 (0.028)<br />

P 0.808 0.938 0.466 0.865 0.949 0.055 0.707 0.798


N363S (rs6195)<br />

AA 222 766 (722-812) 248 (235-262) 0.14 (0.13-0.15) 108 (101-115) 161 696 (637-760) 249 (232-267) 0.15 (0.13-0.17) 103 (94-114)<br />

AG 17 613 (515-731) 263 (225-307) 0.14 (0.09-0.14) 74 (62-88) 8 658 (553-783) 229 (191-274) 0.20 (0.14-0.29) 118 (91-154)<br />

β (SE) -0.096 (0.041) 0.026 (0.036) -0.111 (0.055) -0.165 (0.043) -0.024 (0.039) -0.037 (0.041) 0.130 (0.085) 0.058 (0.058)<br />

P 0.020 0.476 0.043 0.0001 0.534 0.375 0.125 0.327<br />

ER22/23EK (rs6189/rs6190)<br />

GG/GG 231 764 (722-808) 248 (235-262) 0.14 (0.13-0.15) 106 (100-113) 155 690 (632-754) 249 (232-266) 0.15 (0.14-0.17) 104 (94-114)<br />

AA/GG 9 518 (386-694) 294 (205-420) 0.10 (0.08-0.14) 68 (51-90) 14 751 (595-947) 240 (210-274) 0.14 (0.10-0.18) 106 (78-144)<br />

β (SE) -0.169 (0.066) 0.073 (0.080) -0.131 (0.064) -0.197 (0.065) 0.037 (0.052) -0.015 (0.030) -0.047 (0.068) 0.011 (0.069)<br />

P 0.011 0.360 0.040 0.003 0.485 0.618 0.491 0.874<br />

Data are estimated means (95% CI), the β (SE) is the β (SE) of the log transformed variable. Only one participant was homozygous for the N363S variant; the subject was<br />

added to the heterozygous group. For the ER22/23EK SNP, non-carriers are depicted as GG/GG. Heterozygotes are AA/GG. No homozygotes were identified in this cohort.<br />

All variables were log transformed before analysis. P values were computed for additive models using linear generalized estimating equations, which takes into account the<br />

family relatedness when computing the standard errors. First- and second-phase GSIS were adjusted for study center, family relatedness, glucose tolerance status, age, BMI<br />

and ISI. ISI and DI were adjusted for study center, family relatedness, glucose tolerance status, age and BMI. Significant findings are marked in bold. Abbreviations: GSIS:<br />

glucose-stimulated insulin secretion; ISI: insulin sensitivity index; DI: disposition index.<br />

121<br />

6<br />

Chapter 6


Chapter 6 Glucocorticoid receptor SNPs & beta-cell function<br />

DISCUSSIoN<br />

In four cohorts of NGT and IGT subjects, we found that the NR3C1 SNPs N363S and ER22/23EK<br />

were associated with reduced beta-cell function parameters in women. Both first-phase<br />

GSIS and disposition index, which denotes the adaptation of beta cells to prevailing insulin<br />

sensitivity, were influenced. As expected, the corresponding NR3C1 haplotypes containing<br />

these polymorphisms provided identical results. The N363S SNP enhances GC sensitivity by<br />

increasing gene transcription [14]. Indeed, in various studies a link was established between<br />

the N363S SNP and characteristics of a Cushingoid phenotype, including increased BMI and<br />

waist circumference, dyslipidemia and augmented fasting insulin levels, indicating reduced<br />

insulin sensitivity [10, 19]. The ER22/23EK SNP on the other hand, demonstrated reduced GR<br />

activation in vitro, and relative GC resistance in vivo [11, 16]. As such, in men, the ER22/23EK<br />

was associated with a beneficial metabolic phenotype, including increased muscle mass<br />

and strength, lower LDL cholesterol and insulin levels [11, 16]. On the other hand, female<br />

carriers of the ER22/23EK SNP were at increased risk to develop cardiovascular disease<br />

[28]. In another cohort, carriers of the ER22/23EK had higher HbA1c levels as compared<br />

to noncarriers [29], thus raising doubt on the hypothesis that this SNP may induce a more<br />

favorable metabolic profile, especially in women.<br />

More recently, impaired GSIS was shown to be another hallmark of GC-induced adverse<br />

metabolic effects both in vitro and in vivo in humans, where several measures of beta-cell<br />

function were impaired [2, 5-7]. Furthermore, mice overexpressing the GR specifically in<br />

beta-cells developed diabetes through beta-cell failure [20]. Our present data support the<br />

concept that GCs impair beta-cell function.<br />

Interestingly, the associations between SNPs in the NR3C1 gene and beta-cell function<br />

parameters were only observed in women, not in men. As outlined above, genderspecific<br />

effects of NR3C1 gene variants have been observed in various studies for various<br />

anthropometric and metabolic variables [8-10, 16, 17, 19]. Additionally, gender-related<br />

hormonal factors are known to affect beta-cell function [30]. As such, premenopausal women<br />

and women receiving estrogen replacement therapy have reduced prevalence of diabetes,<br />

which has been attributed to the beta-cell protective effects of estrogens [30]. Furthermore,<br />

the male sex hormone testosterone may also affect beta-cell function [31]. The effects of NR3C1<br />

polymorphisms on beta-cell function may therefore interact differently with sex hormones.<br />

An important limitation of the present study is the relatively small number of participants<br />

that were included, although this cohort is the largest to undergo a hyperglycemic clamp in<br />

the context of genetic analysis currently available in the literature. We cannot rule out the<br />

122


possibility to have missed subtle effects of other NR3C1 polymorphisms on beta-cell function<br />

variables. Another limitation is the fact that the SNPs did not tag the whole NR3C1 gene,<br />

therefore we cannot exclude that the associations found were caused by another untested<br />

SNP, although given the extensive literature on the SNPs function, this seems unlikely. We fully<br />

subscribe the need for replication of these data, although such replication is nontrivial because<br />

the hyperglycemic clamp methodology is demanding for both researchers and participants.<br />

In conclusion, this is the first report to show that the N363S and ER22/23EK NR3C1 gene<br />

variants are associated with reduced first-phase GSIS and DI in women, but not in men. This<br />

may point to a differential effect of genetically determined variation in GR activity in women<br />

as compared to men in the adaptation of (first-phase) insulin secretion to insulin sensitivity.<br />

Funding: This paper was written within the framework of project T1-106 of the Dutch Top<br />

Institute <strong>Pharma</strong>. The work in this study was additionally financially supported by the Dutch<br />

Diabetes Research Foundation grant 2006.00.060.<br />

123<br />

6<br />

Chapter 6


Chapter 6 Glucocorticoid receptor SNPs & beta-cell function<br />

REFERENCE LIST<br />

1. Schacke H, Docke WD, Asadullah K. Mechanisms involved in the side effects of glucocorticoids.<br />

<strong>Pharma</strong>col Ther. 2002;96(1):23-43.<br />

2. van Raalte DH, Ouwens DM, Diamant M. Novel insights into glucocorticoid-mediated diabetogenic<br />

effects: towards expansion of therapeutic options? Eur j Clin Invest. 2009;39(2):81-93.<br />

3. Gulliford MC, Charlton j, Latinovic R. Risk of diabetes associated with prescribed glucocorticoids in<br />

a large population. Diabetes Care. 2006;29(12):2728-9.<br />

4. McMahon M, Gerich j, Rizza R. Effects of glucocorticoids on carbohydrate metabolism. Diabetes<br />

Metab Rev. 1988;4(1):17-30.<br />

5. Linssen MM, van Raalte DH, Toonen Ej, Alkema W, van der Zon GC, Dokter WH, Diamant M, Guigas B,<br />

Ouwens DM. Prednisolone-induced beta cell dysfunction is associated with impaired endoplasmic<br />

reticulum homeostasis in INS-1E cells. Cell Signal. 2011;23(11):1708-15.<br />

6. van Raalte DH, Nofrate V, Bunck MC, van Iersel T, Elassaiss Schaap j, Nassander UK, Heine Rj, Mari<br />

A, Dokter WH, Diamant M. Acute and 2-week exposure to prednisolone impair different aspects of<br />

beta-cell function in healthy men. Eur j Endocrinol. 2010;162(4):729-35.<br />

7. van Raalte DH, van Genugten RE, Linssen MM, Ouwens DM, Diamant M. Glucagon-like peptide-1<br />

receptor agonist treatment prevents glucocorticoid-induced glucose intolerance and islet-cell<br />

dysfunction in humans. Diabetes Care. 2011;34(2):412-7.<br />

8. Derijk RH. Single nucleotide polymorphisms related to HPA axis reactivity. Neuroimmunomodulation.<br />

2009;16(5):340-52.<br />

9. van Rossum EF, Lamberts SW. Polymorphisms in the glucocorticoid receptor gene and their<br />

associations with metabolic parameters and body composition. Recent Prog Horm Res. 2004;59:333-<br />

57.<br />

10. Manenschijn L, van den Akker EL, Lamberts SW, van Rossum EF. Clinical features associated with<br />

glucocorticoid receptor polymorphisms. An overview. Ann N Y Acad Sci. 2009;1179:179-98.<br />

11. van Rossum EF, Koper jW, Huizenga NA, Uitterlinden AG, janssen jA, Brinkmann AO, Grobbee DE, de<br />

jong FH, van Duyn CM, Pols HA, Lamberts SW. A polymorphism in the glucocorticoid receptor gene,<br />

which decreases sensitivity to glucocorticoids in vivo, is associated with low insulin and cholesterol<br />

levels. Diabetes. 2002;51(10):3128-34.<br />

12. Derijk RH, Schaaf Mj, Turner G, Datson NA, Vreugdenhil E, Cidlowski j, de Kloet ER, Emery P,<br />

Sternberg EM, Detera-Wadleigh SD. A human glucocorticoid receptor gene variant that increases the<br />

stability of the glucocorticoid receptor beta-isoform mRNA is associated with rheumatoid arthritis.<br />

j Rheumatol. 2001;28(11):2383-8.<br />

13. van Rossum EF, Koper jW, van den Beld AW, Uitterlinden AG, Arp P, Ester W, janssen jA, Brinkmann<br />

AO, de Jong FH, Grobbee DE, Pols HA, Lamberts SW. Identification of the BclI polymorphism in the<br />

glucocorticoid receptor gene: association with sensitivity to glucocorticoids in vivo and body mass<br />

index. Clin Endocrinol (Oxf). 2003;59(5):585-92.<br />

124


14. Russcher H, Smit P, van den Akker EL, van Rossum EF, Brinkmann AO, de jong FH, Lamberts SW,<br />

Koper jW. Two polymorphisms in the glucocorticoid receptor gene directly affect glucocorticoidregulated<br />

gene expression. j Clin Endocrinol Metab. 2005;90(10):5804-10.<br />

15. van Rossum EF, Roks PH, de jong FH, Brinkmann AO, Pols HA, Koper jW, Lamberts SW. Characterization<br />

of a promoter polymorphism in the glucocorticoid receptor gene and its relationship to three other<br />

polymorphisms. Clin Endocrinol (Oxf). 2004;61(5):573-81.<br />

16. van Rossum EF, Voorhoeve PG, te Velde Sj, Koper jW, Delemarre-van de Waal HA, Kemper HC,<br />

Lamberts SW. The ER22/23EK polymorphism in the glucocorticoid receptor gene is associated<br />

with a beneficial body composition and muscle strength in young adults. J Clin Endocrinol Metab.<br />

2004;89(8):4004-9.<br />

17. Syed AA, Irving jA, Redfern CP, Hall AG, Unwin NC, White M, Bhopal RS, Weaver jU. Association of<br />

glucocorticoid receptor polymorphism A3669G in exon 9beta with reduced central adiposity in<br />

women. Obesity (Silver Spring). 2006;14(5):759-64.<br />

18. Rosmond R, Chagnon YC, Holm G, Chagnon M, Perusse L, Lindell K, Carlsson B, Bouchard C,<br />

Bjorntorp P. A glucocorticoid receptor gene marker is associated with abdominal obesity, leptin, and<br />

dysregulation of the hypothalamic-pituitary-adrenal axis. Obes Res. 2000;8(3):211-8.<br />

19. Roussel R, Reis AF, Dubois-Laforgue D, Bellanne-Chantelot C, Timsit j, Velho G. The N363S<br />

polymorphism in the glucocorticoid receptor gene is associated with overweight in subjects with<br />

type 2 diabetes mellitus. Clin Endocrinol (Oxf). 2003;59(2):237-41.<br />

20. Davani B, Portwood N, Bryzgalova G, Reimer MK, Heiden T, Ostenson CG, Okret S, Ahren B, Efendic<br />

S, Khan A. Aged transgenic mice with increased glucocorticoid sensitivity in pancreatic beta-cells<br />

develop diabetes. Diabetes. 2004;53 Suppl 1:S51-9.<br />

21. Fritsche A, Madaus A, Renn W, Tschritter O, Teigeler A, Weisser M, Maerker E, Machicao F, Haring<br />

H, Stumvoll M. The prevalent Gly1057Asp polymorphism in the insulin receptor substrate-2 gene is<br />

not associated with impaired insulin secretion. j Clin Endocrinol Metab. 2001;86(10):4822-5.<br />

22. Ruige jB, Dekker jM, Nijpels G, Popp-Snijders C, Stehouwer CD, Kostense Pj, Bouter LM, Heine Rj.<br />

Hyperproinsulinaemia in impaired glucose tolerance is associated with a delayed insulin response<br />

to glucose. Diabetologia. 1999;42(2):177-80.<br />

23. Simonis-Bik AM, Eekhoff EM, Diamant M, Boomsma DI, Heine Rj, Dekker jM, Willemsen G, van<br />

Leeuwen M, de Geus Ej. The heritability of HbA1c and fasting blood glucose in different measurement<br />

settings. Twin Res Hum Genet. 2008;11(6):597-602.<br />

24. van Haeften TW, Dubbeldam S, Zonderland ML, Erkelens DW. Insulin secretion in normal glucosetolerant<br />

relatives of type 2 diabetic subjects. Assessments using hyperglycemic glucose clamps and<br />

oral glucose tolerance tests. Diabetes Care. 1998;21(2):278-82.<br />

25. van Haeften TW, Pimenta W, Mitrakou A, Korytkowski M, jenssen T, Yki-jarvinen H, Gerich jE.<br />

Disturbances in beta-cell function in impaired fasting glycemia. Diabetes. 2002;51 Suppl 1:S265-70.<br />

125<br />

6<br />

Chapter 6


Chapter 6 Glucocorticoid receptor SNPs & beta-cell function<br />

26. Mitrakou A, Vuorinen-Markkola H, Raptis G, Toft I, Mokan M, Strumph P, Pimenta W, Veneman T,<br />

jenssen T, Bolli G, et al. Simultaneous assessment of insulin secretion and insulin sensitivity using a<br />

hyperglycemia clamp. j Clin Endocrinol Metab. 1992;75(2):379-82.<br />

27. Kahn SE, Prigeon RL, McCulloch DK, Boyko EJ, Bergman RN, Schwartz MW, Neifing JL, Ward WK,<br />

Beard JC, Palmer JP, et al. Quantification of the relationship between insulin sensitivity and betacell<br />

function in human subjects. Evidence for a hyperbolic function. Diabetes. 1993;42(11):1663-72.<br />

28. Koeijvoets KC, van Rossum EF, Dallinga-Thie GM, Steyerberg EW, Defesche jC, Kastelein jj,<br />

Lamberts SW, Sijbrands Ej. A functional polymorphism in the glucocorticoid receptor gene and its<br />

relation to cardiovascular disease risk in familial hypercholesterolemia. j Clin Endocrinol Metab.<br />

2006;91(10):4131-6.<br />

29. Kuningas M, Mooijaart SP, Slagboom PE, Westendorp RG, van Heemst D. Genetic variants in the<br />

glucocorticoid receptor gene (NR3C1) and cardiovascular disease risk. The Leiden 85-plus Study.<br />

Biogerontology. 2006;7(4):231-8.<br />

30. Liu S, Mauvais-jarvis F. Minireview: Estrogenic protection of beta-cell failure in metabolic diseases.<br />

Endocrinology. 2010;151(3):859-64.<br />

31. Morimoto S, Fernandez-Mejia C, Romero-Navarro G, Morales-Peza N, Diaz-Sanchez V. Testosterone<br />

effect on insulin content, messenger ribonucleic acid levels, promoter activity, and secretion in the<br />

rat. Endocrinology. 2001;142(4):1442-7.<br />

126


SUPPLEMENTAL FILES<br />

Supplementary Figure 1<br />

Pairwise Linkage Disequilibrium (LD) values between the SNP quantified using r 2 in the Haploview<br />

program are indicated. Black color indicates full linkage disequilibrium, lower shades of gray indicate<br />

lower LD and white color indicates no LD.<br />

127<br />

6<br />

Chapter 6


Chapter 6 Glucocorticoid receptor SNPs & beta-cell function<br />

Supplemental Table 1. Characteristics of the cohort.<br />

Hoorn*1 Utrecht*2,3 NTR*4 Germany*5<br />

128<br />

IGT NGT IGT NGT IGT NGT IGT<br />

n 137 64 12 116 7 81 32<br />

Sex (male/female) 64/73 15/49 4/8 58/58 0/7 33/48 14/18<br />

Age (years) 60.5±8.6 45.9±6.4 49.5±7.7 31.5±6.5 31.2±3.2 36.7±12.6 45.9±13.3<br />

BMI (kg m -2 ) 28.1±4.0 25.8±3.8 26.7±4.1 24.2±3.5 24.5±3.3 25.0±5.8 26.8±4.5<br />

FPG (mmol/l) 6.3±0.7 4.6±0.4 5.1±0.4 4.6±0.4 4.6±0.6 5.0±0.6 5.5±0.7<br />

FPI (pmol/l) 62 (46-91) 30 (24-42) 66 (42-78) 34 (27-51) 39 (29-60) 40 (30-60) 58 (39-95)<br />

587 (378-895) 885 (644-1217) 678 (461-909) 814 (589-1162) 795 (693-1210) 667 (522-1148) 524 (416-922)<br />

First-phase GSIS<br />

(pmol/l)<br />

255 (176-354) 260 (191-365) 251 (186-307) 218 (162-358) 217 (210-434) 229 (147-343) 196 (145-294)<br />

Second-phase GSIS<br />

(pmol/l)<br />

0.11 (0.07-0.17) 0.19 (0.13-0.29) 0.11 (0.08-0.26) 0.23 (0.15-0.32) 0.12 (0.11-0.18) 0.15 (0.09-0.22) 0.11 (0.06-0.15)<br />

ISI (mmol/min.<br />

kg.pmol/l)<br />

DI (mmol/min.kg) 65 (42-92) 172 (103-238) 72 (55-128) 180 (140-234) 138 (82-151) 109 (71-166) 62 (45-80)<br />

Data are mean ± SD, median (interquartile range) or n. *Original population from which the cohort originated [1-5]. FPG: fasting plasma glucose; FPI: fasting plasma<br />

insulin; GSIS: glucose-stimulated insulin secretion; ISI: insulin sensitivity index; DI: disposition index.<br />

1. Ruige jB, Dekker jM, Nijpels G, Popp-Snijders C, Stehouwer CD, Kostense Pj, et al. Hyperproinsulinaemia in impaired glucose tolerance is associated with a<br />

delayed insulin response to glucose. Diabetologia. 1999 Feb;42(2):177-80.<br />

2. van Haeften TW, Dubbeldam S, Zonderland ML, Erkelens DW. Insulin secretion in normal glucose-tolerant relatives of type 2 diabetic subjects. Assessments<br />

using hyperglycemic glucose clamps and oral glucose tolerance tests. Diabetes Care. 1998 Feb;21(2):278-82.<br />

3. van Haeften TW, Pimenta W, Mitrakou A, Korytkowski M, jenssen T, Yki-jarvinen H, et al. Disturbances in beta-cell function in impaired fasting glycemia.<br />

Diabetes. 2002 Feb;51 Suppl 1:S265-70.<br />

4. Simonis-Bik AM, Eekhoff EM, Diamant M, Boomsma DI, Heine Rj, Dekker jM, et al. The heritability of HbA1c and fasting blood glucose in different measurement<br />

settings. Twin Res Hum Genet. 2008 Dec;11(6):597-602.<br />

5. Fritsche A, Madaus A, Renn W, Tschritter O, Teigeler A, Weisser M, et al. The prevalent Gly1057Asp polymorphism in the insulin receptor substrate-2 gene is<br />

not associated with impaired insulin secretion. j Clin Endocrinol Metab. 2001 Oct;86(10):4822-5.


Supplemental Table 2. Major inclusion criteria of the study cohort.<br />

Cohort Reference Ethnicity Age Relatedness Glucometabolic State FDR T2DM<br />

Hoorn [1] Caucasian 45-74 Unrelated IGT No<br />

Utrecht [2] Caucasian NA Unrelated NGT Yes<br />

[3] Caucasian NA Unrelated NGT/IGT No<br />

NTR [4] Caucasian 20-50 Same sex twins and siblings NGT and IGT No<br />

Tubingen [5] Caucasian NA Unrelated NGT and IGT No<br />

Glucometabolic state was determined following an OGTT. FDR T2DM: first degree relative of patient with type 2 diabetes mellitus; IGT: impaired glucose tolerance;<br />

NA: not applicable; NTR: Netherlands Twin Registry; NGT: normal glucose tolerance.<br />

1. Ruige jB, Dekker jM, Nijpels G, Popp-Snijders C, Stehouwer CD, Kostense Pj, et al. Hyperproinsulinaemia in impaired glucose tolerance is associated with a<br />

delayed insulin response to glucose. Diabetologia. 1999 Feb;42(2):177-80.<br />

2. van Haeften TW, Dubbeldam S, Zonderland ML, Erkelens DW. Insulin secretion in normal glucose-tolerant relatives of type 2 diabetic subjects. Assessments<br />

using hyperglycemic glucose clamps and oral glucose tolerance tests. Diabetes Care. 1998 Feb;21(2):278-82.<br />

3. van Haeften TW, Pimenta W, Mitrakou A, Korytkowski M, jenssen T, Yki-jarvinen H, et al. Disturbances in beta-cell function in impaired fasting glycemia.<br />

Diabetes. 2002 Feb;51 Suppl 1:S265-70.<br />

4. Simonis-Bik AM, Eekhoff EM, Diamant M, Boomsma DI, Heine Rj, Dekker jM, et al. The heritability of HbA1c and fasting blood glucose in different measurement<br />

settings. Twin Res Hum Genet. 2008 Dec;11(6):597-602.<br />

5. Fritsche A, Madaus A, Renn W, Tschritter O, Teigeler A, Weisser M, et al. The prevalent Gly1057Asp polymorphism in the insulin receptor substrate-2 gene is<br />

not associated with impaired insulin secretion. j Clin Endocrinol Metab. 2001 Oct;86(10):4822-5.<br />

129<br />

6<br />

Chapter 6


Chapter 6 Glucocorticoid receptor SNPs & beta-cell function<br />

130<br />

Supplemental Table 3. Insulin response according to NR3C1 haplotype in women and men.<br />

Women Men<br />

DI<br />

(mmol/min.<br />

kg)<br />

ISI<br />

(mmol/min.<br />

kg.pmol/l)<br />

2 nd -phase<br />

GSIS (pmol/l)<br />

1 st -phase<br />

GSIS (pmol/l)<br />

n<br />

DI<br />

(mmol/min.<br />

kg)<br />

ISI<br />

(mmol/min.<br />

kg.pmol/l)<br />

Second-phase<br />

GSIS (pmol/l)<br />

First-phase<br />

GSIS (pmol/l)<br />

Haplotype n<br />

Haplotype 1<br />

0 80 802 (721-892) 269 (245-294) 0.13 (0.11-0.15) 105 (94-118) 44 651 (552-768) 227 (200-257) 0.15 (0.12-0.18) 98 (86-116)<br />

1 108 754 (693-821) 246 (227-267) 0.14 (0.13-0.16) 107 (98-116) 92 719 (641-807) 254 (233-276) 0.15 (0.14-0.17) 108 (96-123)<br />

2 52 694 (625-772) 229 (205-257) 0.15 (0.13-0.16) 102 (91-115) 33 677 (570-804) 260 (226-300) 0.14 (0.12-0.17) 99 (83-119)<br />

P 0.06 0.03 0.18 0.75 0.68 0.12 0.83 0.86<br />

Haplotype 2<br />

0 148 716 (665-771) 240 (224-256) 0.14 (0.13-0.15) 100 (93-108) 114 706 (636-783) 245 (225-267) 0.15 (0.14-0.17) 106 (95-119)<br />

1 81 811 (745-883) 263 (240-288) 0.14 (0.13-0.16) 112 (102-123) 53 660 (569-767) 245 (220-274) 0.15 (0.13-0.18) 101 (86-119)<br />

2 11 937 (687-1278) 292 (238-357) 0.14 (0.09-0.21) 122 (92-163) 2 923 (763-1115) 418 (362-482) 0.06 (0.05-0.07) 79 (59-106)<br />

P 0.02 0.03 0.74 0.04 0.65 0.54 0.71 0.45<br />

Haplotype 3<br />

0 163 730 (684-780) 247 (232-264) 0.14 (0.13-0.15) 104 (97-111) 126 698 (634-768) 250 (232-269) 0.15 (0.13-0.17) 104 (93-116)<br />

1 73 807 (720-904) 252 (229-278) 0.13 (0.12-0.15) 108 (95-122) 37 654 (537-797) 246 (212-284) 0.16 (0.13-0.19) 102 (83-125)<br />

2 4 889 (731-1081) 306 (183-512) 0.12 (0.06-0.21) 109 (72-165) 6 835 (689-1012) 232 (200-270) 0.16 (0.09-0.26) 117 (88-156)<br />

P 0.08 0.51 0.30 0.58 0.98 0.63 0.70 0.87<br />

Haplotype 4<br />

0 189 756 (711-804) 253 (238-268) 0.14 (0.13-0.15) 104 (97-111) 124 711 (645-784) 258 (241-278) 0.15 (0.13-0.17) 106 (95-118)<br />

1 47 781 (681-895) 241 (218-268) 0.15 (0.13-0.17) 113 (98-130) 40 633 (535-748) 215 (188-246) 0.16 (0.13-0.19) 99 (84-116)<br />

2 4 488 (312-762) 198 (131-300) 0.14 (0.09-0.20) 70 (45-109) 5 772 (541-1101) 257 (186-355) 0.13 (0.07-0.25) 98 (66-145)<br />

P 0.58 0.23 0.37 0.96 0.45 0.06 0.95 0.44


Haplotype 5<br />

0 223 767 (724-813) 248 (235-263) 0.14 (0.13-0.15) 108 (101-115) 161 696 (637-760) 249 (232-267) 0.15 (0.13-0.17) 103 (94-114)<br />

1 17 614 (515-760) 264 (226-308) 0.11 (0.09-0.14) 74 (62-88) 8 658 (553-783) 229 (191-274) 0.20 (0.14-0.29) 118 (91-154)<br />

P 0.02 0.47 0.04 0.0001 0.53 0.38 0.13 0.33<br />

Haplotype 6<br />

0 231 764 (722-808) 248 (235-262) 0.14 (0.13-0.15) 106 (100-113) 155 690 (632-754) 249 (232-266) 0.15 (0.14-0.17) 104 (94-114)<br />

1 9 518 (386-694) 294 (205-420) 0.10 (0.08-0.14) 68 (51-90) 14 751 (595-947) 240 (210-274) 0.14 (0.10-0.17) 106 (78-144)<br />

P 0.011 0.36 0.04 0.003 0.49 0.62 0.49 0.87<br />

For the haplotypes dummy variables indicating 0, 1 and 2 copies of each haplotype allele are shown. Data are estimated means (95% CI) unless otherwise<br />

indicated. All variables were log transformed before analysis. P values were computed for additive models using linear generalized estimating equations,<br />

which takes into account the family relatedness when computing the standard errors. First- and second-phase GSIS were adjusted for study center, family<br />

relatedness, glucose tolerance status, age, BMI and ISI. ISI and DI were adjusted for study center, family relatedness, glucose tolerance status, age and BMI.<br />

Significant findings are marked in bold. Abbreviations: GSIS: glucose-stimulated insulin secretion; ISI: insulin sensitivity index; DI: disposition index.<br />

131<br />

6<br />

Chapter 6


Chapter 7<br />

Glucagon-Like Peptide-1 Receptor Agonist<br />

Treatment Prevents Glucocorticoid-<br />

Induced Glucose Intolerance and Islet-Cell<br />

Dysfunction in Humans<br />

D.H. van Raalte*, R.E. van Genugten*, M.M.L. Linssen, D.M.<br />

Ouwens, M. Diamant<br />

*Both authors contributed equally<br />

Diabetes Care. 2011; 34(2):412-7


Chapter 7 GLP-1 prevents glucocorticoid-related metabolic effects<br />

ABSTRACT<br />

objective: Glucocorticoids (GCs) are regarded as diabetogenic since they impair insulin<br />

sensitivity and islet-cell function. In this study we assessed whether treatment with the<br />

glucagon-like peptide receptor agonist (GLP-1 RA) exenatide could prevent GC-induced<br />

glucose intolerance.<br />

Research design and methods: A randomized, placebo-controlled, double blind, crossover<br />

study in 8 healthy males (age: 23.5 (20.0-28.3) years; BMI: 26.4 (24.3-28.0) kg/m 2 ) was<br />

conducted. Participants received 3 therapeutic regimens for 2 consecutive days, i.e. (1) 80<br />

mg oral prednisolone once daily and intravenous (IV.) exenatide infusion (PRED+EXE),<br />

(2) 80 mg oral prednisolone once daily and IV. saline infusion (PRED+SAL), and (3) oral<br />

placebo-prednisolone once daily and IV. saline infusion (PLB+SAL). On day 1, glucose<br />

tolerance was assessed during a meal challenge test. On day 2, participants underwent a<br />

clamp procedure to measure insulin secretion and insulin sensitivity.<br />

Results: PRED+SAL treatment increased postprandial glucose levels (vs. PLB+SAL, P=<br />

0.012), which was prevented by concomitant exenatide (vs. PLB+SAL, P=NS). Exenatide<br />

reduced prednisolone-induced hyperglucagonemia during the meal challenge (P=0.018)<br />

and decreased gastric emptying (vs. PRED+SAL, P=0.028; vs. PLB+SAL, P=0.046). PRED+SAL<br />

decreased first-phase glucose-stimulated and arginine-stimulated C-peptide secretion (vs.<br />

PLB+SAL, P=0.017 and P=0.05 respectively), while PRED+EXE improved first-phase and<br />

second-phase glucose-stimulated, as well as arginine-stimulated C-peptide secretion (vs.<br />

PLB+SAL; P=0.017, 0.012 and 0.093 respectively).<br />

Conclusions: The GLP-1 RA exenatide prevented prednisolone-induced glucose intolerance<br />

and islet-cell dysfunction in healthy humans. Incretin-based therapies should be explored<br />

as a potential strategy to prevent steroid diabetes.<br />

Clinical Trial Registration Number: NCT00744224<br />

134


Glucocorticoids (GCs) are diabetogenic agents since they reduce insulin sensitivity [1], impair<br />

alpha-cell function [2], and, according to more recent findings, impair beta-cell function<br />

[3, 4]. As such, chronic use of GCs was associated with odds ratios between 1.4 and 2.3 to<br />

develop diabetes [5-7]. Loss of glycemic control during GC use is particularly due to impaired<br />

postprandial glucose metabolism, while fasting plasma glucose levels are usually only mildly<br />

elevated [4, 5]. Although the exact prevalence of steroid-related diabetes is unknown, the<br />

widespread use of GCs indicates that it may represent a major clinical problem worldwide.<br />

When initiating GC therapy in current clinical practice, preventive pharmacological measures<br />

are taken to prevent some of the GC-related side-effects, most notably osteoporosis and peptic<br />

ulcer disease [8, 9]. Interestingly, in spite of the above-mentioned highly-prevalent occurrence<br />

of steroid-diabetes, to date, no strategies are undertaken to prevent the adverse metabolic<br />

effects of GC treatment. Previous studies showed that metformin and the thiazolidinedione<br />

(TZD) pioglitazone were unable to mitigate the effects of GCs on glucose tolerance [10], while<br />

the TZD troglitazone prevented GC-induced hyperglycemia by enhancing GC clearance [10,<br />

11]. Due to liver toxicity, however, troglitazone is no longer available for treatment in humans.<br />

The gut hormone glucagon-like peptide (GLP)-1 and synthetic dipeptidyl-peptidase (DPP)-<br />

4 resistant GLP-1 receptor agonists (GLP-1 RA), such as exenatide, lower blood glucose by,<br />

glucose-dependently, enhancing insulin secretion and production and inhibiting glucagon<br />

secretion, and by slowing down gastric emptying [12]. One year of exenatide treatment<br />

was shown to improve clamp-measured beta-cell function in patients with type 2 diabetes<br />

mellitus (T2DM) [13]. Recently, the GLP-1 RA exendin-4 was shown to prevent GC-induced<br />

beta-cell apoptosis in vitro ([14]. In a single patient with Cushing’s disease, GLP-1 infusion<br />

was as effective in lowering blood glucose levels as compared to patients with ‘typical’ T2DM<br />

[15]. Finally, GLP-1 infusion effectively reduced stress hyperglycemia in patients undergoing<br />

coronary artery bypass graft (CABG) [16].<br />

Given these beneficial effects of GLP-1 RA treatment, and the pathophysiologic defects<br />

underlying GC-induced glucose intolerance and diabetes, we aimed to assess whether IV.<br />

infusion of the GLP-1 RA exenatide could prevent the acute adverse effects of prednisolone<br />

treatment on glucose metabolism, islet-cell function and insulin sensitivity in healthy<br />

normoglycemic individuals.<br />

135<br />

7<br />

Chapter 7


Chapter 7 GLP-1 prevents glucocorticoid-related metabolic effects<br />

RESEARCH DESIGN AND METHoDS<br />

Participants: Eight healthy males were recruited via local advertisements. Inclusion criteria<br />

included age = 18-35 years, body mass index (BMI) = 22.0-28.0 kg/m 2 , good physical<br />

health (determined by medical history, physical examination and screening blood tests)<br />

and normoglycemia as defined by fasting plasma glucose < 5.6 mmol/L and 2-hour glucose<br />

< 7.8 mmol/L following a 75g oral glucose tolerance test (OGTT), performed at screening<br />

visit. Exclusion criteria were the presence of any disease, use of any medication, first-degree<br />

relative with type 2 diabetes, smoking, shift work, a history of GC use and recent changes<br />

in weight or physical activity. The study was approved by an independent ethics committee<br />

and the study was conducted in accordance with the Declaration of Helsinki. All participants<br />

provided written informed consent before participation.<br />

Experimental design: The study was a randomized, placebo-controlled, double blind<br />

crossover study. Following assessment of eligibility, participants received for two consecutive<br />

days, in random order: (1) 80 mg oral prednisolone once daily and IV. exenatide infusion<br />

(PRED+EXE), (2) 80 mg oral prednisolone once daily and IV. saline infusion (PRED+SAL),<br />

and (3) oral placebo-prednisolone once daily and IV. saline infusion (PLB+SAL). On day 1,<br />

a standardized meal challenge was performed (Supplemental Figure 1A) and on day 2,<br />

participants underwent a combined clamp procedure, starting with a hyperinsulinemiceuglycemic<br />

clamp, followed by a hyperglycemic clamp with subsequent arginine stimulation<br />

(Supplemental Figure 1B) [13]. The three 2-day treatment blocks were separated by at least<br />

4 weeks. Participants were instructed to refrain from intense physical activity 2 days before<br />

each treatment block.<br />

The primary endpoint was the 4-hour glucose area under the curve (AUC G ) during the meal<br />

challenge test. From previous studies, we expected an increase in AUC G of 400 mmol/L•240min<br />

following prednisolone treatment [4]. A sample size of 8 would provide 80% power to detect<br />

a significant reduction of 50% by exenatide of prednisolone-induced augmentation of AUC . G<br />

Secondary endpoints included first-phase and second-phase C-peptide incremental area<br />

under the curve (iAUC ), and arginine-stimulated C-peptide secretion (ASI- iAUC ) during<br />

CP CP<br />

the hyperglycemic clamp.<br />

Standardized meal challenge: On day 1 of each treatment block, participants underwent a<br />

standardized meal challenge test following an overnight fast of minimally 10 hours. The meal<br />

contained 905 kcal (50 g fat, 75 g carbohydrates, 35 g protein) and 1g liquid acetaminophen<br />

to estimate gastric emptying rates. Samples for determination of glucose, insulin, C-peptide,<br />

136


glucagon, acetaminophen and exenatide were obtained at times -120, -60, -30, 0, 10, 20, 30,<br />

60, 90, 120, 150, 180 and 240 min, with the meal beginning immediately after the time 0<br />

sample and consumed within 15 min. Eighty mg oral prednisolone or placebo was ingested<br />

2 hours before meal consumption, and IV. exenatide or saline infusion started 60 min before<br />

the meal at an infusion rate of 40 ng/min for 30 min, and was decreased to 20 ng/min for the<br />

remainder of the test (Supplemental Figure 1A).<br />

Hyperinsulinemic-euglycemic clamp and hyperglycemic clamp: On day 2 of each block, a<br />

combined hyperinsulinemic-euglycemic and hyperglycemic clamp procedure was done. After<br />

an overnight fast, an indwelling cannula was inserted into an antecubital vein for infusion of<br />

glucose and insulin. To obtain arterialized venous blood samples, a retrograde cannula was<br />

inserted in a contralateral wrist vein and maintained in a heated box at 50°C. Insulin was<br />

infused at a rate of 40 mU/m2•min for 120 min, plasma glucose was kept at 5 mM by a variable<br />

infusion of 20% glucose. The hyperglycemic clamp was started 90 min after cessation of<br />

exogenous insulin infusion. Plasma glucose concentration was then raised to 10 mM by a body<br />

weight-adjusted intravenous bolus of 20% glucose and a variable 20% glucose infusion was<br />

adjusted to maintain the targeted glucose level. After 80 min hyperglycemia, an intravenous<br />

bolus of 5 g arginine (dissolved in 50 mL) was given over 45 seconds, and the glucose level was<br />

maintained at 10 mM for 30 min. Eighty mg oral prednisolone or placebo was administered 2<br />

hours before initiation of hyperinsulinemic-euglycemic clamp. IV. exenatide or saline infusion<br />

was started 60 min before the start of the hyperglycemic clamp at an infusion rate of 40 ng/<br />

min for 30 min, and was decreased to 20 ng/min for the rest of the hyperglycemic clamp<br />

(Supplemental Figure 1B). Note that exenatide was not infused during the hyperinsulinemiceuglycemic<br />

clamp, since, in a pilot study, exenatide strongly induced insulin secretion at a<br />

glucose level of 5 mM. The hyperinsulinemic-euglycemic clamp was performed in order to be<br />

able to calculate the disposition index (see below).<br />

Study medication: Prednisolone tablets were purchased from Pfizer AB (Sollentuna,<br />

Sweden) and placebo tablets were obtained from Xendo Drug Development (Groningen,<br />

The Netherlands). Tablets were capsulated in order to allow the treatment to be blinded [4].<br />

Commercially available exenatide was purchased (<strong>Pharma</strong>cy VU University Medical Center)<br />

and diluted in saline containing 1% human serum albumin, forming a colorless solution. IV.<br />

administration of exenatide was chosen in order to be able to gain steady-state exenatide<br />

levels within a short period of time. The plasma exenatide target level was 100 pg/ml, which<br />

demonstrated good efficacy and tolerability during previous infusion experiments [17].<br />

137<br />

7<br />

Chapter 7


Chapter 7 GLP-1 prevents glucocorticoid-related metabolic effects<br />

Analytical determinations: Blood glucose concentrations were measured using an YSI 2300<br />

STAT Plus analyzer (YSI, Yellow Springs, OH). Insulin and C-peptide levels were determined<br />

using an immunometric assay (Advia, Centaur, Siemens Medical Solutions Diagnostics, USA).<br />

Glucagon (Linco Research, St. Louis, USA) and acetaminophen (Abbott Laboratories, IL, USA)<br />

concentrations were determined by radioimmuno assay. Exenatide levels were determined<br />

by an immunoenzymetric assay as described previously (Amylin, San Diego, CA) (17).<br />

Body fat percentage was estimated by bioelectrical impedance analysis (BF-906, Maltron<br />

International, UK).<br />

Data analyses: Absolute area under the curves (AUCs) for glucose, insulin, C-peptide,<br />

glucagon and acetaminophen were calculated during the 4-hr meal challenge using the<br />

trapezoid method. The Matsuda whole-body insulin sensitivity index was calculated from<br />

the meal challenge. Whole-body insulin sensitivity as obtained from the hyperinsulinemiceuglycemic<br />

clamp was quantified by the M-value, calculated between min 90-120 during<br />

steady-state insulin concentrations. iAUC for the first-phase (min. 0-10), second-phase (min.<br />

CP<br />

10-80) and ASI-iAUC (min. 80-110) were calculated during the hyperglycemic clamp using<br />

CP<br />

the trapezoid method. The disposition index (DI) from the clamp tests was calculated by the<br />

C-peptide iAUC multiplied by the M-value.<br />

Statistical analyses: Data are presented as mean values ± standard error of the mean (SEM)<br />

or, in case of skewed distribution, as median (interquartile range). Between-block differences<br />

were tested non-parametrically with Friedman’s Test and, in case of a significant result,<br />

further analyzed using Wilcoxon Signed Ranks Test. All statistical analyses were run on SPSS<br />

(SPSS, Chicago, IL, USA). A P < 0.05 was considered statistically significant.<br />

RESULTS<br />

Subject characteristics: 8 healthy Caucasian males were included (median (interquartile<br />

range)): age = 23.5 (20.0-28.3) years; BMI = 25.8 (23.2-27.7) kg/m2 ; waist = 91 (82-95) cm;<br />

body fat = 21 (15-26)%; fasting plasma glucose = 5.0 (4.8-5.3) mmol/L; triglycerides = 1.1<br />

(0.7-1.5) mmol/L; systolic blood pressure = 119 (117-125) mmHg; diastolic blood pressure<br />

= 77 (72-82) mmHg.<br />

Standardized meal challenge: PRED+SAL treatment increased AUC G as compared to<br />

PLB+SAL (P=0.012), which was prevented by concomitant exenatide administration (Table<br />

1, Figure 1A). AUC for insulin (AUC ) was not decreased by PRED+SAL, although AUC for<br />

I<br />

138


C-peptide (AUC CP ) tended to be lower (P=0.07). PRED+EXE significantly decreased both AUC I<br />

and AUC CP . (Table 1, Figure 1B+C). PRED+SAL non-significantly increased glucagon secretion<br />

compared to PLB+SAL (P=0.09), which was mitigated by exenatide treatment (Figure 1D).<br />

Exenatide significantly decreased AUC for acetaminophen (AUC ) compared to both<br />

ACET<br />

placebo and prednisolone, compatible with its gastric emptying slowing effects. The Matsuda<br />

whole-body insulin sensitivity index increased during exenatide treatment both vs. placebo<br />

and prednisolone (Table 1).<br />

Table 1. Results from the standardized meal challenge.<br />

AUC glucose<br />

(mmol/L•240min)<br />

AUC insulin<br />

(nmol/L•240min)<br />

AUC c-peptide<br />

(nmol/L•240min)<br />

AUC glucagon<br />

(pmol/L•240min)<br />

AUC acetominophen<br />

(mg/L•240min)<br />

Matsuda index<br />

(No dimension)<br />

PLB+SAL 1199 (1043-1248)<br />

PRED+SAL 1335 (1254-1501)<br />

PRED+EXE 1247 (1156-1260)<br />

PLB+SAL 66.2 (35.2-81.0)<br />

PRED+SAL 52.5 (27.2-70.2)<br />

PRED+EXE 32.2 (22.8-40.1)<br />

PLB+SAL 354 (212-382)<br />

PRED+SAL 270 (191-328)<br />

PRED+EXE 203 (184-221)<br />

PLB+SAL 2845 (1941-2965)<br />

PRED+SAL 3085 (2859-3633)<br />

PRED+EXE 2232 (1850-3149)<br />

PLB+SAL 1272 (1002-1485)<br />

PRED+SAL 1449 (1243-1542)<br />

PRED+EXE 940 (801-1039)<br />

PLB+SAL 22.6 (13.0-39.4)<br />

PRED+SAL 20.4 (16.1-41.2)<br />

PRED+EXE 36.8 (26.9-50.0)<br />

P1 P2 P3<br />

0.012 NS 0.025<br />

NS 0.017 0.017<br />

0.069 0.017 0.017<br />

0.091 NS 0.018<br />

NS 0.028 0.046<br />

NS 0.025 0.012<br />

Abbreviations: AUC: area under the curve; EXE: exenatide; PLB: placebo; PRED: prednisolone; SAL: saline.<br />

P1 indicates PLB+SAL vs PRED+SAL. P2 indicates PLB+SAL vs PRED+EXE. P3 indicates PRED+SAL vs<br />

PRED+EXE.<br />

Combined clamp procedure:<br />

C-peptide secretion, hyperglycemic clamp: Prednisolone decreased first-phase iAUC and CP<br />

ASI-iAUC (vs. PLB+SAL; P=0.017 and P=0.05 respectively), but did not affect second-phase<br />

CP<br />

iAUC (Table 2). Exenatide restored prednisolone-induced reductions in first-phase iAUC CP CP<br />

and ASI-iAUC and significantly improved C-peptide secretion during the entire clamp, both<br />

CP<br />

compared to PRED+SAL and PLB+SAL (Table 2, Figure 2A). Insulin iAUC results were not<br />

different from the C-peptide iAUC results (Figure 2B).<br />

139<br />

7<br />

Chapter 7


Chapter 7 GLP-1 prevents glucocorticoid-related metabolic effects<br />

Table 2. Results from the hyperglycemic clamp<br />

1 st iAUC CP<br />

(nmol•min/L)<br />

2 nd iAUC CP<br />

(nmol•min/L)<br />

1 st +2 nd iAUC CP<br />

(nmol•min/L)<br />

ASI-iAUC CP<br />

(nmol•min/L)<br />

140<br />

PLB+SAL 6 (4-8)<br />

PRED+SAL 4 (2-6)<br />

PRED+EXE 10 (8-13)<br />

PLB+SAL 30 (17-48)<br />

PRED+SAL 26 (18-53)<br />

PRED+EXE 111 (63-117)<br />

PLB+SAL 83 (79-107)<br />

PRED+SAL 71 (41-100)<br />

PRED+EXE 201 (160-249)<br />

PLB+SAL 26 (24-34)<br />

PRED+SAL 18 (17-29)<br />

PRED+EXE 37 (28-43)<br />

P1 P2 P3<br />

0.017 0.017 0.012<br />

0.779 0.012 0.012<br />

0.208 0.012 0.012<br />

0.05 0.093 0.012<br />

Abbreviations: ASI-iAUC CP : arginine-stimulated incremental area under the curve for C-peptide; iAUC CP :<br />

incremental area under the curve for C-peptide. EXE: exenatide; PLB: placebo; PRED: prednisolone; SAL:<br />

saline. P1 indicates PLB+SAL vs PRED+SAL. P2 indicates PLB+SAL vs PRED+EXE. P3 indicates PRED+SAL<br />

vs PRED+EXE.<br />

Figure 1. The effect of prednisolone with or without concomitant exenatide infusion on plasma<br />

glucose (A), insulin (B), C-peptide (C) and glucagon (D) levels during the meal challenge.<br />

Prednisolone increased AUC , which was prevented by exenatide (A), despite lower insulin and C-peptide<br />

G<br />

levels (B+C). Exenatide infusion reduced postprandial glucagon levels as compared Prednisolone (D).<br />

Mean ± SEM are shown. Black solid line with closed squares: PLB+SAL; Grey intersected line with closed<br />

circles: PRED+SAL; Black dotted line with open circles: PRED+EXE.


Figure 2. The effect of prednisolone with or without concomitant exenatide infusion on C-peptide<br />

(A) and insulin (B) concentrations during the hyperglycemic clamp. PRED+SAL decreased first-phase<br />

(min 0-10) and arginine-stimulated C-peptide secretion (min 80-110), which were completely restored<br />

and improved by concomitant exenatide administration (Black solid line with closed squares: PLB+SAL;<br />

Grey intersected line with closed circles: PRED+SAL; Black dotted line with open circles: PRED+EXE).<br />

Panel C depicts prednisolone-induced changes in M-value during the euglycemic clamp. Prednisolone<br />

reduced combined first- and second-phase (min 0-80) disposition index (DI) (panel D) and argininestimulated<br />

(min 80-110) DI (panel E), which was restored and improved by exenatide (Box-and-Whisker<br />

plots (min-max) are shown).<br />

Insulin sensitivity, euglycemic clamp: Insulin levels reached steady-state during min 90-<br />

120 of the euglycemic clamp, averaging 431 ± 62 (PLB+SAL), 418 ± 73 (PRED+SAL) and<br />

422 ± 57 pM (PRED+EXE). Prednisolone acutely decreased the M-value obtained from the<br />

euglycemic clamp by 20% (P=0.018) (Figure 2C). Adjustment of the M-value by insulin levels<br />

during the steady-state part of the clamp (M/I) did not affect the results (data not shown).<br />

Note that the effects of exenatide on whole-body insulin sensitivity were not assessed during<br />

the euglycemic clamp, exenatide was administered during the hyperglycemic clamp only.<br />

Disposition index: PRED+SAL decreased the DI from T = 0-80 min of the hyperglycemic<br />

clamp (combined first- and second-phase; P=0.012) and the DI from T = 80-110 min of the<br />

hyperglycemic clamp (arginine stimulation; P=0.012). The prednisolone-induced decrease in<br />

DI was fully restored by concomitant exenatide infusion, and exenatide significantly improved<br />

the DI as compared to PLB+SAL from T = 0-80 min (Figure 2D+E).<br />

141<br />

7<br />

Chapter 7


Chapter 7 GLP-1 prevents glucocorticoid-related metabolic effects<br />

Exenatide plasma levels and adverse effects: Mean exenatide plasma levels equaled 65 ± 4<br />

pg/mL between T = 0 and T = 240 of the meal challenge (Supplemental Figure 2A) and 80 ±<br />

4 pg/mL between T = -30 and T = 110 of the hyperglycemic clamp (Supplemental Figure 2B).<br />

No adverse effects of either prednisolone or exenatide treatment were experienced by the<br />

participants during the meal or clamp procedure.<br />

DISCUSSIoN<br />

GCs are known to impair glucose metabolism by inducing insulin resistance and, more<br />

recently, beta-cell dysfunction [1, 4]. This study is the first to demonstrate that treatment<br />

with the GLP-1RA exenatide prevents prednisolone-induced glucose intolerance as assessed<br />

by a standardized meal challenge test. During the hyperglycemic clamp, exenatide infusion<br />

restored prednisolone-induced impairment of beta-cell function variables, and even<br />

significantly improved a number of these variables relative to the control situation. In contrast<br />

to the findings observed during the clamp procedure, exenatide treatment given during the<br />

meal challenge, improved glucose tolerance but resulted in decreased insulin plasma levels.<br />

This observation is in line with a previous study in healthy individuals in which subcutaneous<br />

exenatide treatment reduced postprandial glucose excursions despite significantly lower<br />

insulin levels [18]. The glucose-lowering effects of exenatide were attributed to decreased<br />

glucagon secretion and gastric emptying and, due to its glucose-dependent mode of action,<br />

exenatide did not further stimulate insulin secretion in the presence of normoglycemia<br />

[18]. In our study, we similarly found reduced postprandial glucagon secretion and gastric<br />

emptying following exenatide treatment. Studies employing stable isotope techniques have<br />

demonstrated that exenatide may also reduce hepatic glucose output and increase wholebody<br />

glucose disposal in the postprandial state, independent of its more established effects<br />

on islet hormone secretion and gastric emptying [19, 20]. In our study, exenatide improved<br />

whole-body insulin sensitivity during the meal challenge as estimated by the Matsuda index,<br />

however, reduced glucose appearance due to decreased gastric emptying seemed primarily<br />

responsible for improving glucose tolerance. We did not assess the effects of exenatide on<br />

whole-body insulin sensitivity during the hyperinsulinemic-euglycemic clamp for previous<br />

mentioned reasons.<br />

In this proof-of-principle study, both treatment regimens were administered for a short<br />

period of time: i.e. for two consecutive days per study block. Although the acute metabolic<br />

effects of both prednisolone and exenatide may to some extent differ from their effects<br />

following prolonged administration, it provides a good model to study the effects of each of<br />

142


oth drugs as well as their interaction. IV. exenatide infusion was able to prevent the acute<br />

adverse effects of prednisolone on glucose tolerance, and additional benefits from exenatide<br />

treatment may be expected when both compounds are administered for a more prolonged<br />

time period. Chronic GC use is associated with increased appetite, significant weight gain,<br />

increased visceral fat mass, altered secretion of adipocytokines and dyslipidemia [1], all of<br />

which contribute to the adverse effects of GCs on glucose metabolism. In clinical studies<br />

in T2DM patients, chronic exenatide treatment was shown to reduce appetite, resulting in<br />

substantial weight loss, decrease in truncal fat mass, and increased secretion of adiponectin<br />

[13, 21, 22]. Also, exenatide improved postprandial dyslipidemia [23]. The strong reduction<br />

of postprandial glucose levels by exenatide, rather than a pronounced effect on fasting plasma<br />

glucose [13], matches the profile of GC-induced hyperglycemia, which is predominantly<br />

present during the day [5].<br />

During the hyperglycemic clamp experiments, pharmacological concentrations of exenatide<br />

were able to restore prednisolone-induced changes in beta-cell function, including firstphase<br />

and ASI C-peptide secretion and DI calculated for the entire hyperglycemic clamp. GC<br />

exposure was demonstrated to impair various pathways in the beta cell in vitro. These include<br />

both steps in the uptake and metabolism of glucose, but GCs also affected distal pathways in<br />

the insulin exocytosis process, resulting in impaired insulin secretion in response to different<br />

secretagogues [1, 3]. Since exenatide was able to restore insulin secretion, one may speculate<br />

that GCs do not block the pathways mediating GLP-1 action on beta cells. However, it was very<br />

recently reported that a two-week treatment with oral prednisolone reduced the insulinotropic<br />

effects of endogenous GLP-1 and glucose-dependent insulinotropic polypeptide (GIP) [24].<br />

A limitation of our study is that we treated healthy subjects with GCs, while GCs are<br />

prescribed to treat acute and chronic inflammatory, as well as autoimmune diseases. Chronic<br />

inflammation is also associated with whole-body insulin resistance and beta-cell dysfunction,<br />

as was recently reported in patients with rheumatoid arthritis [25]. Therefore, the complex<br />

interrelationship between inflammation, GC and GLP-1RA treatment needs do be studied<br />

prospectively in relevant patient populations.<br />

The plasma levels of exenatide reached with our infusion protocol were lower than<br />

those usually obtained following subcutaneous injection of exenatide 10 mg BID, i.e.<br />

the recommended dose for the current treatment in T2DM. Although good efficacy was<br />

demonstrated by current plasma exenatide levels, the full potential of GLP-1 RA treatment<br />

to prevent prednisolone-induced glucose intolerance may be fully unveiled in clinical studies<br />

administering exenatide at the usual dose.<br />

143<br />

7<br />

Chapter 7


Chapter 7 GLP-1 prevents glucocorticoid-related metabolic effects<br />

Taken together, this study provides evidence that the GLP-1 RA exenatide may prevent<br />

prednisolone-induced glucose intolerance and restore islet-cell functional balance. Longterm<br />

studies in relevant populations should explore the potential of GLP-1 RA treatment as a<br />

novel strategy to prevent steroid diabetes.<br />

Acknowledgments and Funding: The authors express their gratitude to Mark Fineman<br />

(Amylin <strong>Pharma</strong>ceuticals) for the determination of exenatide plasma levels. Drs. Daniël<br />

van Raalte and Drs. Margot Linssen are supported by the Dutch Top Institute <strong>Pharma</strong> (<strong>TI</strong>P)<br />

grant T1-106. Drs. Renate van Genugten is supported by the EFSD/MSD Clinical Research<br />

Programme 2008. Dr. D. Margriet Ouwens is supported by the Ministry of Innovation, Science,<br />

Research and Technology of the German state of North-Rhine Westphalia, and the EU European<br />

Cooperation in the field of Scientific and Technical Research (COST) Action BM0602.<br />

144


REFERENCES<br />

1. van Raalte DH, Ouwens DM, Diamant M. Novel insights into glucocorticoid-mediated diabetogenic<br />

effects: towards expansion of therapeutic options? Eur j Clin Invest. 2009;39(2):81-93.<br />

2. Wise JK, Hendler R, Felig P. Influence of glucocorticoids on glucagon secretion and plasma amino<br />

acid concentrations in man. j Clin Invest. 1973;52(11):2774-82.<br />

3. Lambillotte C, Gilon P, Henquin jC. Direct glucocorticoid inhibition of insulin secretion. An in vitro<br />

study of dexamethasone effects in mouse islets. j Clin Invest. 1997;99(3):414-23.<br />

4. van Raalte DH, Nofrate V, Bunck MC, van Iersel T, Elassaiss Schaap j, Nassander UK, Heine Rj, Mari<br />

A, Dokter WH, Diamant M. Acute and 2-week exposure to prednisolone impair different aspects of<br />

beta-cell function in healthy men. Eur j Endocrinol. 2010;162(4):729-35.<br />

5. Clore jN, Thurby-Hay L. Glucocorticoid-induced hyperglycemia. Endocr Pract. 2009;15(5):469-74.<br />

6. Gulliford MC, Charlton j, Latinovic R. Risk of diabetes associated with prescribed glucocorticoids in<br />

a large population. Diabetes Care. 2006;29(12):2728-9.<br />

7. Gurwitz jH, Bohn RL, Glynn Rj, Monane M, Mogun H, Avorn j. Glucocorticoids and the risk for<br />

initiation of hypoglycemic therapy. Arch Intern Med. 1994;154(1):97-101.<br />

8. Compston j. Management of glucocorticoid-induced osteoporosis. Nat Rev Rheumatol. 2010;6(2):82-<br />

8.<br />

9. Trikudanathan S, McMahon GT. Optimum management of glucocorticoid-treated patients. Nat Clin<br />

Pract Endocrinol Metab. 2008;4(5):262-71.<br />

10. Morita H, Oki Y, Ito T, Ohishi H, Suzuki S, Nakamura H. Administration of troglitazone, but not<br />

pioglitazone, reduces insulin resistance caused by short-term dexamethasone (DXM) treatment by<br />

accelerating the metabolism of DXM. Diabetes Care. 2001;24(4):788-9.<br />

11. Willi SM, Kennedy A, Wallace P, Ganaway E, Rogers NL, Garvey WT. Troglitazone antagonizes<br />

metabolic effects of glucocorticoids in humans: effects on glucose tolerance, insulin sensitivity,<br />

suppression of free fatty acids, and leptin. Diabetes. 2002;51(10):2895-902.<br />

12. Baggio LL, Drucker Dj. Biology of incretins: GLP-1 and GIP. Gastroenterology. 2007;132(6):2131-57.<br />

13. Bunck MC, Diamant M, Corner A, Eliasson B, Malloy jL, Shaginian RM, Deng W, Kendall DM,<br />

Taskinen MR, Smith U, Yki-jarvinen H, Heine Rj. One-year treatment with exenatide improves<br />

beta-cell function, compared with insulin glargine, in metformin-treated type 2 diabetic patients: a<br />

randomized, controlled trial. Diabetes Care. 2009;32(5):762-8.<br />

14. Ranta F, Avram D, Berchtold S, Dufer M, Drews G, Lang F, Ullrich S. Dexamethasone induces cell death<br />

in insulin-secreting cells, an effect reversed by exendin-4. Diabetes. 2006;55(5):1380-90.<br />

15. Ritzel RA, Kleine N, Holst jj, Willms B, Schmiegel W, Nauck MA. Preserved GLP-1 effects in a diabetic<br />

patient with Cushing’s disease. Exp Clin Endocrinol Diabetes. 2007;115(2):146-50.<br />

145<br />

7<br />

Chapter 7


Chapter 7 GLP-1 prevents glucocorticoid-related metabolic effects<br />

16. Sokos GG, Bolukoglu H, German j, Hentosz T, Magovern Gj, jr., Maher TD, Dean DA, Bailey SH, Marrone<br />

G, Benckart DH, Elahi D, Shannon RP. Effect of glucagon-like peptide-1 (GLP-1) on glycemic control<br />

and left ventricular function in patients undergoing coronary artery bypass grafting. Am j Cardiol.<br />

2007;100(5):824-9.<br />

17. Degn KB, Brock B, juhl CB, Djurhuus CB, Grubert j, Kim D, Han j, Taylor K, Fineman M, Schmitz<br />

O. Effect of intravenous infusion of exenatide (synthetic exendin-4) on glucose-dependent insulin<br />

secretion and counterregulation during hypoglycemia. Diabetes. 2004;53(9):2397-403.<br />

18. Kolterman OG, Buse jB, Fineman MS, Gaines E, Heintz S, Bicsak TA, Taylor K, Kim D, Aisporna M,<br />

Wang Y, Baron AD. Synthetic exendin-4 (exenatide) significantly reduces postprandial and fasting<br />

plasma glucose in subjects with type 2 diabetes. j Clin Endocrinol Metab. 2003;88(7):3082-9.<br />

19. Cervera A, Wajcberg E, Sriwijitkamol A, Fernandez M, Zuo P, Triplitt C, Musi N, DeFronzo RA,<br />

Cersosimo E. Mechanism of action of exenatide to reduce postprandial hyperglycemia in type 2<br />

diabetes. Am j Physiol Endocrinol Metab. 2008;294(5):E846-52.<br />

20. Zheng D, Ionut V, Mooradian V, Stefanovski D, Bergman RN. Exenatide sensitizes insulin-mediated<br />

whole-body glucose disposal and promotes uptake of exogenous glucose by the liver. Diabetes.<br />

2009;58(2):352-9.<br />

21. Bunck MC, Diamant M, Eliasson B, Corner A, Shaginian RM, Heine Rj, Taskinen MR, Yki-jarvinen H,<br />

Smith U. Exenatide affects circulating cardiovascular risk biomarkers independently of changes in<br />

body composition. Diabetes Care. 2010;33(8):1734-7.<br />

22. Klonoff DC, Buse jB, Nielsen LL, Guan X, Bowlus CL, Holcombe jH, Wintle ME, Maggs DG. Exenatide<br />

effects on diabetes, obesity, cardiovascular risk factors and hepatic biomarkers in patients with type<br />

2 diabetes treated for at least 3 years. Curr Med Res Opin. 2008;24(1):275-86.<br />

23. Bunck MC, Corner A, Eliasson B, Heine Rj, Shaginian RM, Wu Y, Yan P, Smith U, Yki-jarvinen H, Diamant<br />

M, Taskinen MR. One-year treatment with exenatide vs. insulin glargine: effects on postprandial<br />

glycemia, lipid profiles, and oxidative stress. Atherosclerosis. 2010;212(1):223-9.<br />

24. Hansen KB, Vilsboll T, Bagger jI, Holst jj, Knop FK. Reduced glucose tolerance and insulin resistance<br />

induced by steroid treatment, relative physical inactivity, and high-calorie diet impairs the incretin<br />

effect in healthy subjects. j Clin Endocrinol Metab. 2010;95(7):3309-17.<br />

25. Dessein PH, joffe BI. Insulin resistance and impaired beta cell function in rheumatoid arthritis.<br />

Arthritis Rheum. 2006;54(9):2765-75.<br />

146


SUPPLEMENTAL FIGURES<br />

Supplemental Figure 1. Overview of the meal challenge test (A) and the combined hyperinsulinemiceuglycemic<br />

and hyperglycemic clamp procedure (B). Note that exenatide was infused during the<br />

hyperglycemic clamp, but not during the euglycemic clamp. Abbreviations: EXE: exenatide; PRED:<br />

prednisolone.<br />

Supplemental Figure 2. Exenatide plasma levels equaled 65 ± 4 pg/mL between T = 0 and T = 240 of the<br />

meal challenge (A) and 80 ± 4 pg/mL between T = -30 and T = 110 of the hyperglycemic clamp (B). Mean<br />

± SEM are depicted.<br />

147<br />

7<br />

Chapter 7


PART<br />

Mechanisms of glucocorticoid-induced insulin<br />

resistance<br />

III


Chapter 8<br />

Low-dose Glucocorticoid Treatment<br />

Affects Multiple Aspects of Intermediary<br />

Metabolism in Healthy Humans: a<br />

Randomized Controlled Trial<br />

D.H. van Raalte*, M. Brands*, N.J. van der Zijl, M.H. Muskiet, P.<br />

J.W. Pouwels , M.T. Ackermans, H.P. Sauerwein, M.J. Serlie and<br />

M.Diamant<br />

*Both authors contributed equally<br />

Diabetologia. 2011; 54(8):2102-12


Chapter 8 Glucocorticoids affect intermediary metabolism<br />

ABSTRACT<br />

objectives: To assess whether low-dose glucocorticoid (GC) treatment induces adverse<br />

metabolic effects, as is evident for high GC doses.<br />

Methods: In a randomized, placebo-controlled, double blind, dose-response intervention<br />

study, 32 healthy males (age: 22±3 years; BMI 22.4±1.7 kg/m2 ) were allocated to<br />

prednisolone 7.5 mg once daily (n=12), prednisolone 30 mg once daily (n=12), or placebo<br />

(n=8) for two weeks using block randomization. Mean outcomes measures were glucose-,<br />

lipid- and protein metabolism, as measured by stable isotopes, before and at 2 weeks of<br />

treatment, in the fasted state and during a two-step hyperinsulinemic clamp.<br />

Results: Prednisolone, as compared to placebo, dose-dependently and significantly<br />

increased fasting plasma glucose levels, whereas only prednisolone 30 mg increased<br />

fasting insulin levels (29±15 pmol/l). Prednisolone 7.5 mg and prednisolone 30 mg<br />

decreased the ability of insulin to suppress endogenous glucose production (by 17±6%<br />

and 46±7%, respectively, vs. placebo). Peripheral glucose uptake was not reduced by<br />

prednisolone 7.5 mg, but was decreased by prednisolone 30 mg by 34±6% (P


Glucocorticoids (GCs) are the cornerstone in the treatment of numerous chronic autoimmune<br />

and inflammatory diseases, including rheumatoid arthritis [1], polymyalgia rheumatica<br />

[2] and giant cell arteritis [3], systemic lupus erythematosus [4] and inflammatory bowel<br />

diseases [5]. GC treatment is accompanied by significant metabolic adverse effects, including<br />

insulin resistance, glucose intolerance and diabetes, visceral adiposity, dyslipidemia and<br />

skeletal muscle atrophy [6]. The development of these adverse metabolic effects is dependent<br />

on the administered dose [7], and has mainly been studied with high doses. As such, 36<br />

mg prednisolone equivalent daily impaired glucose tolerance by reducing hepatic [8] and<br />

peripheral insulin sensitivity [9]. Furthermore, 30 mg prednisolone daily stimulated adipose<br />

tissue, but not whole-body, lipolysis [10], and induced dyslipidemia [11]. In addition, 60 mg<br />

prednisolone daily was shown to stimulate breakdown of skeletal muscle tissue [12]. Finally,<br />

chronic exposure to markedly increased endogenous GC levels, as shown in patients with<br />

Cushing’s syndrome, unfavorably affected body fat distribution, by promoting visceral fat<br />

accumulation and hepatic steatosis [13].<br />

However, most patients treated with GCs for prolonged time periods, receive, following<br />

an induction dose, GC dosages in the lower range: ≤ 7.5 mg prednisolone equivalent daily.<br />

Although the efficacy of these doses on disease activity in various conditions is wellestablished<br />

[1-4;7], the extent to which these low dosages induce adverse metabolic effects<br />

is unclear. In large retrospective case-control studies, it was demonstrated that low-dose GC<br />

treatment increased the odds for the initiation of blood-glucose lowering therapy by 63%<br />

[14] and increased the risk to develop diabetes [15]. In contrast, in clinical trials with chronic<br />

use of low-dose GC treatment, incident diabetes is reported less frequently [16]. The main<br />

limitation of these data is that no distinction can be made between disease-induced and agerelated<br />

abnormalities on the one hand, and the metabolic abnormalities induced by low-dose<br />

prednisolone treatment on the other. To date, no studies have assessed the effects of low-dose<br />

GC treatment on the 3 most important pathways of intermediary metabolism. In addition,<br />

previous studies addressing the metabolic adverse effects of GC treatment have only studied<br />

the (hyper)acute effects and typically administered a single dose.<br />

Thus, in the present study, we studied the effects of a two-week, low-dose prednisolone<br />

treatment (7.5 mg daily) on glucose-, lipid-, and protein metabolism using stable isotopes<br />

in healthy males, and included a typical induction dosage of prednisolone (i.e. 30 mg daily),<br />

which is known to impair intermediary metabolism, and a matching placebo treatment.<br />

Through combined infusion of tracers for glucose and protein metabolism, as well as lipolysis,<br />

we assessed differences in the sensitivity of metabolic fluxes to the dose-dependent effects of<br />

GC treatment.<br />

153<br />

8<br />

Chapter 8


Chapter 8 Glucocorticoids affect intermediary metabolism<br />

METHoDS<br />

Participants: Thirty-two healthy white males were recruited via local advertisements. All<br />

participants were in good health as confirmed by medical history, physical examination,<br />

screening blood tests and a 75-g 2-hr oral glucose tolerance test (OGTT), performed at<br />

screening visit. Inclusion criteria included age: 18-35 years, body mass index (BMI): 20-25 kg/<br />

m2 , and normoglycemia as defined by fasting plasma glucose (FPG) < 5.6 mmol/l and 2-hour<br />

glucose < 7.8 mmol/l during OGTT. Exclusion criteria were the presence of any disease, use of<br />

any medication, first-degree relative with type 2 diabetes, smoking, shift work, a history of GC<br />

use, excessive sport activities (i.e. more often than two times/week) and changes in weight in<br />

the 3 months prior to study participation. The study was approved by an independent ethics<br />

committee and the study was conducted in accordance with the Declaration of Helsinki. All<br />

participants provided written informed consent before participation.<br />

Study design: The study was a randomized, double blind, placebo-controlled, doseresponse<br />

intervention study. Following assessment of eligibility and baseline measurements,<br />

participants were randomized to receive either prednisolone 7.5 mg once daily (n=12), or<br />

prednisolone 30 mg once daily (n=12), or placebo (n=8) treatment for a period of 14 days<br />

using block randomization, as carried out by the department of experimental pharmacology of<br />

the VU University Medical Center. On day -2 and day 13 of treatment, body composition, body<br />

fat distribution and liver fat content were quantified. On day -1 and on day 14 of treatment,<br />

glucose kinetics, lipolysis and proteolysis were measured in the basal state and during a twostep<br />

hyperinsulinemic-euglycemic clamp using stable isotopes (Supplementary Figure 1a,b).<br />

All measurements were conducted following an 12-h overnight fast with the subjects in the<br />

semi-supine position. Subjects refrained from drinking alcohol for a period of 24h before the<br />

study days and did not perform strenuous exercise for a period of 48h before the study days.<br />

During all visits, including a follow-up visit at day 7 of treatment, safety and tolerability were<br />

assessed. A patient flow diagram is shown in Supplementary Figure 2.<br />

Experimental protocol – body composition/body fat distribution: Body composition<br />

was measured by Dual Energy X-ray Absorptiometry (DEXA) scans (Delphi A, Hologic,<br />

Waltham, MA). Magnetic resonance imaging (MRI), for determination of visceral (VAT) and<br />

subcutaneous (SAT) adipose tissue area at the level of L3-L4, and proton-magnetic resonance<br />

spectroscopy ( 1H-MRS), to quantify liver fat content, were performed using a 1.5 T MRI<br />

scanner (Sonata; Siemens, Erlangen, Germany), as described previously [17;18]. All magnetic<br />

resonance examinations (DVR) and quantification of abdominal fat compartments (MHM)<br />

were done by a single experienced investigator.<br />

154


Experimental protocol – clamp: After an overnight fast of 12 hours, subjects were admitted<br />

at the clinical research unit at 0730 AM. An indwelling cannula was inserted into an antecubital<br />

vein for infusion of stable-isotope tracers, glucose and insulin. To obtain arterialized venous<br />

blood samples, a retrograde cannula was inserted in a contralateral wrist vein and maintained<br />

in a thermoregulated box at 50°C. 0.9% NaCl was infused to keep the sampling line patent.<br />

[6,6-2H ]Glucose, [1,1,2,3,3,- 2 2H ]glycerol and L-[1- 5 13C]valine were used as tracers (> 99%<br />

enriched; Cambridge Isotopes, Andover, MA) to study glucose kinetics, lipolysis and valine<br />

turnover respectively. At T = 0 h (0800 AM), blood samples were drawn for determination of<br />

background enrichments. Then, a primed, continuous infusion of isotopes was started: [6,6-<br />

2H2 ]glucose (prime: 11 mmol/kg; continuous: 0.11 mmol/kg.min), [1,1,2,3,3,-2H ]glycerol<br />

5<br />

(prime: 1.6 mmol/kg; continuous: 0.11 mmol/kg.min), and L-[1-13C]valine (prime: 13.7<br />

mmol/kg; continuous: 0.153 mmol,kg.min) and continued until the end of the clamp. After<br />

a 2-h equilibrium period (14 h of fasting), 3 blood samples were drawn for determination<br />

of basal glucose concentrations, isotope enrichments, glucoregulatory hormones and<br />

nonesterified fatty acids (NEFA). Thereafter, a 2-step hyperinsulinemic-euglycemic clamp<br />

was started: step 1 included an infusion of insulin at a rate of 20 mU/m2 .min (Actrapid 100<br />

IU/mL; Novo Nordisk Farma BV, Alphen aan den Rijn, Netherlands) to assess hepatic insulin<br />

sensitivity. Glucose 20% was started to maintain a plasma glucose concentration of 5 mmol/l.<br />

[6,6-2H ]Glucose was added to the glucose solution to achieve glucose enrichments of 1% in<br />

2<br />

order to approximate the values for enrichment reached in plasma and thereby minimizing<br />

changes in isotopic enrichment due to changes in the infusion rate of exogenous glucose.<br />

Plasma glucose concentrations were measured every 5 min at bedside. After 2-h of insulin<br />

infusion, 5 blood samples were drawn at 5min intervals for the measurement of glucose<br />

concentrations and isotopic enrichments. Another blood sample was drawn for measurement<br />

of glucoregulatory hormones and NEFA. Hereafter, insulin infusion was increased to a rate of<br />

60 mU/m2 .min (step 2) to assess peripheral insulin sensitivity. After 2 h of insulin infusion,<br />

blood sampling for glucose, isotope enrichments, glucoregulatory hormones and NEFA was<br />

repeated (Supplementary Figure 1b).<br />

Indirect calorimetry: Oxygen consumption (VO 2 ) and carbon dioxide production (VCO 2 )<br />

were measured continuously during the final 20 min of both the basal state and during step<br />

2 of the hyperinsulinemic-euglycemic clamp by indirect calorimetry using a ventilated hood<br />

system (Vmax model 2900; Sensormedics, Anaheim, CA). The VO and VCO measurements<br />

2 2<br />

during the last 10 minutes were used for further calculations.<br />

Study medication: Prednisolone tablets were purchased from Pfizer AB (Sollentuna, Sweden)<br />

and matching placebo tablets were obtained from Xendo Drug Development (Groningen, The<br />

155<br />

8<br />

Chapter 8


Chapter 8 Glucocorticoids affect intermediary metabolism<br />

Netherlands). The tablets were encapsulated in order to allow the treatment to be blinded,<br />

as described previously [19]. Study medication was taken at 08.00 AM during the two-week<br />

treatment except for day 13 and day 14, when it was ingested at 06.00 AM. Patients kept a<br />

diary in which the exact time of medication intake during the study was registered.<br />

Glucose, lipid, and valine measurements: Plasma glucose concentrations were measured<br />

with the glucose oxidase method using a Biosen C-line plus glucose analyzer (EKF Diagnostics,<br />

Barleben/Magedeburg, Germany). Plasma NEFA concentrations were measured with an<br />

enzymatic colorimetric method (NEFA-C test kit; Wako Chemicals GmbH, Neuss, Germany)<br />

with an intra-assay variation of 1%, inter-assay variation of 4-15% and detection limit of<br />

0.02 mmol/l. [6,6-2H ]Glucose, [1,1,2,3,3,- 2 2H ]glycerol and L-[1- 5 13C]valine enrichment were<br />

measured with gas chromatography-mass spectroymetry (GC-MS) as described previously<br />

[20-22]. Briefly, temperature programmed GC with splitinjection (Model 6890 Agilent<br />

Technologies, Palo Alto, USA) was coupled to a mass selctive detector (Model 5973 Agilent<br />

technologies, Palo Alto USA) in the electron impact (EI) ionization mode for glucose and<br />

valine analysis and in the positive chemical ionisation mode for glycerol. Exact GC and MS<br />

parameters are given in ref 20-22.<br />

Glucoregulatory hormones: Insulin was determined on an Immulite 2000 system<br />

(Diagnostic Products Corporation, Los Angeles, CA) with a chemiluminescent immunometric<br />

assay, intra-assay variation of 3-6%, an inter-assay variation of 4-6%, and a detection limit of<br />

15 pmol/l. Glucagon was determined with the Linco 125I radioimmunoassay (St Charles, MO)<br />

with an intra-assay variation of 9-10%, an inter-assay variation of 5-7%, and a detection limit<br />

of 15 ng/l.<br />

Calculations: Endogenous glucose production (EGP) and the peripheral uptake of glucose<br />

(Rate of disappearance, Rd) were calculated using modified versions of the Steele equations<br />

for the non-steady state and were expressed as mmol/kg.min as described previously<br />

[23;24]. Lipolysis (glycerol turnover) and proteolysis (valine turnover) were computed using<br />

formulas for steady state kinetics adapted for stable isotopes [20;22] and were expressed as<br />

mmol/kg.min. The equations are provided in Supplementary Figure 3. The abbreviated Weir<br />

equation was used to calculate the 24-hour energy expenditure. Glucose oxidation and fatty<br />

acid oxidation rates were derived from oxygen consumption and carbon oxide production<br />

as reported previously [25]. Glucose oxidation during insulin infusion was addditionally<br />

expressed as percentage of glucose Rd.<br />

156


Statistics:<br />

Data are presented as mean values ± standard deviation (SD), or as median (interquartile<br />

range) in case of skewed distribution. Absolute changes from baseline (on treatment value<br />

minus pre treatment value) were compared between the groups using the Kruskal-Wallis test.<br />

Non-parametric analysis was chosen due to the relatively small number of participants and the<br />

uneven group sizes. Only in case of a significant finding, prednisolone 7.5 mg and prednisolone<br />

30 mg were compared against placebo by posthoc testing, using the Mann-Whitney U test. To<br />

correct for multiple testing, Bonferroni correction was applied by multiplying the obtained<br />

p-value from the Mann-Whitney U test by the numbers of comparisons (i.e. two) that were<br />

carried out. All statistical analyses were run on SPSS for Windows version 15.0 (SPSS, Chicago,<br />

IL, USA). A p


Chapter 8 Glucocorticoids affect intermediary metabolism<br />

twith prednisolone 7.5 mg (Table 2). Pre-treatment, glucose oxidation increased to to 20.0<br />

(18.3-23.7) mmol/kg.min (9±5% of Rd) during hyperinsulinemia (P


Lipid Metabolism:<br />

Lipolysis in the fasted state tended to be decreased by prednisolone treatment (P=0.062),<br />

which was driven by the prednisolone 30 mg arm (P=0.09) (Figure 2a). Insulin-mediated<br />

suppression of lipolysis was markedly and dose-dependently impaired by prednisolone during<br />

hyperinsulinemic conditions (Figure 2b,c). Similarly to changes in lipolysis, basal NEFA levels<br />

were decreased by prednisolone treatment, which seemed to be driven by prednisolone 30<br />

mg (Table 2). During step 1 of insulin infusion (20 mU m2 min-1 ), insulin-mediated suppression<br />

of plasma NEFA levels was dose-dependently decreased by prednisolone treatment<br />

(Supplementary Table 2). During the second step of insulin infusion (60 mU m2 min-1 ), NEFA<br />

levels were reduced to detection level in the placebo group and prednisolone 7.5 mg group,<br />

but remained detectable in participants in the prednisolone 30 mg arm (Supplementary Table<br />

2). Fasting triacylglycerol (TG) levels were increased by prednisolone 30 mg only (Table 3).<br />

In the total study population, pre-treatment, fasting fatty acid oxidation rate was 1.5 (1.3-<br />

1.8) mmol/kg.min and decreased during insulin infusion to 0.4 (0.2-0.7) mmol kg-1 min-1 (p


Chapter 8 Glucocorticoids affect intermediary metabolism<br />

Table 2. Resting energy expenditure and glucose- and fatty acid oxidation in the fasted state and<br />

during insulin infusion before and during two-week treatment with placebo, prednisolone 7.5 mg<br />

daily or prednisolone 30 mg daily.<br />

Basal REE<br />

(kJ/kg.day)<br />

REE during clamp<br />

(kJ/kg.day)<br />

Basal glucose oxidation<br />

(mmol/kg.min)<br />

Glucose oxidation clamp<br />

(mmol/kg.min)<br />

Glucose oxidation clamp<br />

(% of Rd)<br />

Basal fatty acid oxidation<br />

(mmol/kg.min)<br />

Fatty acid oxidation clamp<br />

(mmol/kg.min)<br />

160<br />

Pre-treatment On-treatment P1 P2 P3<br />

PLB 95 (86-110) 89 (86-98)<br />

7.5 98 (91-107) 99 (92-109) 0.139 NA NA<br />

30 106 (91-114) 107 (98-110)<br />

PLB 102 (95-114) 100 (95-111)<br />

7.5 105 (97-110) 106 (99-111) 0.352 NA NA<br />

30 111 (96-111) 109 (96-111)<br />

PLB 7.7 (4.9-9.0) 5.5 (4.2-8.0)<br />

7.5 5.8 (3.0-8.5) 5.8 (3.2-12.6) 0.006 0.430 0.002<br />

30 5.1 (3.0-7.7) 11.2 (7.7-14.6)<br />

PLB 20.3 (18.8-22.2) 19.0 (15.8-25.2)<br />

7.5 19.6 (17.5-24.0) 16.1 (11.8-20.1) 0.133 NA NA<br />

30 19.4 (15.5-25.4) 20.0 (17.4-22.8)<br />

PLB 11.4 (7.8-16.1) 9.0 (5.8-15.9)<br />

7.5 9.4 (4.9-12.0) 9.3 (4.8-13.6) < 0.001 0.728 < 0.001<br />

30 8.5 (4.9-9.8) 25.1 (15.8-41.3)<br />

PLB 1.4 (1.2-1.5) 1.3 (1.2-1.8)<br />

7.5 1.4 (1.2-1.6) 1.6 (1.1-1.7) 0.02 0.640 0.022<br />

30 1.7 (1.5-1.9) 1.1 (1.0-1.5)<br />

PLB 0.4 (0.1-0.6) 0.5 (0.1-0.7)<br />

7.5 0.5 (0.1-0.7) 0.7 (0.3-1.0) 0.083 NA NA<br />

30 0.5 (0.2-0.8) 0.5 (0.2-0.7)<br />

Median (interquartile range) are provided. Between-group changes from baseline were tested by by<br />

Kruskal-Wallis (P1). In case of a significant finding, posthoc testing (by Mann-Whitney U) was done (P2<br />

and P3). Abbreviations: Rd: rate of disappearance; REE: resting energy expenditure. Treatment groups:<br />

PLB=placebo; 7.5=prednisolone 7.5 mg daily; 30=prednisolone 30 mg daily.<br />

Glucoregulatory hormones:<br />

Prednisolone 7.5 mg did not change fasting plasma insulin levels or glucagon<br />

levels. Prednisolone 30 mg, in contrast, induced both fasting hyperinsulinemia and<br />

hyperglucagonemia (Table 3). Before treatment, insulin levels in the entire study population<br />

during the clamp were 171 (153-183) pmol/l (step 1) and 533 (485-564) pmol/l (step 2).<br />

These levels remained unchanged during the on-treatment clamps, although there was a<br />

tendency towards increased insulin levels in the placebo group (Supplementary Table 2).<br />

Glucagon levels remained increased during step 1, but not step 2, of the hyperinsulinemic<br />

clamp during prednisolone 30 mg treatment (Supplementary Table 2).


Figure 3. The effect of prednisolone treatment on protein metabolism. a Valine turnover in the basal state,<br />

was not affected by prednisolone treatment. b Overall, valine turnover was increased during step 1 of<br />

the hyperinsulinemic-euglycemic clamp by prednisolone treatment. This seemed driven by prednisolone<br />

30 mg in post-hoc analysis, but failed to reach statistical significance. c A similar pattern was observed<br />

during step 2 of the clamp, where overall prednisolone treatment increased valine turnover, however<br />

in post-hoc testing, no significance was reached for either dose as compared to placebo. Data represent<br />

mean ± SEM. Black bars, before treatment; white bars, day 14 of treatment. Between-group changes from<br />

baseline were tested by Kruskal-Wallis (indicated by top line). Posthoc tests were done by Mann-Whitney<br />

U with Bonferroni correction for multiple testing (indicated by line with brackets). *P


Chapter 8 Glucocorticoids affect intermediary metabolism<br />

Safety and Tolerability<br />

One subject in the prednisolone group complained of gastric discomfort which was mild of<br />

nature and did not require any intervention. Otherwise, no treatment-related side effects<br />

were reported in any of the groups.<br />

DISCUSSIoN<br />

Low-dose GC treatment is chronically prescribed to a great number of patients, but to date it is<br />

uncertain whether low-dose GC treatment induces adverse metabolic effects, as is evident for<br />

higher GC doses. The present study is the first to demonstrate that low-dose GC therapy, that<br />

is just above the normal daily cortisol replacement dose for adrenal insufficiency, significantly<br />

impairs the effects of insulin on glucose and lipid metabolism, but not on whole-body protein<br />

metabolism. Using a relatively long exposure time, i.e. two weeks, we report that treatment<br />

with prednisolone 7.5 mg once daily impaired the ability of insulin to suppress EGP, wholebody<br />

lipolysis and plasma NEFA levels. These effects were present after only two weeks of<br />

treatment and occurred in the absence of changes in body weight, LBM, liver fat content and<br />

body fat distribution. Our data, demonstrating clear low-dose prednisolone-induced hepatic<br />

and adipose tissue insulin resistance, provide supportive mechanisms for observations made<br />

in large retrospective, case-control studies that reported increased risk to develop diabetes<br />

[15] and increased need to initiate blood-glucose lowering therapy [14] during or following<br />

low-dose GC therapy.<br />

In prospective studies, the reported incidence of diabetes at lower GC doses has been more<br />

modest, although there was considerable variation depending on the patient group exposed<br />

[16]. The numbers reported in these clinical trials may underestimate the actual incidence of<br />

diabetes. Although not well specified in the different studies, most studies relied on fasting<br />

glucose measurements to monitor changes in glucose tolerance. Our data demonstrate a<br />

very mild increase in fasting glucose and unchanged fasting insulin levels, but show evident<br />

changes in metabolic fluxes under insulin-stimulated conditions during treatment with<br />

low-dose prednisolone. This observation is in line with previous studies reporting large<br />

postprandial glucose excursions with only modest changes in fasting glucose levels [19;26].<br />

These data imply that, in order to properly assess the effects of GCs on glucose metabolism,<br />

measurements under stimulated conditions should be performed, such as an OGTT or a<br />

glucose day curve.<br />

162


By including an additional treatment arm, we were able to study dose-dependent effects of<br />

prednisolone treatment. Prednisolone 30 mg, an induction dosage often used in the clinic for<br />

short-term treatment, impaired the effects of insulin on EGP, glucose disposal and lipolysis<br />

to a greater extent than prednisolone 7.5 mg. In addition, prednisolone 30 mg increased<br />

proteolysis under hyperinsulinemic conditions, increased glucagon levels and augmented<br />

fasting TG levels. Surprisingly, prednisolone 30 mg increased both basal glucose oxidation<br />

(absolute levels) and glucose oxidation during the clamp (expressed as % of Rd). Increased<br />

basal glucose oxidation has, to our knowledge, not been reported previously following GC<br />

treatment. We hypothesize that prednisolone treatment induced metabolic inflexibility.<br />

This phenomenon refers to the inability to adjust fuel oxidation to substrate availability.<br />

As such, impaired fat oxidation in the fasted state was extensively shown in obese, insulin<br />

resistant and type 2 diabetic individuals [27]. In addition, following high-dose prednisolone<br />

treatment, fasting plasma glucose levels were, albeit it marginally, increased, which could<br />

have contributed to augmented glucose oxidation in the basal state. The finding that GC<br />

treatment increased glucose oxidation under hyperinsulinemic conditions, has been reported<br />

previously in nondiabetic individuals who were exposed to dexamethasone treatment<br />

[28]. GCs are well to known to impair the insulin-stimulated glycogen synthesis pathway<br />

[29] and therefore, increased glucose oxidation was proposed to serve as a mechanisms to<br />

compensate for reduced glycogen synthesis, in the presence of higher glucose levels within<br />

the skeletal muscle [28]. Our data similarly indicate preferential glucose oxidation compared<br />

to glycogen synthesis pathways, since absolute glucose oxidation rates were not altered<br />

despite lower glucose uptake. Glucose oxidation as percentage of Rd was thus increased.<br />

Finally, prednisolone 30 mg tended to decrease basal lipolysis and NEFA levels as compared<br />

to placebo. In acute studies, cortisol infusion was shown to increase lipolysis in the fasted<br />

state. In these studies, hyperinsulinemia was prevented by performing a pancreatic clamp<br />

with insulin infusion at basal levels [30;31]. In studies addressing the effects of chronic GC<br />

treatment, typically no changes were observed in whole-body lipolysis in the fasted state [32],<br />

and fasting insulin levels were increased [33-35]. In our study we found a tendency towards<br />

decreased lipolysis. Since lipolysis is more sensitive to small changes in insulin concentration<br />

then EGP [36;37], and plasma NEFA levels are closely correlated to changes in lipolysis,<br />

we hypothesize that decreased basal lipolysis and NEFA levels in individuals treated with<br />

prednisolon 30 mg daily may be due to the observed prednisolone-induced 80% increase<br />

in fasting plasma insulin levels. The negative correlation between the treatment-induced<br />

change in fasting plasma insulin and fasting NEFA levels (r=-0.564; p=0.001) supports our<br />

hypothesis. Another hypothesis is that prednisolone 30 mg decreased the sympathetic tone<br />

to adipose tissue resulting in lower rates of basal lipolysis [38].<br />

163<br />

8<br />

Chapter 8


Chapter 8 Glucocorticoids affect intermediary metabolism<br />

The mechanisms underlying prednisolone-induced insulin resistance on the various metabolic<br />

fluxes have not been elucidated. GCs were shown to interfere with insulin signaling in vitro and<br />

in vivo in rodents in liver [39], adipose tissue [40] and skeletal muscle[41]. On the other hand,<br />

GCs may also directly activate genes involved in the regulation of various metabolic fluxes. As<br />

such, GCs were shown to induce phosphoenolpyruvate carboxykinase, the rate controlling<br />

enzyme in hepatic glucose production [42]. In addition, hormone-sensitive lipase, a major<br />

regulator of adipose tissue lipolysis was demonstrated to be regulated by GCs [43]. Moreover,<br />

GCs directly activated muscle ring finger-1 and Atrogin-1, proteins involved in skeletal muscle<br />

proteolysis [44]. Finally, GCs impaired glucose transport into skeletal muscle independent of<br />

changes in insulin signaling [6].<br />

In the present study we exposed healthy volunteers to different doses of prednisolone,<br />

allowing us to specifically measure the metabolic effects of prednisolone. In clinical practice,<br />

prednisolone is used to treat patients with chronic inflammatory diseases. Since systemic<br />

inflammation also impairs insulin sensitivity [45], the effects of GCs on various fluxes, given<br />

their combined anti-inflammatory and metabolic effects, could be more complex in this<br />

population.<br />

One potential, yet inevitable limitation of our experimental design when addressing the effects<br />

of real-life oral prednisolone treatment, as compared to previously reported, more artificial<br />

cortisol infusion studies, is that our clamp tests were performed under non-steady state<br />

prednisolone levels. Since prednisolone has a T of 4-6 hours, the final part of the clamp may<br />

1/2<br />

have been performed under lower prednisolone levels. However, plasma prednisolone levels<br />

may be a poor marker of its pharmacodynamics, since many effects exerted by prednisolone<br />

are genomic of nature, which take a number of hours to become effected. In addition, our<br />

participants were treated for two weeks, in contrast to the acute studies that have been mostly<br />

conducted up to now, and therefore, differences caused by the acute effects of prednisolone<br />

treatment may be very limited.<br />

We conclude that GC treatment, even at a low, so-called maintenance dose that is prescribed<br />

to large numbers of patients for prolonged periods of time, impairs both glucose metabolism<br />

and lipolysis by impairing insulin’s metabolic actions on the liver and adipose tissue. The<br />

results of this study uncover the mechanisms by which GC dosages just above cortisol<br />

replacement levels, especially when used chronically, may impair glucose tolerance in<br />

susceptible patients. Physicians treating patients with low-dose GCs should be aware of the<br />

induction of metabolic disturbances and should not solely rely on fasting measurements. In<br />

addition, our study indicates that insulin-sensitizing therapies could be considered when<br />

164


treating patients with GC therapy. As such, the thiazolidinedione troglitazone was shown to<br />

prevent GC-induced glucose intolerance in healthy humans [46]. Troglitazone, however, is<br />

no longer available for use in humans, and therefore a search for other options to alleviate<br />

GC-induced insulin resistance seems justified. In this regard, the current development of<br />

the so called dissociated glucocorticoid receptor agonists, which aim to separate the antiinflammatory<br />

and dysmetabolic effects of GCs, may be highly interesting [47].<br />

Acknowledgements<br />

This paper was written within the framework of project T1-106 of the Dutch Top Institute<br />

<strong>Pharma</strong>.<br />

165<br />

8<br />

Chapter 8


Chapter 8 Glucocorticoids affect intermediary metabolism<br />

REFERENCE LIST<br />

1. Bijlsma jW, van der Goes MC, Hoes jN, jacobs jW, Buttgereit F, Kirwan j. Low dose glucocorticoid<br />

166<br />

therapy in rheumatoid arthritis: an obligatory therapy. Ann N Y Acad Sci. 2010;1193:123-6.<br />

2. Hernandez-Rodriguez j, Cid MC, Lopez-Soto A, Espigol-Frigole G, Bosch X. Treatment of polymyalgia<br />

rheumatica: a systematic review. Arch Intern Med. 2009;169(20):1839-50.<br />

3. Salvarani C, Cantini F, Hunder GG. Polymyalgia rheumatica and giant-cell arteritis. Lancet.<br />

2008;372(9634):234-45.<br />

4. Parker Bj, Bruce IN. High dose methylprednisolone therapy for the treatment of severe systemic<br />

lupus erythematosus. Lupus. 2007;16(6):387-93.<br />

5. Lichtenstein GR, Abreu MT, Cohen R, Tremaine W. American Gastroenterological Association<br />

Institute medical position statement on corticosteroids, immunomodulators, and infliximab in<br />

inflammatory bowel disease. Gastroenterology. 2006;130(3):935-9.<br />

6. van Raalte DH, Ouwens DM, Diamant M. Novel insights into glucocorticoid-mediated diabetogenic<br />

effects: towards expansion of therapeutic options? Eur j Clin Invest. 2009;39(2):81-93.<br />

7. Buttgereit F, Da Silva jA, Boers M, Burmester GR, Cutolo M, jacobs j, Kirwan j, Kohler L, van Riel P,<br />

Vischer T, Bijlsma jW. Standardised nomenclature for glucocorticoid dosages and glucocorticoid<br />

treatment regimens: current questions and tentative answers in rheumatology. Ann Rheum.Dis.<br />

2002;61(8):718-22.<br />

8. Wajngot A, Giacca A, Grill V, Vranic M, Efendic S. The diabetogenic effects of glucocorticoids are more<br />

pronounced in low- than in high-insulin responders. Proc Natl Acad Sci U S A. 1992;89(13):6035-9.<br />

9. Larsson H, Ahren B. Insulin resistant subjects lack islet adaptation to short-term dexamethasoneinduced<br />

reduction in insulin sensitivity. Diabetologia. 1999;42(8):936-43.<br />

10. Gravholt CH, Dall R, Christiansen jS, Moller N, Schmitz O. Preferential stimulation of abdominal<br />

subcutaneous lipolysis after prednisolone exposure in humans. Obes Res. 2002;10(8):774-81<br />

11. Taskinen MR, Kuusi T, Yki-jarvinen H, Nikkila EA. Short-term effects of prednisone on serum lipids<br />

and high density lipoprotein subfractions in normolipidemic healthy men. j Clin Endocrinol Metab.<br />

1988;67(2):291-9.<br />

12. Short KR, Bigelow ML, Nair KS. Short-term prednisone use antagonizes insulin’s anabolic effect on<br />

muscle protein and glucose metabolism in young healthy people. Am j Physiol Endocrinol Metab.<br />

2009;297(6): E1260-8.<br />

13. Fernandez-Rodriguez E, Stewart PM, Cooper MS. The pituitary-adrenal axis and body composition.<br />

Pituitary. 2009;12(12):105-15.<br />

14. Gurwitz jH, Bohn RL, Glynn Rj, Monane M, Mogun H, Avorn j. Glucocorticoids and the risk for<br />

initiation of hypoglycemic therapy. Arch Intern Med. 1994;154(1):97-101.<br />

15. Gulliford MC, Charlton j, Latinovic R. Risk of diabetes associated with prescribed glucocorticoids in<br />

a large population. Diabetes Care. 2006;29(12):2728-9.


16. Hoes jN, jacobs jW, Verstappen SM, Bijlsma jW, Van der Heijden Gj. Adverse events of low- to<br />

medium-dose oral glucocorticoids in inflammatory diseases: a meta analysis. Ann Rheum Dis.<br />

2009;68(12):1833-8.<br />

17. Tushuizen ME, Bunck MC, Pouwels Pj, Bontemps S, van Waesberghe jH, Schindhelm RK, Mari A,<br />

Heine Rj, Diamant M. Pancreatic fat content and beta-cell function in men with and without type 2<br />

diabetes. 2007. Diabetes Care;30(3):2916-21.<br />

18. Tushuizen ME, Pouwels Pj, Bontemps S, Rustemeijer C, Matikanen N, Heine Rj, Taskinen MR,<br />

Diamant M. Postprandial lipid and apolipoprotein responses following three consecutive meals<br />

associate with liver fat content in type 2 diabetes and the metabolic syndrome. Atherosclerosis.<br />

2010;211(1): 308-14.<br />

19. van Raalte DH, Nofrate V, Bunck MC, van Iersel T, Elassaiss Schaap j, Nassander UK, Heine Rj, Mari<br />

A, Dokter WH, Diamant M. Acute and 2-week exposure to prednisolone impair different aspects of<br />

beta-cell function in healthy men. Eur j Endocrinol. 2010;162(4):729-35.<br />

20. Ackermans MT, Ruiter AF, Endert E. Determination of glycerol concentrations and glycerol isotopic<br />

enrichments in human plasma by gas chromatography/mass spectrometry. Anal Biochem.<br />

1998;258(1):80-6.<br />

21. Ackermans MT, Pereira Arias AM, Bisschop PH, Endert E, Sauerwein HP, Romijn JA. The quantification<br />

of gluconeogenesis in healthy men by (2)H2O and [2-(13)C] glycerol yields different results: rates<br />

of gluconeogenesis in healthy men measured with (2)H2O are higher than those measured with [2-<br />

(13)C]glycerol. j Clin Endocrinol Metab. 2001;86(5):2220-6.<br />

22. Geukers VG, Oudshoorn jH, Taminiau jA, van der Ent CK, Schilte P, Ruiter AF, Ackermans MT, Endert<br />

E, jonkers-Schuitema CF, Heymans HS, Sauerwein HP. Short-term protein intake and stimulation of<br />

protein synthesis in stunted children with cystic fibrosis. Am J Clin Nutr. 2005;81(3):605-10.<br />

23. Finegood DT, Bergman RN, Vranic M. Estimation of endogenous glucose production during<br />

hyperinsulinemic-euglycemic glucose clamps. Comparison of unlabeled and labeled exogenous<br />

glucose infusates. Diabetes. 1987;36(8):914-24.<br />

24. Steele R. Influences of glucose loading and of injected insulin on hepatic glucose output. Ann NY<br />

Acad Sci. 1959;82:420-30.<br />

25. Frayn KN. Calculation of substrate oxidation rates in vivo from gaseous exchange. j Appl Physiol.<br />

1983;55(2):628-34.<br />

26. Clore jN, Thurby-Hay L. Glucocorticoid-induced hyperglycemia. Endocr Pract. 2009;15(5):469-74.<br />

27. Corpeleijn E, Saris WH, Blaak EE. Metabolic flexibility in the development of insulin resistance and<br />

type 2 diabetes: effects of lifestyle. Obes Rev. 2009;10(2):178-193.<br />

28. Henriksen jE, Alford F, Vaag A, Handberg A, Beck-Nielsen H. Intracellular skeletal muscle glucose<br />

metabolism is differentially altered by dexamethasone treatment of normoglycemic relatives of type<br />

2 diabetic patients. Metabolism. 1999;48(9):1128-35.<br />

167<br />

8<br />

Chapter 8


Chapter 8 Glucocorticoids affect intermediary metabolism<br />

29. Ruzzin j, Wagman AS, jensen j. Glucocorticoid-induced insulin resistance in skeletal muscles: defects<br />

168<br />

in insulin signalling and the effects of a selective glycogen synthase kinase-3 inhibitor. Diabetologia.<br />

2005;48(10):2119-30.<br />

30. Djurhuus CB, Gravholt CH, Nielsen S, Mengel A, Christiansen jS, Schmitz OE, Moller N. Effects of<br />

cortisol on lipolysis and regional interstitial glycerol levels in humans. Am j Physiol Endocrinol<br />

Metab. 2002;283(1):E172-7.<br />

31. Djurhuus CB, Gravholt CH, Nielsen S, Pedersen SB, Moller N, Schmitz O. Additive effects of cortisol<br />

and growth hormone on regional and systemic lipolysis in humans. Am.j Physiol Endocrinol Metab.<br />

2004;286(3):E488-94.<br />

32. Macfarlane DP, Forbes S, Walker BR. Glucocorticoids and fatty acid metabolism in humans: fuelling<br />

fat redistribution in the metabolic syndrome. j Endocrinol. 2008;197(2):189-204.<br />

33. Birkenhager JC, Timmermans HA, Lamberts SW. Depressed plasma FFA turnover rate in Cushing’s<br />

syndrome. j Clin Endocrinol Metab. 1976;42(1):28-32.<br />

34. Gravholt CH, Dall R, Christiansen jS, Moller N, Schmitz O. Preferential stimulation of abdominal<br />

subcutaneous lipolysis after prednisolone exposure in humans. Obes.Res. 2002;10(8):774-81<br />

35. Miyoshi H, Shulman GI, Peters Ej, Wolfe MH, Elahi D, Wolfe RR. Hormonal control of substrate cycling<br />

in humans. j Clin Invest. 1988;81(5):1545-55.<br />

36. Bonadonna RC, Groop LC, Zych K, Shank M, DeFronzo RA. Dose-dependent effect of insulin on<br />

plasma free fatty acid turnover and oxidation in humans. Am j Physiol. 1990;259(5 Pt 1):E736-50.<br />

37. Gelding SV, Coldham N, Anyaoku V, Heslop K, Halliday D, johnston DG. Differential metabolic actions<br />

of biosynthetic insulin analogues in normal man assessed by stable isotopic tracers. Diabet Med.<br />

1993;10(5):470-6.<br />

38. Nakada MT, Stadel jM, Poksay KS, Crooke ST. Glucocorticoid regulation of beta-adrenergic receptors<br />

in 3T3-L1 preadipocytes. Mol <strong>Pharma</strong>col. 1987;31(4):377-84.<br />

39. Rojas FA, Hirata AE, Saad Mj. Regulation of insulin receptor substrate-2 tyrosine phosphorylation in<br />

animal models of insulin resistance. Endocrine. 2003;21(2):115-22.<br />

40. Lundgren M, Buren j, Ruge T, Myrnas T, Eriksson jW. Glucocorticoids down-regulate glucose uptake<br />

capacity and insulin-signaling proteins in omental but not subcutaneous human adipocytes. j Clin<br />

Endocrinol Metab. 2004;89(6):2989-97.<br />

41. Saad Mj, Folli F, Kahn jA, Kahn CR. Modulation of insulin receptor, insulin receptor substrate-1,<br />

and phosphatidylinositol 3-kinase in liver and muscle of dexamethasone-treated rats. j Clin Invest.<br />

1993;92(4):2065-72.<br />

42. Vegiopoulos A, Herzig S. Glucocorticoids, metabolism and metabolic diseases. Mol Cell Endocrinol.<br />

2007;275(1-2):43-61.<br />

43. Slavin BG, Ong jM, Kern PA. Hormonal regulation of hormone-sensitive lipase activity and mRNA<br />

levels in isolated rat adipocytes. j Lipid Res. 1994;35(9):1535-41.


44. Schakman O, Gilson H, Thissen jP. Mechanisms of glucocorticoid-induced myopathy. j Endocrinol.<br />

2008;197(1):1-10.<br />

45. Dessein PH, joffe BI. Insulin resistance and impaired beta cell function in rheumatoid arthritis.<br />

Arthritis Rheum. 2006;54(9):2765-75.<br />

46. Willi SM, Kennedy A, Wallace P, Ganaway E, Rogers NL, Garvey WT. Troglitazone antagonizes<br />

metabolic effects of glucocorticoids in humans: effects on glucose tolerance, insulin sensitivity,<br />

suppression of free fatty acids, and leptin. Diabetes. 2002;51(10):2895-902.<br />

47. Rosen j, Miner jN. The search for safer glucocorticoid receptor ligands. Endocr Rev 2005;26(3):452-<br />

64.<br />

169<br />

8<br />

Chapter 8


Chapter 8 Glucocorticoids affect intermediary metabolism<br />

SUPPLEMENTARy MATERIAL<br />

Supplementary Figure 1. Study Design (a) and overview of the 2-step hyperinsulinemic-euglycemic<br />

clamp protocol (b).<br />

Day -14 -2 -1 0<br />

7<br />

13 14<br />

S T1 T2 M<br />

V T1 T2<br />

170<br />

a<br />

Baseline<br />

PLB, PRED 7.5 mg or PRED 30 mg QD<br />

Offtreatment<br />

S: Screening T1: MRI/DEXA scan T2: Clamp M: start study medication V: Follow-up visits<br />

b<br />

Insulin 60 mU/m2 Insulin 20 mU/m .min<br />

Glucose 5.0 mM<br />

2 .min<br />

Variable Glucose 20% enriched with 6,6 –2H 2 Glucose<br />

6,6 –2H 2 Glucose bolus + Continuous infusion<br />

1-13C 1 -Valine bolus + continuous infusion<br />

1,1,2,3,3-2H 5 -Glycerol bolus + continuous infusion<br />

0 120 Time (min)<br />

240<br />

360<br />

Supplementary Figure 2. Patient flow diagram. 100% of the randomised participants completed the<br />

study<br />

28<br />

V


Supplementary Figure 3. Kinetic equations for the steady-state (a) and for the non-steady state (b).<br />

Where: R a = rate of appearance; TTR tracer = tracer/tracee ratio of the tracer infused (=1); I tracer = tracer<br />

infusion rate; TTR p = tracer/tracee ratio in plasma; TTR exo = tracer/tracee ratio of exogenous glucose<br />

infusion; I exo : infusion rate of exogenous glucose; R d = rate of disappearance; R a (t)= average R a between t=1<br />

and t=2; TTR p (t)= average TTR p between t=1 and t=2; pV= distribution volume; C unl (t)= average plasma<br />

concentration tracee between t=1 and t=2; dTTR p (t)/dt = change in plasma TTR p between t=1 and t=2;<br />

R d (t) = average R d between t=1 and t=2; dC unl (t)/dt = change in tracee concentration between t=1 and t=2;<br />

EP(t) = average endogenous production between t=1 and t=2; I unl = tracee infusion rate.<br />

171<br />

8<br />

Chapter 8


Chapter 8 Glucocorticoids affect intermediary metabolism<br />

Supplementary Table 1. Parameters of body composition and body fat distribution before and during<br />

two-week treatment with placebo, prednisolone 7.5 mg or prednisolone 30 mg.<br />

BMI<br />

(kg/m2 )<br />

LBM<br />

(%)<br />

Fat mass<br />

(%)<br />

Liver fat content<br />

(%)<br />

SAT area<br />

(cm2 )<br />

VAT area<br />

(cm2 )<br />

172<br />

Pre-treatment On-treatment P<br />

PLB 23.0±2.3 22.8±2.2<br />

7.5 22.3±2.0 22.3±1.8<br />

0.449<br />

30 22.6±1.1 22.6±1.1<br />

PLB 78.6±7.9 80.2±10.0<br />

7.5 82.1±3.9 81.8±3.7<br />

0.275<br />

30 80.7±2.5 80.9±2.0<br />

PLB 16.9±4.7 17.1±4.6<br />

7.5 15.9±2.8 16.1±3.7<br />

0.503<br />

30 14.8±3.5 14.7±3.2<br />

PLB 1.3 (0.3-2.5) 1.5 (0.4-3.5)<br />

7.5 0.8 (0.4-2.1) 1.3 (0.7-2.4)<br />

0.537<br />

30 1.0 (0.5-1.6) 1.6 (1-1.8)<br />

PLB 131 (91-202) 130 (102-201)<br />

7.5 127 (94-154) 130 (89-160)<br />

0.447<br />

30 129 (114-151) 132 (114-147)<br />

PLB 82 (48-107) 71 (48-112)<br />

7.5 72 (69-99) 86 (66-93)<br />

0.291<br />

30 77 (67-88) 80 (72-88)<br />

Mean±SD or median (interquartile range) are provided. Between-group changes from baseline were<br />

tested by Kruskal-Wallis (P). No significnant differences were observed during treatment. Abbreviations:<br />

BMI: body mass index; LBM: lean body mass; SAT: subcutaneous adipose tissue; VAT: visceral adipose<br />

tissue. Treatment groups: PLB=placebo; 7.5=prednisolone 7.5 mg daily; 30=prednisolone 30 mg daily.


Supplementary Table 2. Metabolic parameters during the two-step hyperinsulinemic-euglycemic clamp<br />

before and during two-week treatment with placebo, prednisolone 7.5 mg or prednisolone 30 mg.<br />

Glucose step 1<br />

(mmol/l)<br />

Glucose step 2<br />

(mmol/l)<br />

Insulin step 1<br />

(pmol/l)<br />

Insulin step 2<br />

(pmol/l)<br />

Glucagon step 1<br />

(pmol/l)<br />

Glucagon step 2<br />

(pmol/l)<br />

NEFA step 1<br />

(mmol/l)<br />

NEFA step 2<br />

(mmol/l)<br />

Pre-treatment On-treatment P1 P2 P3<br />

PLB 5.0±0.1 5.0±0.2<br />

7.5 4.9±0.1 4.9±0.2<br />

0.389 NA NA<br />

30 5.0±0.2 5.0±0.1<br />

PLB 5.1±0.2 5.1±0.2<br />

7.5 5.1±0.2 5.0±0.1<br />

0.860 NA NA<br />

30 5.0±0.2 5.0±0.2<br />

PLB 172±36 179±35<br />

7.5 171±26 161±32<br />

0.398 NA NA<br />

30 165±33 151±39<br />

PLB 529±74 571±102<br />

7.5 531±73 522±81<br />

0.055 NA NA<br />

30 497±86 481±105<br />

PLB 41±8 36±5<br />

7.5 48±6 52±13<br />

0.016 0.232 0.02<br />

30 38±9 58±20<br />

PLB 31±8 32±7<br />

7.5 35±8 41±12<br />

0.156 NA NA<br />

30 32±8 44±15<br />

PLB 0.03 (0.02-0.04) 0.03 (0.02-0.04)<br />

7.5 0.03 (0.02-0.05) 0.05 (0.04-0.1) < 0.0001 0.034 0.0002<br />

30 0.02 (0.02-0.03) 0.12 (0.09-0.16)<br />

PLB


Chapter 9<br />

Glycosphingolipid Metabolism and<br />

Mitochondrial Function do not Explain<br />

Glucocorticoid-Induced Insulin Resistance<br />

in Healthy Men<br />

D.H. van Raalte*, M. Brands*, M. João Ferraz, H.P. Sauerwein,<br />

A.J.Verhoeven, J.F.G.Aerts, M.Diamant and M.J. Serlie<br />

*Both authors contributed equally<br />

Submitted


Chapter 9 Glucocorticoids, mitochondrial function & glycosphingolipid metabolism<br />

Abstract<br />

objective: Glucorticoids (GCs) are well known to induce skeletal muscle insulin resistance,<br />

however, the mechanisms underlying this phenomenon are presently incompletely<br />

understood. In this study we explored the involvement of changes in muscle ceramide and<br />

ganglioside GM3 levels, and the potential role of mitochondrial dysfunction in GC-induced<br />

insulin resistance in healthy men.<br />

Research design and methods: In a randomized, placebo-controlled, double blind, dose-<br />

response intervention study, 32 healthy men (age: 22±3 years; BMI 22.4±1.7 kg/m 2 ) were<br />

allocated to prednisolone 7.5 mg once daily (n=12), prednisolone 30 mg once daily (n=12),<br />

or placebo (n=8) for two weeks. At baseline and on day 14 of treatment, insulin sensitivity<br />

was measured by hyperinsulinemic-euglycemic clamp. In skeletal muscle biopsies, levels of<br />

ceramide and GM3 were measured, and mitochondrial function was determined by ex vivo<br />

respirometry.<br />

Results: Prednisolone treatment dose-dependently impaired skeletal muscle insulin<br />

sensitivity. Muscle ceramide and GM3 concentrations were not affected by low- or highdose<br />

prednisolone treatment (P>0.05 for both). In addition, mitochondrial respiratory<br />

fluxes were not affected by prednisolone treatment (all P>0.05).<br />

Conclusion: No apparent role for altered muscle ceramide and GM3 levels, or mitochondrial<br />

dysfunction was observed in GC-induced insulin resistance in healthy males. Further studies<br />

are warranted to investigate the mechanisms by which GCs impair insulin sensitivity in<br />

skeletal muscle.<br />

176


INTRoDUC<strong>TI</strong>oN<br />

Glucocorticoids (GCs) are important anti-inflammatory and immunosuppressive agents that<br />

are clinically used in a broad spectrum of disease entities [1, 2]. However, GCs also induce<br />

metabolic side effects, which limit their use. Patients exposed to excess GCs levels develop<br />

weight gain, in specific central adiposity, breakdown of skeletal muscle mass [3], dyslipidemia<br />

characterized by increased nonesterified fatty acid (NEFA) levels [4], hepatic and peripheral<br />

insulin resistance [5], glucose intolerance, and overt diabetes [6-8]. Recently we reported on<br />

the metabolic effects of a two-week treatment with prednisolone 7.5 mg daily and prednisolone<br />

30 mg daily and described a dose-dependent decrease of insulin sensitivity with concurrent<br />

elevated fasting triglyceride levels and increased NEFA levels during hyperinsulinemia. This<br />

effect occurred in the absence of significant changes in body weight, lean body mass, liver fat<br />

content and body fat distribution [5].<br />

Despite extensive research, the mechanisms by which GCs induce skeletal muscle insulin<br />

resistance remain at present to be elucidated. It was demonstrated in rodents that GCs impair<br />

insulin signaling. As such, GC treatment in rats was shown to reduce the phosphorylation<br />

of insulin receptor substrate (IRS)-1, phosphatidylinositol 3-kinase (PI3-K) and Akt/protein<br />

kinase B (PKB). This impairment in insulin signaling induced a reduction in transportation<br />

of glucose transporter (GLUT)-4 to the plasma membrane and reduced activation of glycogen<br />

synthase kinase (GSK)-3, resulting in impaired glucose uptake and glycogen synthesis<br />

respectively [9-13]. More recently, decreased phosphorylation of insulin signaling proteins<br />

[14] and glycogen synthase kinase [15] was also shown in skeletal muscle of healthy humans<br />

treated with GCs. However, pathophysiological mechanisms that underlie GC-induced defects<br />

in insulin signaling have not yet been elucidated.<br />

GC treatment increases lipolysis and plasma NEFA concentrations. This may endorse processes<br />

commonly referred to as lipotoxicity, which is induced through several mechanisms, amongst<br />

others by formation and accumulation of intracellular lipid metabolites [16]. Two of these<br />

metabolites are ceramide and the downstream ganglioside GM3. Muscle ceramide levels<br />

seem to be increased in patients with type 2 diabetes mellitus (T2DM) [17] and correlate in<br />

some, but not all studies, with insulin sensitivity [17-21]. Ceramide interferes with insulin<br />

signaling by reducing insulin-mediated phosphorylation of Akt/PKB [22-24]. Interestingly, it<br />

was shown that impairment of ceramide synthesis in rats prevented dexamethasone-induced<br />

insulin resistance in skeletal muscle [25]. GM3 has also been shown to be involved in insulin<br />

sensitivity in mice [26]. Gangliosides reside within the plasma membrane and are able to<br />

modulate insulin signaling at the level of the insulin receptor [27-31].<br />

177<br />

9<br />

Chapter 9


Chapter 9 Glucocorticoids, mitochondrial function & glycosphingolipid metabolism<br />

Intracellular lipid accumulation and peripheral insulin resistance have also been associated<br />

with decreased mitochondrial function [32]. Interestingly, chronic GC treatment in patients<br />

causes myopathy, characterized by increased proteolysis, oxidative stress and mitochondrial<br />

dysfunction [33]. Whether decreased mitochondrial function plays a role in GC-induced<br />

insulin resistance has not been elucidated.<br />

In the present study we investigated whether GC-induced peripheral insulin resistance<br />

could be explained by either accumulation of intramyocellular ceramide and GM3 levels or<br />

mitochondrial dysfunction in healthy men.<br />

METHoDS<br />

Participants: Thirty-two healthy normoglycemic Caucasian men were recruited via local<br />

advertisements. All participants were in good health as confirmed by medical history,<br />

physical examination, screening blood tests and a 75-g 2-hr oral glucose tolerance test<br />

(OGTT), performed at screening visit. Inclusion criteria were: age between 18-35 years and<br />

body mass index (BMI) ≤ 25 kg/m2 . Exclusion criteria were any previous or current illness,<br />

use of any medication, first-degree relative with T2DM, smoking, shift work and a history of<br />

GC use. In addition, participants who exercised more than two times per week were excluded.<br />

Participants were instructed not to change their physical activity or diet during the study. To<br />

achieve homogeneity of the study group, a physical fitness test was performed as described<br />

previously [34]. Participants with a maximal oxygen uptake (VO2 max) between 45 and<br />

60 ml/kg.min were included. The study was approved by an independent ethics committee<br />

and the study was conducted in accordance with the Declaration of Helsinki. All participants<br />

provided written informed consent before participation.<br />

Study design: Details on study design were reported previously [5]. Briefly, in a randomized,<br />

placebo-controlled, double-blind, dose-response intervention trial, 32 healthy volunteers<br />

were allocated to a treatment with prednisolone 7.5 mg once daily (n=12), prednisolone 30<br />

mg once daily (n=12) or placebo (n=8) for a period of two weeks using block randomization<br />

as carried out by the department of Experimental <strong>Pharma</strong>cology of the VU University Medical<br />

Center. Before and at day 14 of treatment, peripheral insulin sensitivity was measured<br />

during a hyperinsulinemic-euglycemic clamp using a stable glucose isotope. At the end<br />

of the clamp, biopsies from the vastus lateralis muscle were collected for measurement of<br />

glycosphingolipids and mitochondrial function. During all visits, including a follow-up visit at<br />

day 7 of treatment, safety and tolerability were assessed.<br />

178


Experimental protocol: The design of the hyperinsulinemic-euglycemic clamp was described<br />

in detail elsewhere [5]. In short, following an overnight fast and following measurement of<br />

the background enrichment of glucose, a primed, continuous infusion of [6,6-2H ]glucose<br />

2<br />

(prime: 11 mmol/kg; continuous: 0.11 mmol/kg.min) was started and continued until the<br />

end of the clamp. After a total of 4 hours of insulin infusion, blood samples were drawn for<br />

determination of glucose concentrations, isotope enrichment, and additional parameters of<br />

interest. The first 2 hours, insulin was infused at a rate of 20 mU/m2 .min, thereafter, insulin<br />

was infused at a rate of 60 mU/ m2 .min for 2 hours. Plasma glucose was kept at 5 mmol/l by<br />

a variable infusion of glucose 20% solution enriched with [6,6-2H ]glucose. Peripheral uptake<br />

2<br />

of glucose (rate of disappearance, R ) was calculated during the final 20 min of the clamp<br />

d<br />

using a modified version of the Steele equation as described [5].<br />

Muscle biopsies: Immediately after the last blood sample was drawn and with continuation<br />

of glucose and insulin infusion, a muscle biopsy was taken from the vastus lateralis muscle<br />

under local anesthesia with Lidocaine 20% (Fresenius Kabi; Den Bosch, the Netherlands).<br />

Biopsies were obtained using an automatic biopsy instrument (Pro-Mag I 2.5; Medical Device<br />

Technologies Inc, Gainesville, USA). This procedure yields muscle biopsies of 10-15 mg each.<br />

Two muscle biopsies were collected in a test tube on ice with 10 ml of ice cold BIOPS solution<br />

containing 10 mmol/L Ca-EGTA buffer, 0.1 µmol/L free calcium, 20 mmol/L imidazole, 20<br />

mmol/L Taurine, 50 mmol/L K-Mes, 0.5 mmol/L DTT, 6.65 mmol/L MgCl , 5.77 mmol/L ATP,<br />

2<br />

15 mmol/L phosphocreatinine, pH 7.1 and were used to measure mitochondrial respiration<br />

after permeabilisation (see below). Additional muscle biopsies were washed with 0.9% NaCl<br />

fortified with 10mmol/L Na-Hepes to reduce blood contamination, snap-frozen in liquid<br />

nitrogen and stored at -80˚C until further analysis.<br />

Study medication: Prednisolone tablets were purchased from Pfizer (Sollentuna, Sweden)<br />

and matching placebo tablets were obtained from Xendo Drug Development (Groningen, the<br />

Netherlands). The tablets were encapsulated in order to allow the treatment to be blinded,<br />

as described previously [35]. Study medication was taken at 08:00 hours during the 2-week<br />

treatment period except on days 13 and 14, when it was ingested at 06:00 hours. Patients kept<br />

a diary in which the exact time of medication intake during the study was registered.<br />

Laboratory analysis: Plasma glucose concentrations were measured with the glucose oxidase<br />

method using a Biosen C-line plus glucose analyzer (EKF Diagnostics, Barleben/Magedeburg,<br />

Germany). Insulin was determined on an Immulite 2000 system (Diagnostic Products<br />

Corporation, Los Angeles, CA) with a chemiluminescent immunometric assay. Plasma NEFA<br />

concentrations were measured with an enzymatic colorimetric method (NEFA-C test kit;<br />

179<br />

9<br />

Chapter 9


Chapter 9 Glucocorticoids, mitochondrial function & glycosphingolipid metabolism<br />

Wako Chemicals GmbH, Neuss, Germany). [6,6- 2 H 2 ] glucose enrichment was measured with<br />

gas chromatography-mass spectrometry (GC-MS) as described in detail elsewhere [36].<br />

Muscle glycosphingolipids: Ceramide concentrations were measured as described<br />

previously [37]. Briefly, 100 μL of muscle homogenate was extracted with 600 μL of CHCl 3 /<br />

MeOH 1/2 (vol/vol). The extract was centrifuged for 10 min at 13,200 rpm and the pellet<br />

discarded. 460 μL of CHCl /MQ-H O 1/1.3 (vol/vol) was added, mixed, and centrifuged for 3<br />

3 2<br />

min at 13,200 rpm to separate the phases. The lower phase was collected and the upper phase<br />

re-extracted with 400 μL of CHCl . Ceramide and glycosphingolipid content (glucosylceramide<br />

3<br />

(GlCer), lactosylceramide (Lac-cer) and globocylceramide (GB3)) were determined as<br />

previously described [37] with slight modifications. The combined lower phases were dried<br />

under N flow, taken up in 500 μL of freshly prepared 0.1M NaOH in MeOH, and deacetylated<br />

2<br />

in a microwave (SAM-155, CEM corp.) for 60 min. 50 μL of this solution was derivatized with<br />

25 μL o-phtaldehyde (OPA) reagent. The OPA-derivatized lipids were separated by HPLC and<br />

quantified as previously described, using C17 sphinganine as an Internal Standard (Avanti<br />

Polar Lipids, Alabama, USA) [37]. All measurements were done in duplicate from two different<br />

biopsies. Biopsies with a GlCer content above 5 nmol/g, and a GlCer/ceramide ratio above 0.5<br />

were excluded for further analysis, because this was interpreted as blood contamination.<br />

Muscle GM3: The ganglioside GM3 was detected by analysis of the acidic glycolipid fraction<br />

obtained by Bligh-Dyer extraction, as previously described [38]. Gangliosides were desalted<br />

on a disposable SPE C18 column (Bakerbond, Mallinckrodt Baker Inc., Phillipsburg, Nj, USA),<br />

eluted in 1 mL MeOH and concentrated in an Eppendorf concentrator 5301 at 45 °C. The<br />

samples were digested with ceramide glycanase (Recombinant endoglycoceramidase II,<br />

Takara Bio Inc., Otsu, Shiga, japan) at 37 °C for 18 h in order to release the oligosaccharides<br />

from glycosphingolipids. The enzyme was used according to the manufacturer’s instructions.<br />

Oligosaccharides were then fluorescently labeled at their reducing end with 80 μL anthranilic<br />

acid (2-aminobenzoic acid) labeling mixture, at 80 °C, 300 rpm for 45 min. The excess labeling<br />

was removed by gravity flow using Discovery DPA-6S columns and the samples were eluted<br />

with 600 μL of water. Purified 2-AA-labeled oligosaccharides were separated by normalphase<br />

HPLC and quantified as previously described [38], using monosialoganglioside-GM1<br />

(Sigma, St Louis, Mo, USA) as an internal standard for muscles measurements and Gt1b as an<br />

internal standard for plasma measurements.<br />

Mitochondrial function: Mitochondrial respiration was measured ex-vivo with high<br />

resolution respirometry using polarographic oxygen sensors in a two-chamber Oxygraph<br />

(OROBOROS ® Instruments, Innsbruck, Austria) as described before [34]. To evaluate<br />

180


oxidative phosphorylation (OXPHOS) capacity, oxygen consumption was measured during<br />

subsequent addition of different substrates. First malate (2 mmol/L) was added to obtain<br />

state 2 respiration, followed by glutamate (20 mmol/L) as a substrate for complex 1. Then,<br />

an excess of ADP (2 mmol/L) was introduced to measure state 3 respiration. Cytochrome C<br />

(10 mmol/L) was subsequently added to check the integrity of the OXPHOS chain. Thereafter<br />

succinate (10 mmol/L) was added as substrate for complex 2. Additionally, aurovertin was<br />

introduced. Aurovertin inhibits the respiratory chain by binding to ATP synthesis. The oxygen<br />

consumption after the addition of aurovertin (1umol/L) therefore indicates the amount of<br />

leakage under stimulated conditions. Maximal oxygen flux rates were measured with the<br />

chemical uncoupler carbonylcyanide-4-(trifluoromethoxy)-phenylhydrazone (FCCP). FCCP<br />

was titrated (0.25 µmol/L per addition) until no further stimulation of respiration could<br />

be detected. Rotenone (0.1 ummol/L) was subsequently included as inhibitor of complex 1.<br />

Finally, antimycin A (2.5 mmol/L) was added which blocks the respiratory chain at complex 3.<br />

Mitochondrial DNA: Mitochondrial DNA (mtDNA) copy number reflects the tissue<br />

concentration of mitochondria. MtDNA copy number was determined as the ratio of NADH<br />

dehydrogenase subunit 1 (ND1) to lipoprotein lipase (LPL) (mtDNA/nuclear DNA) as<br />

described previously [39].<br />

Statistics: Data are presented as median and range. Absolute changes from baseline (on<br />

treatment value minus pre treatment value) were compared between the groups using the<br />

Kruskal-Wallis test. Non-parametric analysis was chosen due to the relatively small number<br />

of participants and the uneven group sizes. Only in case of a significant finding, prednisolone<br />

7.5 mg and prednisolone 30 mg were compared against placebo by posthoc testing, using<br />

the Mann-Whitney U test. Correction for multiple testing was performed by Bonferroni.<br />

Correlations were made using the non parametric Spearman’s rank test. All statistical<br />

analyses were run on SPSS for Windows version 15.0 (SPSS, Chicago, IL, USA). A P


Chapter 9 Glucocorticoids, mitochondrial function & glycosphingolipid metabolism<br />

Table 1. Subject characteristics at inclusion<br />

182<br />

Placebo Prednisolone<br />

7.5 mg 30 mg P<br />

N 8 12 12<br />

Age (y) 22±3 22±2 22±3 0.740<br />

Weight (kg) 76.9±8.7 74.8±7.2 77.0±5.4 0.250<br />

BMI (kg m -2 ) 22.7±2.1 22.2±1.9 22.4±1.2 0.906<br />

Lean body mass (%) 78.6±7.9 82.1±3.9 80.7±2.5 0.731<br />

Fat mass (%) 16.9±4.7 15.9±2.8 14.8±3.5 0.531<br />

Fasting plasma glucose (mmol/l) 5.1±0.3 5.0±0.2 5.0±0.4 0.899<br />

2-hr glucose OGTT (mmol/l) 4.2±0.8 4.7±0.9 4.2±1.1 0.407<br />

VO2 max (ml.kg.min) 49.9 (44.6-58.2) 53.8 (45.0-60.3) 48.6 (45.4-55.2) 0.159<br />

Data are mean ± SD or median (interquartile range). Significance was tested by Kruskal-Wallis. No<br />

differences were observed between the groups at baseline [5].<br />

Table 2. Muscle glycosphingolipid concentrations before and during two-week treatment with<br />

placebo, prednisolone 7.5 mg or prednisolone 30 mg.<br />

Ceramide<br />

pmol/mg musle<br />

GlCeramide<br />

pmol/mg musle<br />

Lac-Ceramide<br />

pmol/mg musle<br />

GB3<br />

pmol/mg musle<br />

GM3<br />

pmol/mg musle<br />

Pre-treatment On-treatment P1 P2 P3<br />

PLB 20.3 (14.4-37.9) 21.6 (14.0-33.7)<br />

7.5 19.4 (15.0-27.0) 19.6 (6.3-26.3) 0.954 NA NA<br />

30 19.4 (13.5-35.9) 21.9 (17.4-25.7)<br />

PLB 1.03 (0.47-4.34) 1.21 (0.29-1.91)<br />

7.5 1.18 (0.53-2.67) 1.12 (0.54-3.68) 0.558 NA NA<br />

30 1.58 (0.53-2.57) 1.44 (0.87-3.61)<br />

PLB 12.9 (10.2-30.5) 10.6 (5.0-12.9)<br />

7.5 12.6 (7.2-23.6) 12.2 (3.2-16.0) 0.038 0.155 0.013<br />

30 12.5 (6.2-24.0) 17.3 (10.4-22.7)<br />

PLB 5.1 (2.4-13.5) 5.3 (1.6-10.4)<br />

7.5 4.9 (2.2-14.0) 4.2 (3.4-8.0)<br />

0.899 NA NA<br />

30 6.1 (2.2-18.2) 5.9 (3.8-11.8)<br />

PLB 9.3 (5.8-13.3) 9.2 (6.2-16.8)<br />

7.5 10.8 (3.6-29.9) 11.8 (3.3-30.4) 0.310 NA NA<br />

30 10.4 (6.8-53.2) 12.5 (3.6-55.0)<br />

Data are median (range). Bonferroni was applied to correct for multiple testing. Between-group changes<br />

from baseline were tested by Kruskal-Wallis (P1). P2: Placebo vs. prednisolone 7.5 mg and P3: placebo<br />

vs. prednisolone 30 mg (post hoc testing by Mann-Whitney U in the case of a significant finding with<br />

Kruskal-Wallis). 7.5: prednisolone 7.5 mg; 30: prednisolone 30 mg; GB3: globocylceramide; GlCeramide:<br />

glucosylceramide; Lac-Ceramide: lactosylceramide; PLB: placebo.


Table 3. Mitochondrial respiration rates corrected for mitochondrial density before and during<br />

two-week treatment with placebo, prednisolone 7.5 mg or prednisolone 30 mg.<br />

Malate<br />

pmol/mg.sec.mtDNA<br />

Glutamate<br />

pmol/mg.sec.mtDNA<br />

ADP<br />

pmol/mg.sec.mtDNA<br />

Cytochrome C<br />

pmol/mg.sec.mtDNA<br />

Succinate<br />

pmol/mg.sec.mtDNA<br />

Aurovertin<br />

pmol/mg.sec.mtDNA<br />

FCCP<br />

pmol/mg.sec.mtDNA<br />

Rotenone<br />

pmol/mg.sec.mtDNA<br />

Antimycin A<br />

pmol/mg.sec.mtDNA<br />

Pre-treatment On-treatment P<br />

PLB 1.6 (1.0-3.2) 1.4 (0.6-4.2)<br />

7.5 1.4 (0.6-3.5) 1.3 (0.8-4.2)<br />

NS<br />

30 1.6 (1.0-3.7) 2.2 (1.3-5.8)<br />

PLB 2.1 (0.8-3.3) 1.8 (0.8-4.4)<br />

7.5 2.2 (1.0-3.5) 2.6 (1.1-5.9)<br />

NS<br />

30 2.2 (1.2-3.9) 2.4 (1.7-7.0)<br />

PLB 23.0 (8.3-25.5) 15.9 (6.6-25.2)<br />

7.5 17.6 (9.4-31.0) 17.1 (10.4-57.4)<br />

NS<br />

30 21.8 (12.8-38.0) 20.0 (13.4-50.5)<br />

PLB 22.2 (8.3-29.4) 16.1 (7.6-22.6)<br />

7.5 20.9 (10.0-30.0) 18.4 (10.5-58.2)<br />

NS<br />

30 21.9 (12.5-38.5) 19.2 (14.5-33.8)<br />

PLB 32.7 (12.8-41.4) 28.7 (14.8-35.2)<br />

7.5 30.7 (15.1-40.0) 27.4 (15.4-79.6)<br />

NS<br />

30 31.4 (17.7-43.6) 28.4 (21.7-54.3)<br />

PLB 7.7 (2.9-9.7) 6.8 (3.4-11.6)<br />

7.5 7.3 (3.4-12.3) 7.6 (3.6-21.8)<br />

NS<br />

30 7.6 (4.2-9.8) 8.7 (6.1-13.9)<br />

PLB 57.0 (17.7-61.3) 39.9 (17.6-74.0)<br />

7.5 44.2 (22.2-82.4) 38.8 (22.0-124.8)<br />

NS<br />

30 51.3 (30.6-66.9) 43.8 (19.5-130.0)<br />

PLB 26.3 (10.0-30.7) 22.6 (11.1-38.1)<br />

7.5 20.3 (11.2-40.0) 20.3 (12.1-71.3)<br />

NS<br />

30 25.0 (14.2-45.0) 23.0 (10.7-62.0)<br />

PLB 1.8 (0.7-3.1) 1.6 (0.6-4.9)<br />

7.5 1.8 (0.7-6.3) 1.4 (0.4-4.0)<br />

NS<br />

30 1.8 (0.4-2.9) 1.9 (0.8-4.0)<br />

Data are median (range). Between-group changes from baseline were tested by Kruskal-Wallis. No<br />

significant differences were observed between the groups. Abbreviations 7.5: prednisolone 7.5 mg; 30:<br />

prednisolone 30 mg; FCCP: carbonylcyanide-4-(trifluoromethoxy)-phenylhydrazone. PLB: placebo.<br />

Peripheral insulin sensitivity: As described previously [5], peripheral glucose uptake was<br />

non-significantly reduced by prednisolone 7.5 mg 5 (Rd= 61.05 [47.85-76.8] vs. 60 .19 [43.92-<br />

7.09] mmol/kg.min ; P= 0.090), but decreased significantly after treatment with prednisolone<br />

30 mg by 34±6% (Rd = 67.35 [50.87-93.53] vs. 42.42 [28.83-60.40]; P < 0.001).<br />

183<br />

9<br />

Chapter 9


Chapter 9 Glucocorticoids, mitochondrial function & glycosphingolipid metabolism<br />

Muscle glycosphingolipids and GM3: As compared to placebo, muscle ceramide<br />

concentrations were not altered by prednisolone treatment (Table 2). Concentrations of the<br />

sphingolipids GlCer and GB3 were also not affected by prednisolone treatment (Table 2). Due<br />

to suspected blood contamination, the GlCer data from two participants in the prednisolone<br />

7.5 mg group and one participant in the prednisolone 30 mg group were excluded from<br />

analysis. Muscle concentrations of Lac-Cer showed a significance increase during high-dose<br />

prednisolone treatment (Table 2). There was no relation between the change in Lac-Cer levels<br />

and treatment-induced changes in insulin sensitivity. Muscle GM3 concentrations were not<br />

significantly altered from baseline by prednisolone treatment (Table 2).<br />

Mitochondrial function: Mitochondrial respiration rates, corrected for mitochondrial<br />

density, were not altered by prednisolone treatment (Table 3). Uncorrected mitochondrial<br />

respiration rates as well as mitochondrial density were also not changed by prednisolone<br />

treatment (data not shown).<br />

DISCUSSIoN<br />

In the present study, we demonstrate that GC-induced insulin resistance cannot be explained<br />

by an increase in muscle ceramide concentrations or the downstream ganglioside GM3. In<br />

addition, muscle mitochondrial function was not changed during two-week treatment with<br />

low or high-dose prednisolone, while insulin sensitivity was dose-dependently decreased.<br />

This suggests that other mechanisms than mitochondrial dysfunction and accumulation of<br />

glycolipids and GM3 are responsible for the induction of GC-induced insulin resistance in<br />

young healthy men.<br />

Both decreased affinity of the insulin receptor and impaired insulin signaling may importantly<br />

contribute to reduced insulin sensitivity during GC treatment [40, 41]. Furthermore, GCs<br />

may also directly inhibit the glucose transport system, by affecting GLUT4 subcellular<br />

trafficking [12]. In this respect, we were interested to assess the effects of GC treatment on<br />

muscle ceramide since this intermediate in lipid metabolism was shown to inhibit insulin<br />

signaling by impairing PKB/Akt activation [22-24]. But although ceramide was shown to play<br />

a crucial role in GC-induced insulin resistance in rodents [25], we could not detect an effect<br />

of GC treatment on muscle ceramide levels in humans. In addition, the more downstream<br />

glycolipids GlCer and GB were also not affected by treatment with study medication. We did<br />

observe a significant increase in the glycolipid Lac-Cer during prednisolone 30 mg treatment<br />

as compared to placebo, however, as the increase in Lac-Cer was not significant within the<br />

184


prednisolone 30 mg group, and the change in Lac-Cer concentrations did not correlate with<br />

changes in Rd, this does not suggest a causal relation with the induction of insulin resistance.<br />

Finally, the ganglioside GM3, which is known to be involved in insulin sensitivity [26], was not<br />

changed by prednisolone treatment.<br />

Thus, we conclude that changes in ceramide, other glycosphingolipids and the ganglioside<br />

GM3 do not seem to play an important role in the impairment of in insulin sensitivity induced<br />

by short-term GC treatment in healthy men. We cannot rule out that other lipids may have<br />

negatively affected insulin sensitivity. Diacylglycerol and several unsaturated fatty acids that<br />

were not measured in the present study due to limited amount of available skeletal muscle<br />

tissue, have been shown in previous studies in obese subjects and patients with T2DM to be<br />

associated with impaired insulin sensitivity [42].<br />

In addition to alterations in myocellular glycoshingolipids, we investigated a potential role for<br />

muscle mitochondrial dysfunction in GC-induced insulin resistance. Decreased mitochondrial<br />

capacity has been related to reduced metabolic flexibility, and may play an important role in<br />

the pathogenesis of lipid accumulation and insulin resistance [32, 42]. Interestingly, chronic<br />

GC treatment is well known to induce myopathy. Patients treated with GCs during 1 year<br />

for different indications were shown to have increased lactate production during exercise<br />

and decreased muscle strength. In addition, analysis of individual mitochondrial respiratory<br />

chain complex (I–IV) activities in these patients showed that complex 1 was significantly<br />

decreased and production of reactive oxygen species was significantly increased, indication<br />

mitochondrial dysfunction [33]. In our study, however, no differences in muscle mitochondrial<br />

respiration rates were observed during prednisolone treatment. Our finding of preserved<br />

mitochondrial capacity despite the occurrence of insulin resistance is in agreement with a<br />

study conducted by Short et al who showed that 6 days treatment with 0.5 mg/kg prednisolone<br />

in healthy men did not affect oxidative enzyme activity or mitochondrial ATP production [43].<br />

The difference results of these studies may be explained by several factors, including the<br />

treatment duration (1 year vs. 1-2 weeks) and the population studied (healthy volunteers<br />

vs. patients). In addition, in the clinic, it is common to administer GCs to the patients with<br />

most active disease, and therefore the association between GC treatment and skeletal muscle<br />

mitochondrial dysfunction may have been caused by disease activity (i.e. bias by indication).<br />

Moreover, no correction for mitochondrial density was done in the chronic study in GC-treated<br />

patients [33]. Furthermore, the good exercise capacity of our participants at baseline may<br />

have contributed to the absence of prednisolone-induced changes in mitochondrial function.<br />

Finally, it should be mentioned that an additional difficulty is that currently no gold-standard<br />

measure exists to assess mitochondrial function. Ex vivo measurement of mitochondrial<br />

185<br />

9<br />

Chapter 9


Chapter 9 Glucocorticoids, mitochondrial function & glycosphingolipid metabolism<br />

function has the advantage to measure intrinsic mitochondrial capacity and function of the<br />

different oxidative phosphorylation complexes, but cytosolic factors that might influence<br />

oxidative phosphorylation are not accounted for.<br />

Several other mechanisms have been suggested that may contribute to GC-induced insulin<br />

resistance. GCs were shown to induce the acute phase response in skeletal muscle tissue<br />

which could reduce insulin sensitivity [44]. In addition, changes in amino acid metabolism<br />

are associated with impaired insulin sensitivity [45]. Since GC treatment induces skeletal<br />

muscle proteolysis [5], this could be a contributing factor, especially during prolonged GC<br />

treatment. Moreover, the endoplasmic reticulum (ER) has been identified as a major regulator<br />

of metabolic homeostasis and inflammatory responses and ER stress was shown to have<br />

unbeneficial effects on insulin sensitivity [46]. Whereas GCs were recently shown to induce<br />

ER stress in beta cells [47], it is uncertain whether GCs may also alter ER function in skeletal<br />

muscle tissue. Finally, GCs may impair insulin sensitivity by impairing the delivery of insulin<br />

and glucose to skeletal muscle by reducing capillary recruitment through the induction of<br />

vascular insulin resistance [14].<br />

In conclusion, short-term treatment with GCs induces insulin resistance in lean men. This<br />

reduction in insulin sensitivity was not related to a change in muscle ceramide, GM3 or<br />

mitochondrial function. Which specific mechanisms are involved in the induction of insulin<br />

resistance by GC treatment remains to be elucidated.<br />

Acknowledgements<br />

The authors thank M. van der Plas for performing the VO2 max measurements. Furthermore,<br />

P. Schrauwen and E. Kornips are acknowledged for the measurement of MT-DNA.<br />

Funding<br />

This paper was written within the framework of project T1-106 of the Dutch Top Institute<br />

<strong>Pharma</strong>.<br />

186


REFERENCES<br />

1. Schacke H, Docke WD, Asadullah K. Mechanisms involved in the side effects of glucocorticoids.<br />

<strong>Pharma</strong>col Ther. 2002;96(1):23-43.<br />

2. van Raalte DH, Ouwens DM, Diamant M. Novel insights into glucocorticoid-mediated diabetogenic<br />

effects: towards expansion of therapeutic options? Eur j Clin Invest. 2009;39(2):81-93.<br />

3. Khaleeli AA, Edwards RH, Gohil K, McPhail G, Rennie Mj, Round j, Ross Ej. Corticosteroid myopathy:<br />

a clinical and pathological study. Clin Endocrinol (Oxf). 1983;18(2):155-66.<br />

4. Taskinen MR, Kuusi T, Yki-jarvinen H, Nikkila EA. Short-term effects of prednisone on serum lipids<br />

and high density lipoprotein subfractions in normolipidemic healthy men. j Clin Endocrinol Metab.<br />

1988;67(2):291-9.<br />

5. van Raalte DH, Brands M, van der Zijl Nj, Muskiet MH, Pouwels Pj, Ackermans MT, Sauerwein HP,<br />

Serlie Mj, Diamant M. Low-dose glucocorticoid treatment affects multiple aspects of intermediary<br />

metabolism in healthy humans: a randomised controlled trial. Diabetologia. 2011;54(8):2103-12.<br />

6. Gulliford MC, Charlton j, Latinovic R. Risk of diabetes associated with prescribed glucocorticoids in<br />

a large population. Diabetes Care. 2006;29(12):2728-9.<br />

7. Gurwitz jH, Bohn RL, Glynn Rj, Monane M, Mogun H, Avorn j. Glucocorticoids and the risk for<br />

initiation of hypoglycemic therapy. Arch Intern Med. 1994;154(1):97-101.<br />

8. Clore jN, Thurby-Hay L. Glucocorticoid-induced hyperglycemia. Endocr Pract. 2009;15(5):469-74.<br />

9. Ruzzin j, Wagman AS, jensen j. Glucocorticoid-induced insulin resistance in skeletal muscles: defects<br />

in insulin signalling and the effects of a selective glycogen synthase kinase-3 inhibitor. Diabetologia.<br />

2005;48(10):2119-30.<br />

10. Long W, Barrett Ej, Wei L, Liu Z. Adrenalectomy enhances the insulin sensitivity of muscle protein<br />

synthesis. Am j Physiol Endocrinol Metab. 2003;284(1):E102-9.<br />

11. Ewart HS, Somwar R, Klip A. Dexamethasone stimulates the expression of GLUT1 and GLUT4<br />

proteins via different signalling pathways in L6 skeletal muscle cells. FEBS Lett. 1998;421(2):120-4.<br />

12. Weinstein SP, Wilson CM, Pritsker A, Cushman SW. Dexamethasone inhibits insulin-stimulated<br />

recruitment of GLUT4 to the cell surface in rat skeletal muscle. Metabolism. 1998;47(1):3-6.<br />

13. Saad Mj, Folli F, Kahn jA, Kahn CR. Modulation of insulin receptor, insulin receptor substrate-1,<br />

and phosphatidylinositol 3-kinase in liver and muscle of dexamethasone-treated rats. j Clin Invest.<br />

1993;92(4):2065-72.<br />

14. van Raalte DH, Diamant M, Ouwens DM, Ijzerman RG, Linssen MML, Guigas B, Eringa EC, Serne EH.<br />

Glucocorticoid treatment impairs microvascular function in healthy males, in association with its<br />

adverse effects on blood pressure and glucose metabolism. Submitted.<br />

15. Henriksen jE, Alford F, Vaag A, Handberg A, Beck-Nielsen H. Intracellular skeletal muscle glucose<br />

metabolism is differentially altered by dexamethasone treatment of normoglycemic relatives of type<br />

2 diabetic patients. Metabolism. 1999;48(9):1128-35.<br />

187<br />

9<br />

Chapter 9


Chapter 9 Glucocorticoids, mitochondrial function & glycosphingolipid metabolism<br />

16. Kewalramani G, Bilan Pj, Klip A. Muscle insulin resistance: assault by lipids, cytokines and local<br />

macrophages. Curr Opin Clin Nutr Metab Care. 2010;13(4):382-90.<br />

17. Haus jM, Kashyap SR, Kasumov T, Zhang R, Kelly KR, Defronzo RA, Kirwan jP. Plasma ceramides are<br />

elevated in obese subjects with type 2 diabetes and correlate with the severity of insulin resistance.<br />

Diabetes. 2009;58(2):337-43.<br />

18. Skovbro M, Baranowski M, Skov-jensen C, Flint A, Dela F, Gorski j, Helge jW. Human skeletal muscle<br />

ceramide content is not a major factor in muscle insulin sensitivity. Diabetologia. 2008;51(7):1253-<br />

60.<br />

19. Adams jM, 2nd, Pratipanawatr T, Berria R, Wang E, DeFronzo RA, Sullards MC, Mandarino Lj.<br />

Ceramide content is increased in skeletal muscle from obese insulin-resistant humans. Diabetes.<br />

2004;53(1):25-31.<br />

20. Straczkowski M, Kowalska I, Baranowski M, Nikolajuk A, Otziomek E, Zabielski P, Adamska A,<br />

Blachnio A, Gorski j, Gorska M. Increased skeletal muscle ceramide level in men at risk of developing<br />

type 2 diabetes. Diabetologia. 2007;50(11):2366-73.<br />

21. Coen PM, Dube jj, Amati F, Stefanovic-Racic M, Ferrell RE, Toledo FG, Goodpaster BH. Insulin<br />

resistance is associated with higher intramyocellular triglycerides in type I but not type II myocytes<br />

concomitant with higher ceramide content. Diabetes. 2010;59(1):80-8.<br />

22. Summers SA. Sphingolipids and insulin resistance: the five Ws. Curr Opin Lipidol. 2010;21(2):128-<br />

35.<br />

23. Holland WL, Summers SA. Sphingolipids, insulin resistance, and metabolic disease: new insights<br />

from in vivo manipulation of sphingolipid metabolism. Endocr Rev. 2008;29(4):381-402.<br />

24. Bikman BT, Summers SA. Ceramides as modulators of cellular and whole-body metabolism. j Clin<br />

Invest. 2011;121(11):4222-30.<br />

25. Holland WL, Brozinick jT, Wang LP, Hawkins ED, Sargent KM, Liu Y, Narra K, Hoehn KL, Knotts<br />

TA, Siesky A, Nelson DH, Karathanasis SK, Fontenot GK, Birnbaum Mj, Summers SA. Inhibition<br />

of ceramide synthesis ameliorates glucocorticoid-, saturated-fat-, and obesity-induced insulin<br />

resistance. Cell Metab. 2007;5(3):167-79.<br />

26. Yamashita T, Hashiramoto A, Haluzik M, Mizukami H, Beck S, Norton A, Kono M, Tsuji S, Daniotti jL,<br />

Werth N, Sandhoff R, Sandhoff K, Proia RL. Enhanced insulin sensitivity in mice lacking ganglioside<br />

GM3. Proc Natl Acad Sci U S A. 2003;100(6):3445-9.<br />

27. Nojiri H, Stroud M, Hakomori S. A specific type of ganglioside as a modulator of insulin-dependent<br />

cell growth and insulin receptor tyrosine kinase activity. Possible association of ganglioside-induced<br />

inhibition of insulin receptor function and monocytic differentiation induction in HL-60 cells. j Biol<br />

Chem. 1991;266(7):4531-7.<br />

28. Kabayama K, Sato T, Kitamura F, Uemura S, Kang BW, Igarashi Y, Inokuchi j. TNFalpha-induced<br />

insulin resistance in adipocytes as a membrane microdomain disorder: involvement of ganglioside<br />

GM3. Glycobiology. 2005;15(1):21-9.<br />

188


29. Kabayama K, Sato T, Saito K, Loberto N, Prinetti A, Sonnino S, Kinjo M, Igarashi Y, Inokuchi j.<br />

Dissociation of the insulin receptor and caveolin-1 complex by ganglioside GM3 in the state of<br />

insulin resistance. Proc Natl Acad Sci U S A. 2007;104(34):13678-83.<br />

30. Inokuchi j. Insulin resistance as a membrane microdomain disorder. Yakugaku Zasshi.<br />

2007;127(4):579-86.<br />

31. Cohen AW, Combs TP, Scherer PE, Lisanti MP. Role of caveolin and caveolae in insulin signaling and<br />

diabetes. Am j Physiol Endocrinol Metab. 2003;285(6):E1151-60.<br />

32. Roden M. Muscle triglycerides and mitochondrial function: possible mechanisms for the development<br />

of type 2 diabetes. Int j Obes (Lond). 2005;29 Suppl 2:S111-5.<br />

33. Mitsui T, Azuma H, Nagasawa M, Iuchi T, Akaike M, Odomi M, Matsumoto T. Chronic corticosteroid<br />

administration causes mitochondrial dysfunction in skeletal muscle. j Neurol. 2002;249(8):1004-9.<br />

34. Brands M, Hoeks j, Sauerwein HP, Ackermans MT, Ouwens M, Lammers NM, van der Plas MN,<br />

Schrauwen P, Groen AK, Serlie Mj. Short-term increase of plasma free fatty acids does not interfere<br />

with intrinsic mitochondrial function in healthy young men. Metabolism. 2011;60(10):1398-405.<br />

35. van Raalte DH, Nofrate V, Bunck MC, van Iersel T, Elassaiss Schaap j, Nassander UK, Heine Rj, Mari<br />

A, Dokter WH, Diamant M. Acute and 2-week exposure to prednisolone impair different aspects of<br />

beta-cell function in healthy men. Eur j Endocrinol. 2010;162(4):729-35.<br />

36. Ackermans MT, Pereira Arias AM, Bisschop PH, Endert E, Sauerwein HP, Romijn JA. The quantification<br />

of gluconeogenesis in healthy men by (2)H2O and [2-(13)C]glycerol yields different results: rates of<br />

gluconeogenesis in healthy men measured with (2)H2O are higher than those measured with [2-<br />

(13)C]glycerol. j Clin Endocrinol Metab. 2001;86(5):2220-6.<br />

37. Groener jE, Poorthuis Bj, Kuiper S, Helmond MT, Hollak CE, Aerts jM. HPLC for simultaneous<br />

quantification of total ceramide, glucosylceramide, and ceramide trihexoside concentrations in<br />

plasma. Clin Chem. 2007;53(4):742-7.<br />

38. Ghauharali-van der Vlugt K, Langeveld M, Poppema A, Kuiper S, Hollak CE, Aerts jM, Groener jE.<br />

Prominent increase in plasma ganglioside GM3 is associated with clinical manifestations of type I<br />

Gaucher disease. Clin Chim Acta. 2008;389(1-2):109-13.<br />

39. Phielix E, Schrauwen-Hinderling VB, Mensink M, Lenaers E, Meex R, Hoeks j, Kooi ME, Moonen-<br />

Kornips E, Sels jP, Hesselink MK, Schrauwen P. Lower intrinsic ADP-stimulated mitochondrial<br />

respiration underlies in vivo mitochondrial dysfunction in muscle of male type 2 diabetic patients.<br />

Diabetes. 2008;57(11):2943-9.<br />

40. Grunfeld C, Baird K, Van Obberghen E, Kahn CR. Glucocorticoid-induced insulin resistance in vitro:<br />

evidence for both receptor and postreceptor defects. Endocrinology. 1981;109(5):1723-30.<br />

41. McMahon M, Gerich j, Rizza R. Effects of glucocorticoids on carbohydrate metabolism. Diabetes<br />

Metab Rev. 1988;4(1):17-30.<br />

42. Samuel VT, Petersen KF, Shulman GI. Lipid-induced insulin resistance: unravelling the mechanism.<br />

Lancet. 2010;375(9733):2267-77.<br />

189<br />

9<br />

Chapter 9


Chapter 9 Glucocorticoids, mitochondrial function & glycosphingolipid metabolism<br />

43. Short KR, Bigelow ML, Nair KS. Short-term prednisone use antagonizes insulin’s anabolic effect on<br />

muscle protein and glucose metabolism in young healthy people. Am j Physiol Endocrinol Metab.<br />

2009;297(6):E1260-8.<br />

44. Viguerie N, Picard F, Hul G, Roussel B, Barbe P, Iacovoni jS, Valle C, Langin D, Saris WH. Multiple<br />

effects of a short-term dexamethasone treatment in human skeletal muscle and adipose tissue.<br />

Physiol Genomics. 2012;44(2):141-51.<br />

45. Krebs M, Krssak M, Bernroider E, Anderwald C, Brehm A, Meyerspeer M, Nowotny P, Roth E,<br />

Waldhausl W, Roden M. Mechanism of amino acid-induced skeletal muscle insulin resistance in<br />

humans. Diabetes. 2002;51(3):599-605.<br />

46. Hotamisligil GS. Endoplasmic reticulum stress and the inflammatory basis of metabolic disease. Cell.<br />

2010;140(6):900-17.<br />

47. Linssen MM, van Raalte DH, Toonen Ej, Alkema W, van der Zon GC, Dokter WH, Diamant M, Guigas B,<br />

Ouwens DM. Prednisolone-induced beta cell dysfunction is associated with impaired endoplasmic<br />

reticulum homeostasis in INS-1E cells. Cell Signal. 2011;23(11):1708-15.<br />

190


Chapter 10<br />

Glucocorticoid Treatment Impairs<br />

Microvascular Function in Healthy Males,<br />

in Association with its Adverse Effects on<br />

Glucose Metabolism and Blood Pressure<br />

D.H. van Raalte, M. Diamant, D.M. Ouwens, R.G. IJzerman, M.M.L.<br />

Linssen, B. Guigas, E.C. Eringa, E.H. Serné<br />

Manuscript in Preparation


Chapter 10 Glucocorticoids and microvascular function<br />

ABSTRACT<br />

Glucocorticoids (GCs) are anti-inflammatory agents but also induce side effects, including<br />

insulin resistance, glucose intolerance and hypertension. In this study, we investigated the<br />

role of microvascular function in the development of these adverse effects. In a randomized,<br />

placebo-controlled, double-blind, dose-response intervention study, 32 healthy males (age:<br />

21±2 years; BMI: 21.9±1.7 kg/m2 ) were allocated to prednisolone 30 mg once daily (n=12),<br />

prednisolone 7.5 mg once daily (n=12), or placebo (n=8) for two weeks. Compared to placebo,<br />

prednisolone treatment dose-dependently decreased capillary recruitment. Additionally,<br />

prednisolone treatment dose-dependently impaired insulin sensitivity and glucose<br />

tolerance, but only prednisolone 30 mg increased systolic blood pressure. Prednisoloneinduced<br />

changes in insulin-stimulated capillary recruitment were associated with insulin<br />

sensitivity (r=+0.76), postprandial glucose levels (r=-0.52) and systolic blood pressure<br />

(r=-0.62). To further investigate underlying mechanisms, male Wistar rats were treated<br />

with prednisolone or saline for 7 days. Prednisolone treatment attenuated the vasodilatory<br />

actions of insulin in muscle resistance arteries by impairing insulin signaling through the<br />

Akt pathway. We conclude that prednisolone dose-dependently impaired insulin-stimulated<br />

capillary recruitment by inducing vascular insulin resistance through direct interference<br />

with vascular insulin signaling. We propose that GC-induced impairments of microvascular<br />

function may contribute to the adverse effects of GC treatment on glucose metabolism.<br />

Clinical Trial Registration Number: ISRTCN 78149983<br />

194


Glucocorticoids (GCs) represent the most important and frequently used class of antiinflammatory<br />

drugs, but their use is hampered by an increased risk of serious side effects,<br />

particularly hypertension, insulin resistance, reduced glucose tolerance and overt diabetes<br />

in susceptible individuals [1-5]. Although the exact mechanisms underlying these adverse<br />

effects remain to be identified, GC-induced insulin resistance may underlie part of these side<br />

effects [6]. Insulin resistance is characterized by the diminished ability of insulin to initiate<br />

intracellular signaling, primarily in liver, adipose tissue and skeletal muscle [7]. Impaired<br />

insulin signaling in these tissues results in reduced glucose uptake, insufficient suppression of<br />

hepatic glucose output, compensatory hyperinsulinemia and dyslipidemia, which collectively<br />

constitute the so-called metabolic syndrome [7].<br />

Insulin action is not restricted to metabolically active tissues. Recently, it has become clear<br />

that vascular tissue, and particularly endothelial cells, represents an important physiological<br />

target for insulin and a significant regulator of overall insulin-stimulated glucose uptake [8-<br />

10]. Insulin promotes its own access to muscle interstitial space by increasing blood flow and<br />

by recruiting capillaries to expand the endothelial transporting surface available for nutrient<br />

exchange. In addition, the local actions of insulin on (micro)vascular tissue may be of relevance<br />

to the pathogenesis of hypertension, an important feature of the metabolic syndrome [8,<br />

10-12]. Insulin exerts a vasodilatory action by promoting nitric oxide (NO) release from<br />

the endothelial cells. This involves the phosphorylation of endothelial NO synthase (eNOS),<br />

which on its turn is mediated by the phosphatidylinositol 3-kinase (PI3K)/Akt pathway [13].<br />

Concomitantly, insulin activates extracellular signal-regulated kinase (ERK)1/2 signaling<br />

in endothelial cells, which results in enhanced expression of the vasoconstrictor regulator<br />

endothelin-1 [10, 13]. In healthy subjects, the vasodilatory signal prevails, but if activation<br />

of the PI3K/Akt/eNOS pathway is impaired, as observed in insulin resistant states, insulinregulated<br />

vasodilatation becomes compromised. In this manner, vascular insulin resistance<br />

may contribute to the development of hypertension and impaired whole-body insulinstimulated<br />

glucose uptake [14].<br />

It is presently unclear whether vascular insulin resistance could be involved in the unfavorable<br />

effects of GCs on glucose metabolism and blood pressure. Thus, in the present study, we<br />

investigated whether the metabolic effects of the GC prednisolone, given at both a clinically<br />

relevant low- and high dosage for 2 weeks, would concomitantly induce vascular insulin<br />

resistance in healthy humans. Subsequently, we assessed associations among prednisoloneinduced<br />

vascular insulin resistance and changes in blood pressure, whole-body insulinmediated<br />

glucose uptake and glucose tolerance. In order to detail underlying mechanisms,<br />

vasoreactivity and Akt-signaling and MAPK pathway signaling in the endothelium were<br />

examined in cremaster arterioles of Wistar rats treated with prednisolone.<br />

195<br />

Chapter 10<br />

10


Chapter 10 Glucocorticoids and microvascular function<br />

METHoDS<br />

Clinical study<br />

Participants: Thirty-two healthy Caucasian subjects were recruited by local advertisement.<br />

Inclusion criteria were: age between 18-35 years, body mass index (BMI) between 20.0-<br />

25.0 kg/m2 , good physical health (determined by medical history, physical examination and<br />

screening blood tests) and normoglycemia as defined by fasting plasma glucose (FPG) < 5.6<br />

mmol/L and 2-hour glucose < 7.8 mmol/L following a 75 g oral glucose tolerance test (OGTT),<br />

performed at screening visit. Exclusion criteria were any previous or current illness, use of<br />

any medication, first-degree relative with type 2 diabetes, smoking, shift work, a history of<br />

GC use, excessive sports activities (i.e. more often than two times/week) and recent changes<br />

in weight or physical activity. The study was approved by an independent ethics committee<br />

and the study was conducted in accordance with the Declaration of Helsinki. All participants<br />

provided written informed consent before participation.<br />

Study design: The study was a randomized, placebo-controlled, double blind, doseresponse<br />

intervention study. Following assessment of eligibility and baseline measurements,<br />

participants were randomized to receive either prednisolone 30 mg once daily (n=12),<br />

prednisolone 7.5 mg once daily (n=12), or placebo treatment (n=8) for a period of 14 days using<br />

block randomization, as carried out by the Department of Experimental <strong>Pharma</strong>cology of the<br />

VU University Medical Center. Prior to treatment and on day 13 of treatment, microvascular<br />

function was measured by capillary microscopy in the fasted state and during insulin infusion.<br />

Insulin sensitivity was determined by hyperinsulinemic-euglycemic clamp. Prior to treatment<br />

and on day 14 of treatment, a standardized consecutive meal challenge test was performed<br />

to assess glucose tolerance. All measurements were conducted in a temperature-controlled<br />

room (23.4±0.4°C) starting at 0730 AM, following a 12-h overnight fast with the subjects in<br />

the supine position. Subjects refrained from drinking alcohol for a period of 24h before the<br />

study day and did not perform strenuous exercise for a period of 48h before each study day.<br />

Baseline measurements were obtained following 30 min of rest and acclimatization. During<br />

all visits, including a follow-up visit at day 7 of treatment, safety and tolerability were assessed<br />

(Supplementary Figure 1). A patient flow diagram is shown in Supplementary Figure 2.<br />

Capillary microscopy: Nail fold capillary studies were performed in the fasted state (T=-<br />

120 min) and during hyperinsulinemia (T=120 min) as described previously [15] by a single<br />

experienced investigator (Figure 1). Baseline capillary density was defined as the number<br />

of continuously erythrocyte-perfused capillaries per square millimeter of nail fold skin.<br />

Postocclusive reactive hyperemia after 4 min of arterial occlusion was used to assess functional<br />

196


capillary recruitment. Capillary recruitment was calculated by dividing the increase in<br />

perfused capillary density during postocclusive reactive hyperemia by the baseline perfused<br />

capillary density. In addition, venous congestion to expose a maximal number of capillaries<br />

was done. All procedures were performed twice, and the mean of both measurements was<br />

used for analyses. The day-to-day coefficient of variation of functional capillary recruitment<br />

was 15.9±8.0 % as determined in 10 healthy individuals on 2 separate days. Skin temperature<br />

was monitored during the tests. One subject in the prednisolone 7.5 arm could not be analyzed<br />

due to insufficient quality of microscopic images during treatment with study medication and<br />

was left out of the analyses.<br />

Blood pressure: Systolic and diastolic blood pressure were manually measured (Welch Allyn,<br />

Delft, Netherlands) by a single experienced investigator following the 30 min acclimatization<br />

period. The average of three consecutive blood pressure measurements with 5-min interval<br />

was used for further analyses.<br />

Figure 1. Design of the experimental protocol. A two-hour hyperinsulinemic-euglycemic clamp<br />

was performed for 180 min. Microcirculation: nailfold capillary microscopy; BP: blood pressure<br />

measurements; biopsy: percutaneous skeletal muscle biopsy. Arrows indicate measurement of plasma<br />

insulin levels.<br />

Hyperinsulinemic-euglycemic clamp: Whole-body insulin sensitivity was assessed by<br />

hyperinsulinemic-euglycemic clamp method as described previously [16]. Briefly, a primed,<br />

continuous (40 mU/m.min) insulin infusion (Actrapid 100 IU/mL; Novo Nordisk, Bagsvaerd,<br />

Denmark) was given for 120 min; plasma glucose was kept at 5 mmol/l by adjusting the<br />

rate of a 20% glucose infusion based on plasma glucose measurements performed at 5-min<br />

intervals (Figure 1). Whole-body insulin sensitivity was quantified by the M-value (mg/<br />

kg.min), calculated between min 90-120 during steady-state insulin concentrations as<br />

described previously [16].<br />

197<br />

Chapter 10<br />

10


Chapter 10 Glucocorticoids and microvascular function<br />

Standardized meal test: Glucose tolerance was assessed following two consecutive identical<br />

meals that were served as breakfast at 09.00 AM and as lunch at 1.00 PM. The meals contained<br />

905 kcal (50 g fat, 75 g carbohydrates, 35 g protein) each. Samples for determination of<br />

glucose were obtained at 30 min intervals, with the meal beginning immediately after the<br />

time 0 sample and consumed within 15 min. Area under the postprandial glucose curve<br />

(AUC ) was calculated by the trapezoid method. Insulin sensitivity in the postprandial state<br />

G<br />

was calculated as the oral glucose sensitivity index (OGIS) [17].<br />

Insulin signaling in skeletal muscle biopsies: Since skeletal muscle tissue represents the<br />

major site of glucose clearance in the postprandial state, characterized by a classic intracellular<br />

insulin signaling cascade, we assessed the effects of prednisolone on key proteins in this<br />

cascade in skeletal muscle tissue. To this end, a percutaneous skeletal muscle biopsy was<br />

taken from the vastus lateralis muscle using a Bergstrom needle [18] under local anesthesia<br />

with lidocaine 1% (B. Braun, Melsungen, Germany) before and after 30 min of insulin infusion.<br />

Muscle biopsies were washed with sterile NaCl 0.9% to reduce blood contamination and were<br />

thereafter snap-frozen in liquid nitrogen and stored at -80°C for further analysis. Western<br />

blotting of proteins involved in insulin signaling, including phosphorylation of Akt and its<br />

downstream substrate proline-rich Akt substrate of 40 kDa (PRAS40) was done as described<br />

previously [19]. Antibodies for PRAS40, Phospho-Akt-Ser473 and Tubulin were obtained from<br />

Cell Signaling Technology (Boston, MA), for total Akt from Millipore Corporation (Billerica,<br />

MA), for the insulin receptor from Santa Cruz Biotechnology Inc (Santa Cruz, CA) and for<br />

Phospho-PRAS40-Thr246 from Biosource (Carlsbad, CA).<br />

Study medication: Prednisolone tablets were purchased from Pfizer AB (Sollentuna,<br />

Sweden) and placebo tablets were obtained from Xendo Drug Development (Groningen, The<br />

Netherlands). Tablets were capsulated in order to allow the treatment to be blinded [20].<br />

Study medication was taken at 08.00 AM during the two-week treatment except for day 13<br />

and day 14, when it was ingested at 06.00 AM. Patients kept a diary in which the exact time of<br />

medication intake during the study was registered.<br />

Biochemical methods: Blood glucose concentrations were measured using an YSI 2300<br />

STAT Plus analyzer (YSI, Yellow Springs, OH). Insulin levels were determined using an<br />

immunometric assay (Advia, Centaur, Siemens Medical Solutions Diagnostics, USA).<br />

Statistical analyses: Data are presented as mean values ± standard deviation (SD), or as<br />

median (interquartile range) in case of skewed distribution. Absolute changes from baseline<br />

were calculated (on treatment value minus pre treatment value) for all parameters. Nonparametric<br />

analysis was chosen due to uneven sample size in the different groups and the<br />

198


elatively small numbers in each group. Baseline values were compared between the groups<br />

by Kruskal-Wallis. For treatment-induced effects, Kruskal-Wallis with trend analysis was<br />

performed (‘the Jonckheere-Terpstra test’). Only in case of a significant finding, prednisolone<br />

7.5 mg and prednisolone 30 mg were compared against placebo by posthoc testing, using<br />

the Mann-Whitney U test. To correct for multiple testing, Bonferroni correction was applied.<br />

Correlations between the various parameters were assessed with Spearman correlations. All<br />

statistical analyses were run using SPSS version 18.0 for Mac OS X (Chicago, IL, USA). A P <<br />

0.05 was considered statistically significant.<br />

Animal Study<br />

Animals and treatments: The investigation conformed to the Guide for the Care and Use<br />

of Laboratory Animals published by the US National Institutes of Health. The local ethics<br />

committee for animal experiments approved the procedures. Male Wistar rats (300-350<br />

g; Charles-River, Maastricht, the Netherlands) were treated with prednisolone (8 mg/kg<br />

per day; n=6) or saline (n=6) by intraperitoneal injection for 7 days. Two hours after the<br />

final injection, the rats were sacrificed by isoflurane overdose and resistance arteries of the<br />

cremaster muscles were isolated as described previously [21].<br />

Vasoreactivity experiments: After dissection, a first order cremaster arteriole was placed<br />

in a pressure myograph. Only vessels that developed substantial spontaneous constriction<br />

(>40% of the passive diameter) to pressure (65 mmHg) and then showed a clear vasodilator<br />

response (>10% of the diameter) to the endothelium-dependent vasodilator acetylcholine<br />

(0.1 mm; Sigma) were used [21]. Acute effects of insulin (Novo Nordisk, Alphen a/d Rijn, the<br />

Netherlands) on the diameter were studied at three different concentrations: 0.02, 0.2 and 2<br />

nmol/l, and diameter changes at each concentration were recorded for 30 min.<br />

Western blots: Segments of cremaster arteries from the same rat were exposed to insulin<br />

or solvent for 15 min at 34°C as described [22]. The protein lysates were stained with a<br />

specific antibody against phosphorylated Akt, total Akt, phosphorylated ERK1/2 and total<br />

ERK1/2 (Cell Signaling Technology, Boston, MA) and visualized with a chemiluminescence<br />

kit (Amersham). Differences in phosphorylated protein were adjusted for differences in the<br />

corresponding total protein staining or actin.<br />

Statistical analyses: Steady-state responses are reported as mean changes in diameter from<br />

baseline (in percent) ± SEM. The baseline diameter was defined as the arterial diameter just<br />

before addition of the first concentration of insulin. Western blot data were expressed as<br />

relative to unstimulated controls, assigning a value of 1 to the control. Differences between<br />

199<br />

Chapter 10<br />

10


Chapter 10 Glucocorticoids and microvascular function<br />

the various parameters were assessed by the Mann-Whitney U test. A P < 0.05 was considered<br />

statistically significant.<br />

RESULTS<br />

Clinical study<br />

Baseline characteristics and anthropometrics: Prior to treatment, all treatment groups<br />

had comparable anthropometrics, blood pressure and glucose tolerance (Table 1). Treatment<br />

with study medication did not alter body weight (Table 2).<br />

Table 1. Baseline characteristics<br />

Placebo Prednisolone<br />

7.5 mg 30 mg P<br />

N 8 12 12<br />

Age (y) 21±3 21±2 21±2 0.670<br />

Weight (kg) 75±8 75±6 73±10 0.886<br />

Height (cm) 186±7 184±6 185±5 0.718<br />

BMI (kg/m2 ) 21.6±1.5 22.1±1.5 21.4±2.2 0.656<br />

Systolic blood pressure (mmHg) 126±7 124±9 120±9 0.284<br />

Diastolic blood pressure (mmHg) 79±8 79±6 78±10 0.872<br />

Fasting plasma glucose (mmol/l) 4.4±0.3 4.4±0.2 4.5±0.2 0.852<br />

2-hr glucose OGTT (mmol/l) 3.7±1.0 3.9±0.9 3.6±0.8 0.782<br />

Mean±SD are provided. Significance was tested by Kruskal-Wallis. No statistically significant differences<br />

were observed between the groups at baseline. 2-hr glucose OGTT denotes plasma glucose concentrations<br />

2 h after ingestion of 75 g glucose during an oral glucose tolerance test.<br />

Prednisolone impaired capillary recruitment: In the fasted state prior to insulin infusion,<br />

prednisolone treatment did not affect baseline capillary density, capillary recruitment or<br />

maximal number of capillaries following venous congestion (Figure 2A; Table 3). Insulin<br />

infusion increased capillary recruitment by 13±4% (P


Table 2. Metabolic parameters and blood pressure before and during treatment.<br />

Weight<br />

(kg)<br />

Systolic blood pressure<br />

(mmHg)<br />

Diastolic blood pressure<br />

(mmHg)<br />

Fasting plasma glucose<br />

(mmol/l)<br />

Fasting plasma insulin<br />

(pmol/l)<br />

Area under glucose curve<br />

(mmol/l.8hr)<br />

OGIS<br />

(ml/min.m2 )<br />

M-value<br />

(mg/kg.min)<br />

Clamp glucose levels<br />

(mmol/l)<br />

Clamp insulin levels<br />

(pmol/l)<br />

Pre-treatment On-treatment P1 P2 P3<br />

PLB 75±8 76±7<br />

7.5 75±6 75±6<br />

0.539 NA NA<br />

30 73±10 74±10<br />

PLB 126±7 123±9<br />

7.5 124±9 124±11<br />

0.002 0.710 0.006<br />

30 120±9 124±8<br />

PLB 79±8 76±7<br />

7.5 79±6 76±6<br />

0.042 1.0 0.164<br />

30 78±10 80±9<br />

PLB 4.4±0.3 4.5±0.3<br />

7.5 4.4±0.2 4.7±0.3<br />

0.04 0.418 0.09<br />

30 4.5±0.2 4.9±0.4<br />

PLB 31 (28-40) 33 (28-39)<br />

7.5 36 (29-47) 36 (27-65) 0.001 1.0 0.008<br />

30 32 (26-44) 56 (41-72)<br />

PLB 2197±193 2182±92<br />

7.5 2250±121 2492±127


Chapter 10 Glucocorticoids and microvascular function<br />

Figure 2. The effects of prednisolone (PRED) on capillary recruitment. Capillary recruitment in the<br />

fasted state was not impaired by either prednisolone dosage (A), however, prednisolone treatment dosedependently<br />

impaired insulin-stimulated microvascular function (B). Data represent absolute change<br />

from baseline; mean ± SEM are provided. White bars: placebo; grey bars: prednisolone 7.5 mg daily; black<br />

bars: prednisolone 30 mg daily. Between-group changes from baseline were tested by Kruskal-Wallis<br />

with trend analysis (indicated by top line). Posthoc tests were done by Mann-Whitney U with Bonferroni<br />

correction for multiple testing (indicated by line with brackets). ***p


Table 3. Capillary density before and during treatment.<br />

Baseline density fasting<br />

(n/mm2 )<br />

Peak density fasting<br />

(n/mm2 )<br />

Capillary recruitment<br />

fasting<br />

(%)<br />

Venous occlusion<br />

(n/mm2 )<br />

Baseline density<br />

hyperinsulinemia<br />

(n/mm2 )<br />

Peak density<br />

hyperinsulinemia<br />

(n/mm2 )<br />

Capillary recruitment<br />

hyperinsulinemia<br />

(%)<br />

Pre-treatment On-treatment P1 P2 P3<br />

PLB 37.6±3.9 40.9±5.1<br />

7.5 39.4±2.9 40.2±4.3 0.688 NA NA<br />

30 40.1±6.3 41.8±6.6<br />

PLB 47.6±5.4 52.1±6.0<br />

7.5 51.3±5.6 54.3±7.1 0.189 NA NA<br />

30 52.0±8.9 54.8±9.8<br />

PLB 26.6±2.8 26.4±8.4<br />

7.5 30.0±5.8 30.8±7.7<br />

1.0 NA NA<br />

30 29.7±6.6 28.5±8.5<br />

PLB 51.1±6.1 54.5±6.4<br />

7.5 59.6±7.8 60.0±7.6 0.742 NA NA<br />

30 57.0±7.9 60.3±10.6<br />

PLB 37.1±2.1 41.4±6.5<br />

7.5 39.3±4.0 41.0±5.6 0.701 NA NA<br />

30 39.1±6.5 41.7±7.4<br />

PLB 51.3±2.2 60.8±8.1<br />

7.5 57.1±8.0 57.9±8.4 0.011 0.03 0.038<br />

30 55.1±9.7 54.4±10.3<br />

PLB 38.7±4.2 44.8±9.8<br />

7.5 44.9±8.2 41.5±12.9 0.001 0.18 0.002<br />

30 41.2±5.2 30.6±8.2<br />

Data are mean±SD or median (interquartile range). Between-group changes from baseline were tested by<br />

Kruskal-Wallis with trend analysis (P1). In case of a significant finding, posthoc testing (by Mann-Whitney<br />

U) was done (P2 to compare prednisolone 7.5 mg vs. placebo and P3 to compare prednisolone 30 mg vs.<br />

placebo, respectively). Treatment groups: 7.5=prednisolone 7.5 mg daily; 30=prednisolone 30 mg daily.<br />

Abbrevations: NA: not applicable.<br />

Prednisolone dose-dependently impaired glucose metabolism and insulin sensitivity:<br />

Prednisolone 30 mg, but not prednisolone 7.5 mg increased FPG levels relative to placebo<br />

(P=0.04), despite fasting hyperinsulinemia (P=0.008) (Table 2). As compared to placebo,<br />

glucose tolerance during the standardized meal test was dose-dependently reduced by<br />

prednisolone treatment: glucose levels, measured as area under the curve (AUC), increased by<br />

11±5% and 27±9%, for prednisolone 7.5 mg and Prednisolone 30 mg respectively (Table 2).<br />

Clamp-measured whole-body insulin sensitivity was decreased by prednisolone treatment as<br />

compared to placebo in a dose-dependent manner: mean differences: -2.1±0.8 mg/kg.min for<br />

prednisolone 7.5 mg and -4.5±0.7 mg/kg.min for prednisolone 30 mg. Similarly, postprandial<br />

insulin sensitivity, calculated by OGIS, was dose-dependently reduced by prednisolone<br />

treatment as compared to placebo (Table 2).<br />

203<br />

Chapter 10<br />

10


Chapter 10 Glucocorticoids and microvascular function<br />

Figure 4. Associations among prednisolone (PRED)-induced changes in insulin-stimulated<br />

microvascular function, and PRED-induced changes in metabolic and vascular parameters,<br />

including fasting plasma glucose (A), area under the postpandrial glucose curve (B), insulin-stimulated<br />

glucose disposal (C), postprandial insulin sensitivity (D) and systolic blood pressure (E). Correlations<br />

were assessed with Spearman correlations.<br />

204


Prednisolone and insulin signaling in skeletal muscle biopsies: As compared to<br />

placebo, prednisolone treatment did not result in significant reduction in phosphorylation<br />

of Akt (Figure 3A) and PRAS40 (Figure 3B) before and during insulin infusion. Of note, when<br />

within-group changes were assessed, the characteristic insulin-mediated increase in PRASphosphorylation<br />

as observed prior to treatment, was no longer present following high-dose<br />

prednisolone treatment (Figure 4B), indicating reduced insulin-stimulated phosphorylation.<br />

Associations among insulin-stimulated capillary, metabolic parameters and blood<br />

pressure: Prednisolone-induced reductions in insulin-stimulated capillary recruitment were<br />

negatively associated with changes in fasting (P


Chapter 10 Glucocorticoids and microvascular function<br />

reduced the insulin-stimulated increase in Akt phosphorylation (Figure 5B). Basal and<br />

insulin-stimulated phosphorylation of ERK1/2 were not affected by prednisolone treatment<br />

(Figure 5C).<br />

Figure 5. The effects of prednisolone treatment on isolated rat cremaster resistance arterioles.<br />

Prednisolone treatment impaired the vasoactive actions of insulin ex vivo (A). Prednisolone impaired<br />

insulin-stimulated phosphorylation of Akt (B), but did not affect phosphorylation of ERK1/2 (C). A<br />

representative blot is shown in (D). Data indicate percentual change from baseline for vasoreactivity<br />

experiments (A) and the ratio between phosphorylated and total protein content in arbitrary units for<br />

Western Blotting experiments (B) and (C); mean ± SEM are provided. White bars: fasting state; black<br />

bars: insulin infusion. Between-group changes from baseline were tested by Mann-Whitney-U test.<br />

**p


DISCUSSIoN<br />

This is the first study describing a dose-dependent impairment of insulin-stimulated<br />

microvascular function following GC treatment in healthy individuals, as a possible<br />

mechanism contributing to the concomitantly occurring decrease in insulin sensitivity and<br />

glucose tolerance and elevation of blood pressure. These findings were substantiated by the<br />

demonstration of reductions in insulin-stimulated vasodilatation through impaired insulinmediated<br />

phosphorylation of Akt, but unchanged MAPK pathway signaling in rat resistance<br />

arteries. These findings are consistent with a role for vascular insulin resistance in the<br />

development of GC-related adverse metabolic effects.<br />

As observed previously [6, 20], a two-week treatment with prednisolone dose-dependently<br />

reduced whole-body insulin sensitivity and glucose tolerance, while high-dose prednisolone<br />

treatment significantly increased systolic blood pressure. An additional confirmatory finding<br />

of the present study is the identification of the vasoactive properties of insulin, as insulin<br />

increased capillary recruitment by 13% in these healthy subjects. A novel finding was that<br />

prednisolone dose-dependently impaired these vasodilatory actions of insulin in vivo, while<br />

prednisolone treatment did not affect microvascular function in the fasted state. It has been<br />

reported previously that GCs particularly impair metabolism under stimulated conditions, i.e.<br />

during hyperinsulinemic-euglycemic clamps or in the postprandial state. Accordingly, we have<br />

recently shown that prednisolone dose-dependently impaired metabolic fluxes during insulin<br />

infusion, but not in the fasted state when prednisolone induced fasting hyperinsulinemia [6].<br />

Moreover, fasting glucose levels are often only mildly elevated by GC treatment, whereas<br />

during the day significant hyperglycemia develops, especially in the postprandial period [20,<br />

23]. This seems also the case for the vascular effects of prednisolone, because in the present<br />

study we did not observe prednisolone-induced changes in capillary recruitment in the fasted<br />

state, when prednisolone augmented fasting insulin levels. Collectively, the data demonstrated<br />

that prednisolone induced vascular insulin resistance.<br />

Subsequently, we analyzed whether vascular insulin resistance was related to the<br />

prednisolone-induced impairment of glucose metabolism and blood pressure rise. Indeed,<br />

prednisolone-induced impairment in capillary recruitment was significantly associated<br />

with insulin-stimulated glucose disposal during the hyperinsulinemic-euglycemic clamp, as<br />

well as to postprandial glucose levels and postprandial insulin sensitivity, suggesting that<br />

capillary recruitment may also be an important factor contributing to postprandial glucose<br />

metabolism. In line with this finding, Keske and colleagues recently demonstrated using<br />

contrast-enhanced ultrasound that in obese, insulin resistant subjects, capillary recruitment<br />

in the postprandial state was impaired [24].<br />

207<br />

Chapter 10<br />

10


Chapter 10 Glucocorticoids and microvascular function<br />

Additionally, the approximately 6 mmHg increase in systolic blood pressure induced by highdose<br />

prednisolone treatment was related to the decrease in insulin-stimulated capillary<br />

recruitment. Previously, various mechanisms have been proposed to underlie the GC-induced<br />

increase blood pressure, including salt and water retention, altered sympathovagal balance,<br />

increased responsiveness to pressor stimuli such as catecholamines and angiotensin II and<br />

reduced NO availability [25]. We suggest that GC-induced vascular insulin resistance with<br />

impaired insulin signaling via PI3K/Akt to eNOS may be an alternative mechanism to explain<br />

the increase in blood pressure observed during GC treatment. A similar concept has been<br />

proposed for the development of hypertension in obesity and other insulin resistant states<br />

[10-12]. However, the contribution of insulin signaling to eNOS in the regulation of blood<br />

pressure in different states of insulin resistance is not unequivocal [12].<br />

Although it seems very likely that prednisolone treatment also directly affects insulin<br />

signaling in skeletal muscle tissue, we did not detect altered insulin signaling as measured<br />

by phosphorylated Akt and its substrate PRAS40 in skeletal muscle biopsies obtained in the<br />

healthy participants, when compared to placebo. In rat skeletal muscle, it has been shown<br />

previously that the GC dexamethasone impaired various components of the insulin signaling<br />

cascade, including decreased phosphorylation of IRS-1, PI-3K and Akt, resulting in impaired<br />

translocation of glucose transporter (GLUT) 4 to the plasma membrane [26-30]. These<br />

seemingly discrepant findings may not be easily explained. Since we did find statistically<br />

significant changes when within-group comparisons were made, we may have been limited by<br />

the relatively small size of the study. Furthermore, since proteins remain in a phosphorylated<br />

state for a limited time period, the timing of biopsy collection, i.e. at 30 min following onset of<br />

hyperinsulinemia, may have influenced our findings. Taken together, our data are compatible<br />

with an important role for prednisolone-induced vascular insulin resistance in the reduction<br />

in glucose uptake by skeletal muscle during hyperinsulinemia and in the postprandial state<br />

following prednisolone treatment.<br />

Since muscle tissue is the main peripheral site of insulin-mediated glucose uptake and<br />

vascular resistance, it would have been more straightforward to measure insulin-mediated<br />

microvascular recruitment in muscle instead of skin. However, the human skin is the only site<br />

available in humans to directly and to non-invasively assess capillary density and recruitment<br />

of capillaries. Moreover, the effects of obesity and free fatty acids on insulin-mediated<br />

microvascular recruitment in muscle [31, 32] can be reproduced in human skin [15, 33],<br />

suggesting that the vascular responses observed in skin reflect those in muscle. In a recent<br />

study, we could demonstrate concurrent insulin-mediated microvascular recruitment in skin<br />

and skeletal muscle. Both were mutually related and strongly associated with whole-body<br />

208


glucose uptake suggesting that the cutaneous microcirculation is a representative vascular<br />

bed to examine insulin’s actions on the microcirculation [34].<br />

In order to study underlying mechanisms of prednisolone-induced vascular insulin<br />

resistance, Wistar rats were additionally treated with prednisolone. Also in rats, insulin<br />

dose-dependently increased the diameter of isolated cremaster resistance arterioles,<br />

which was impaired by prednisolone treatment on all insulin levels. At the molecular level,<br />

prednisolone attenuated the vascular effects of insulin by reducing phosphorylation of the<br />

Akt pathway. Phosphorylation of the ERK1/2 pathway was not affected. Thus, by reducing<br />

Akt phosphorylation, prednisolone impaired insulin’s vasodilatory actions. Unfortunately, the<br />

amount of eNOS protein in one cremaster resistance artery is not sufficient to measure eNOS<br />

phosphorylation, but Akt phosphorylation has been shown to be a reliable marker of eNOS<br />

phosphorylation in this model [21, 35].<br />

Interestingly, prednisolone treatment specifically attenuated the vasodilatory effects<br />

of insulin, since endothelium-dependent vasodilation as assessed by acetylcholine<br />

administration remained intact. This may indicate that specifically vascular insulin signaling<br />

is disturbed, whereas eNOS function is intact. Nevertheless, acetylcholine may also induce<br />

vasodilation by additional mechanisms than NO production, i.e. by releasing prostanoids and<br />

other endothelium-derived hyperpolarizing factors [36].<br />

In conclusion, prednisolone treatment in healthy volunteers dose-dependently impaired<br />

insulin-stimulated capillary recruitment. In addition, prednisolone-induced changes in<br />

insulin-stimulated capillary recruitment were strongly related to unfavorable metabolic<br />

effects induced by prednisolone treatment, including impaired glucose tolerance, decreased<br />

insulin sensitivity as well as increased systolic blood pressure. Importantly, a substantial part<br />

of the decrease in insulin-stimulated glucose disposal could be explained by prednisoloneinduced<br />

microvascular dysfunction. In isolated rat cremaster resistance arteries, we found<br />

evidence that prednisolone specifically impaired insulin-induced vasodilation by attenuating<br />

insulin-stimulated phosphorylation of the Akt pathway. Thus, we propose that vascular insulin<br />

resistance may contribute to the cardiovascular side effects associated with GC treatment.<br />

Funding: This paper was written within the framework of project T1-106 of the Dutch Top<br />

Institute <strong>Pharma</strong>. Dr E.H. Serné is financially supported by a fellowship from The Netherlands<br />

Heart Foundation (grant no. 2010T041).<br />

209<br />

Chapter 10<br />

10


Chapter 10 Glucocorticoids and microvascular function<br />

REFERENCES<br />

1. Gulliford MC, Charlton j, Latinovic R. Risk of diabetes associated with prescribed glucocorticoids in<br />

a large population. Diabetes Care. 2006;29(12):2728-9.<br />

2. Schacke H, Docke WD, Asadullah K. Mechanisms involved in the side effects of glucocorticoids.<br />

<strong>Pharma</strong>col Ther. 2002;96(1):23-43.<br />

3. van Raalte DH, Ouwens DM, Diamant M. Novel insights into glucocorticoid-mediated diabetogenic<br />

effects: towards expansion of therapeutic options? Eur j Clin Invest. 2009;39(2):81-93.<br />

4. Walker BR. Glucocorticoids and cardiovascular disease. Eur j Endocrinol. 2007;157(5):545-59.<br />

5. Williamson PM, Kelly jj, Whitworth jA. Dose-response relationships and mineralocorticoid activity<br />

in cortisol-induced hypertension in humans. j Hypertens Suppl. 1996;14(5):S37-41.<br />

6. van Raalte DH, Brands M, van der Zijl Nj, Muskiet MH, Pouwels Pj, Ackermans MT, Sauerwein HP,<br />

Serlie Mj, Diamant M. Low-dose glucocorticoid treatment affects multiple aspects of intermediary<br />

metabolism in healthy humans: a randomised controlled trial. Diabetologia. 2011.<br />

7. Saltiel AR, Kahn CR. Insulin signalling and the regulation of glucose and lipid metabolism. Nature.<br />

2001;414(6865):799-806.<br />

8. Barrett Ej, Eggleston EM, Inyard AC, Wang H, Li G, Chai W, Liu Z. The vascular actions of insulin<br />

control its delivery to muscle and regulate the rate-limiting step in skeletal muscle insulin action.<br />

Diabetologia. 2009;52(5):752-64.<br />

9. Kubota T, Kubota N, Kumagai H, Yamaguchi S, Kozono H, Takahashi T, Inoue M, Itoh S, Takamoto<br />

I, Sasako T, Kumagai K, Kawai T, Hashimoto S, Kobayashi T, Sato M, Tokuyama K, Nishimura S,<br />

Tsunoda M, Ide T, Murakami K, Yamazaki T, Ezaki O, Kawamura K, Masuda H, Moroi M, Sugi K, Oike<br />

Y, Shimokawa H, Yanagihara N, Tsutsui M, Terauchi Y, Tobe K, Nagai R, Kamata K, Inoue K, Kodama T,<br />

Ueki K, Kadowaki T. Impaired insulin signaling in endothelial cells reduces insulin-induced glucose<br />

uptake by skeletal muscle. Cell Metab. 2011;13(3):294-307.<br />

10. Serne EH, de jongh RT, Eringa EC, RG Ij, Stehouwer CD. Microvascular dysfunction: a potential<br />

pathophysiological role in the metabolic syndrome. Hypertension. 2007;50(1):204-11.<br />

11. Kim jA, Montagnani M, Koh KK, Quon Mj. Reciprocal relationships between insulin resistance<br />

and endothelial dysfunction: molecular and pathophysiological mechanisms. Circulation.<br />

2006;113(15):1888-904.<br />

12. Li R, Zhang H, Wang W, Wang X, Huang Y, Huang C, Gao F. Vascular insulin resistance in prehypertensive<br />

rats: role of PI3-kinase/Akt/eNOS signaling. Eur j <strong>Pharma</strong>col. 2010;628(1-3):140-7.<br />

13. Muniyappa R, Montagnani M, Koh KK, Quon Mj. Cardiovascular actions of insulin. Endocr Rev.<br />

2007;28(5):463-91.<br />

14. Bakker W, Eringa EC, Sipkema P, van Hinsbergh VW. Endothelial dysfunction and diabetes: roles of<br />

hyperglycemia, impaired insulin signaling and obesity. Cell Tissue Res. 2009;335(1):165-89.<br />

210


15. de jongh RT, Serne EH, RG Ij, de Vries G, Stehouwer CD. Impaired microvascular function in<br />

obesity: implications for obesity-associated microangiopathy, hypertension, and insulin resistance.<br />

Circulation. 2004;109(21):2529-35.<br />

16. DeFronzo RA, Tobin jD, Andres R. Glucose clamp technique: a method for quantifying insulin<br />

secretion and resistance. Am j Physiol. 1979;237(3):E214-23.<br />

17. Mari A, Pacini G, Murphy E, Ludvik B, Nolan jj. A model-based method for assessing insulin sensitivity<br />

from the oral glucose tolerance test. Diabetes Care. 2001;24(3):539-48.<br />

18. Bergstrom j. Percutaneous needle biopsy of skeletal muscle in physiological and clinical research.<br />

Scand j Clin Lab Invest. 1975;35(7):609-16.<br />

19. Nascimento EB, Fodor M, van der Zon GC, jazet IM, Meinders AE, Voshol Pj, Vlasblom R, Baan B,<br />

Eckel j, Maassen jA, Diamant M, Ouwens DM. Insulin-mediated phosphorylation of the prolinerich<br />

Akt substrate PRAS40 is impaired in insulin target tissues of high-fat diet-fed rats. Diabetes.<br />

2006;55(12):3221-8.<br />

20. van Raalte DH, Nofrate V, Bunck MC, van Iersel T, Elassaiss Schaap j, Nassander UK, Heine Rj, Mari<br />

A, Dokter WH, Diamant M. Acute and 2-week exposure to prednisolone impair different aspects of<br />

beta-cell function in healthy men. Eur j Endocrinol. 2010;162(4):729-35.<br />

21. Eringa EC, Stehouwer CD, Merlijn T, Westerhof N, Sipkema P. Physiological concentrations of insulin<br />

induce endothelin-mediated vasoconstriction during inhibition of NOS or PI3-kinase in skeletal<br />

muscle arterioles. Cardiovasc Res. 2002;56(3):464-71.<br />

22. Bradley EA, Eringa EC, Stehouwer CD, Korstjens I, van Nieuw Amerongen GP, Musters R, Sipkema<br />

P, Clark MG, Rattigan S. Activation of AMP-activated protein kinase by 5-aminoimidazole-4carboxamide-1-beta-D-ribofuranoside<br />

in the muscle microcirculation increases nitric oxide<br />

synthesis and microvascular perfusion. Arterioscler Thromb Vasc Biol. 2010;30(6):1137-42.<br />

23. Clore jN, Thurby-Hay L. Glucocorticoid-induced hyperglycemia. Endocr Pract. 2009;15(5):469-74.<br />

24. Keske MA, Clerk LH, Price Wj, jahn LA, Barrett Ej. Obesity blunts microvascular recruitment in<br />

human forearm muscle after a mixed meal. Diabetes Care. 2009;32(9):1672-7.<br />

25. Whitworth jA, Schyvens CG, Zhang Y, Mangos Gj, Kelly jj. Glucocorticoid-induced hypertension: from<br />

mouse to man. Clin Exp <strong>Pharma</strong>col Physiol. 2001;28(12):993-6.<br />

26. Dimitriadis G, Leighton B, Parry-Billings M, Sasson S, Young M, Krause U, Bevan S, Piva T, Wegener<br />

G, Newsholme EA. Effects of glucocorticoid excess on the sensitivity of glucose transport and<br />

metabolism to insulin in rat skeletal muscle. Biochem j. 1997;321 ( Pt 3):707-12.<br />

27. Giorgino F, Almahfouz A, Goodyear Lj, Smith Rj. Glucocorticoid regulation of insulin receptor<br />

and substrate IRS-1 tyrosine phosphorylation in rat skeletal muscle in vivo. j Clin Invest.<br />

1993;91(5):2020-30.<br />

211<br />

Chapter 10<br />

10


Chapter 10 Glucocorticoids and microvascular function<br />

28. Ruzzin j, Wagman AS, jensen j. Glucocorticoid-induced insulin resistance in skeletal muscles: defects<br />

212<br />

in insulin signalling and the effects of a selective glycogen synthase kinase-3 inhibitor. Diabetologia.<br />

2005;48(10):2119-30.<br />

29. Saad Mj, Folli F, Kahn jA, Kahn CR. Modulation of insulin receptor, insulin receptor substrate-1,<br />

and phosphatidylinositol 3-kinase in liver and muscle of dexamethasone-treated rats. j Clin Invest.<br />

1993;92(4):2065-72.<br />

30. Weinstein SP, Wilson CM, Pritsker A, Cushman SW. Dexamethasone inhibits insulin-stimulated<br />

recruitment of GLUT4 to the cell surface in rat skeletal muscle. Metabolism. 1998;47(1):3-6.<br />

31. Liu Z, Liu j, jahn LA, Fowler DE, Barrett Ej. Infusing lipid raises plasma free fatty acids and induces<br />

insulin resistance in muscle microvasculature. j Clin Endocrinol Metab. 2009;94(9):3543-9.<br />

32. Clerk LH, Vincent MA, jahn LA, Liu Z, Lindner jR, Barrett Ej. Obesity blunts insulin-mediated<br />

microvascular recruitment in human forearm muscle. Diabetes. 2006;55(5):1436-42.<br />

33. de jongh RT, Serne EH, Ijzerman RG, de Vries G, Stehouwer CD. Free fatty acid levels modulate<br />

microvascular function: relevance for obesity-associated insulin resistance, hypertension, and<br />

microangiopathy. Diabetes. 2004;53(11):2873-82.<br />

34. Meijer R, De Boer MP, Groen, MR, Eringa EC, Smulders YM, Serne EH. Insulin-induced Microvascular<br />

Recruitment in Skeletal Muscle is Paralleled by Capillary Recruitment in Skin and Both Are<br />

Associated with Whole-Body Glucose Uptake. ADA scientific sessions, 219-OR 2011.<br />

35. Eringa EC, Stehouwer CD, Walburg K, Clark AD, van Nieuw Amerongen GP, Westerhof N, Sipkema P.<br />

Physiological concentrations of insulin induce endothelin-dependent vasoconstriction of skeletal<br />

muscle resistance arteries in the presence of tumor necrosis factor-alpha dependence on c-jun<br />

N-terminal kinase. Arterioscler Thromb Vasc Biol. 2006;26(2):274-80.<br />

36. Versari D, Daghini E, Virdis A, Ghiadoni L, Taddei S. Endothelium-dependent contractions and<br />

endothelial dysfunction in human hypertension. Br j <strong>Pharma</strong>col. 2009;157(4):527-36.


SUPPLEMENTARy MATERIAL<br />

Supplemental Figure 1. Study Design.<br />

Baseline PLB, PRED 7.5 mg or PRED 30 mg once daily Off-treatment<br />

Day S -2 -1 1 7<br />

13 14<br />

T1 T2 M<br />

V T1 T2<br />

S: Screening T1: Meal Tolerance Test T2: Euglycemic M: study medication V: Follow up<br />

Clamp + Capillary<br />

Visit<br />

Supplemental Figure 2. Patient flow diagram. 100% of the randomized participants completed the<br />

study.<br />

28<br />

V<br />

213<br />

Chapter 10<br />

10


Chapter 10 Glucocorticoids and microvascular function<br />

Supplemental Figure 3. Prednisolone treatment did not affect endothelium-dependent vasodilation<br />

as assessed by acetylcholine administration. A dose-response curve is presented. Data are mean ± SD.<br />

Significance was tested by Mann-Whitney U test. Prednisolone-treated rats, n=6; Saline-treated rates,<br />

n=6. Ach: acetylcholine.<br />

Supplemental Table 1. General characteristics of isolated cremaster arterioles following a 7-day<br />

treatment with prednisolone or saline.<br />

General Characteristics Prednisolone 8 mg/kg Saline P<br />

A. Passive Diameter (mm) 184±14 173±8 0.180<br />

B. Basal Diameter (mm) 75±25 83±19 0.485<br />

C. Basal Tone (%) 60±11 52±12 0.310<br />

D. Diameter change after Ach (%) 85±16 65±28 0.310<br />

Data are mean ± SD. Passive diameter was measured after isolation of cremaster arterioles. The basal<br />

diameter was measured at a pressure of 65 mmHg and 37°C. Basal arterial tone was calculated by<br />

{[(A-B)/A]*100}. Significance was tested by Mann-Whitney U test. Prednisolone-treated rats, n=6; Salinetreated<br />

rates, n=6. Ach: acetylcholine.<br />

214


Chapter 11<br />

Glucocorticoids alter Subcutaneous<br />

Adipose Tissue Function and Adipokine<br />

Levels in vivo in Healthy Men<br />

D.H. van Raalte, S. Lehr, H. Mueller, D.M. Ouwens and M Diamant.<br />

Manuscript in Preparation


Chapter 11 Glucocorticoids, adipose tissue & adipokines<br />

ABSTRACT<br />

Introduction: Glucocorticoid (GC) treatment is associated with diabetogenic side effects.<br />

However, the effects on adipose tissue function are currently not completely clear. Evidence<br />

for insulin resistance and increased lipolysis was observed in vitro. In the present study<br />

we assessed the effects of low- and high-dose prednisolone treatment on adipose tissue<br />

function and circulating levels of adipokines in vivo in healthy men.<br />

Research design and methods: Healthy men (n=24; age 21.1±2.1 years; BMI 21.9±1.7<br />

kg/m2 ) were allocated to a two-week treatment with prednisolone 7.5 mg or 30 mg daily<br />

for two weeks. At baseline and on day 14 of treatment, insulin sensitivity was measured by<br />

hyperinsulinemic-euglycemic clamp and plasma levels of adiponectin, resistin and leptin<br />

were determined. An abdominal subcutaneous adipose tissue (SAT) biopsy was taken<br />

in the fasted state and during insulin infusion before and after the intervention to study<br />

insulin signaling (protein kinase B (PKB/Akt) and proline-rich Akt substrate (PRAS)-40<br />

phosphorylation). In addition, adipose tissue protein expression of adipogenesis markers<br />

(peroxisome-proliferator activated receptor (PPAR)-γ and Kruppel-like factor (KLF)-<br />

4), lipolytic enzymes (adipose triglyceride lipase (ATGL) and hormone-sensitive lipase<br />

(HSL)) and adiponectin gene expression was measured. Treatment-induced changes were<br />

compared to baseline using paired statistical tests.<br />

Results: Prednisolone 30 mg increased fasting glucose levels (P


Glucocorticoids (GCs) are important regulators of metabolic homeostasis, but have<br />

diabetogenic effects, in particular when present in supraphysiological concentrations [1].<br />

Although the adverse metabolic effects of elevated GC levels on glucose [2] and protein<br />

metabolism [3] are reasonably well defined, the effects on lipid metabolism are less clear [4].<br />

In addition, the involvement of a number of organs in the development of the diabetogenic<br />

effects of GC treatment, including pancreatic islet cells, skeletal muscle and liver are<br />

understood to a greater extent than the role of adipose tissue. However, several observations<br />

indicate that GCs may exert untoward effects on adipose tissue.<br />

As such, chronic GC excess in Cushing’s syndrome or during GC treatment induces fat<br />

deposition in the visceral compartment [5] and promotes liver fat accumulation [6], with<br />

wasting of subcutaneous adipose tissue depots. This increment in visceral adipose tissue<br />

(VAT) by GCs may be induced by several factors, including increased intake of high-caloric<br />

“comfort food” [7], increased lipoprotein lipase (LPL) activity particularly in VAT [8, 9],<br />

increased VAT adipogenesis through stimulation of differentiation of pre-adipocytes into<br />

mature adipocytes [10] and possibly by enhancing de novo lipogenesis [4, 11]. GC-induced<br />

increase in VAT is very relevant since VAT is well known to be associated with an unfavorable<br />

metabolic profile as opposed to subcutaneous adipose tissue (SAT) [12].<br />

In contrast to their lipogenic actions, GCs are catabolic hormones and, as such, have lipolytic<br />

properties [4, 11]. In most performed in vitro studies, GCs increased lipolysis [4] by enhancing<br />

the activity of key lipolytic enzymes including adipose triglyceride lipase (ATGL) and hormone<br />

sensitive lipase (HSL) [13, 14]. It should be noted that differences were observed between<br />

adipose tissue depots with increased lipolysis in VAT as compared to SAT [15]. In addition,<br />

antilipolytic effects of GC treatment have also been observed in vitro [4]. In line with most<br />

of the in vitro observations, acute GC administration was shown to increase fasting wholebody<br />

lipolysis in healthy humans [16, 17], however, during more prolonged GC exposure,<br />

this effect was no longer present, most likely due to compensatory fasting hyperinsulinemia<br />

[18-20]. Indeed, we could recently show that a two-week treatment with prednisolone 7.5<br />

mg did not increase, while prednisolone 30 mg daily even reduced basal lipolysis in healthy<br />

men [21]. However, in the same study, prednisolone treatment dose-dependently impaired<br />

insulin-stimulated suppression of lipolysis and thus increased plasma nonesterified fatty<br />

acids (NEFA) during hyperinsulinemia [21]. It is noteworthy that these effects occurred in<br />

the absence of significant changes in body weight, body composition, body fat distribution<br />

and liver fat content. Increased lipolysis and consequently enhanced plasma NEFA levels<br />

are detrimental since they impair glucose metabolism by reducing muscle and liver insulin<br />

sensitivity [22].<br />

219<br />

Chapter 11<br />

11


Chapter 11 Glucocorticoids, adipose tissue & adipokines<br />

Finally, another mechanism by which GC-induced changes in adipose tissue may exert its<br />

diabetogenic effects is via alteration of the secretion of various adipokines, as was shown<br />

in in vitro studies [23]. An important limitation of the above-mentioned studies that have<br />

addressed the effects of GCs on adipose tissue function is that they were mostly carried out in<br />

vitro or in vivo in rodents, and no data are available from in vivo studies in humans. Therefore,<br />

in the present study, we assessed changes in SAT function and plasma levels of adipokines<br />

following a two-week treatment with low- or high dose prednisolone treatment in healthy<br />

men.<br />

RESEARCH DESIGN AND METHoDS<br />

Participants: Twenty-four healthy normoglycemic Caucasian men (age 18-35 years, BMI<br />

20-25 kg/m2 ) were recruited. Health status was confirmed by history, physical examination,<br />

screening blood tests and 75-g 2-h OGTT during the screening visit. Exclusion criteria<br />

were the presence of any disease, use of any medication, first-degree relative with type 2<br />

diabetes, smoking, shift work, a history of glucocorticoid use, sport activities > twice/<br />

week and changes in weight in the 3 months prior to study participation. The study was<br />

approved by an independent ethics committee and the study was conducted in accordance<br />

with the Declaration of Helsinki. All participants provided written informed consent before<br />

participation.<br />

Study design: Participants were randomly assigned to a 14-day treatment with prednisolone<br />

7.5 mg once daily or prednisolone 30 mg once daily. At baseline and after two weeks of<br />

treatment, fasting blood samples were obtained, insulin sensitivity was measured by<br />

hyperinsulinemic-euglycemic clamp and subcutaneous adipose tissue biopsies were obtained<br />

both in the fasted state and following 45-min insulin infusion. Subjects were asked to refrain<br />

from drinking alcohol and to perform no strenuous exercise for a period of 48h before the<br />

study days.<br />

Hyperinsulinemic-euglycemic clamp: A two-hour hyperinsulinemic-euglycemic clamp<br />

at 5.0 mM was performed to assess insulin sensitivity as described [24]. The mean glucose<br />

infusion rate during steady state (last 30 min of the clamp) was used to assess insulin<br />

sensitivity.<br />

Adipose tissue biopsy: An abdominal subcutaneous adipose tissue biopsy was collected<br />

6-8 cm lateral from the umbilicus under local anesthesia (2% lidocaine) by needle biopsy.<br />

220


Adipose tissue was washed with sterile saline and was snap frozen in liquid nitrogen and<br />

stored at -80°C until analysis.<br />

Biochemical analyses: Blood glucose concentrations were measured using an YSI 2300<br />

STAT Plus analyzer (YSI, Yellow Springs, OH). Insulin levels were determined using an<br />

immunometric assay (Advia Centaur; Siemens Medical Solutions Diagnostics, Deerfield,<br />

IL). A single-plex human magnetic adiponectin and a three-plex human leptin, visfatin, and<br />

resistin assay (all from Biorad, Hercules, CA, USA) were used to determine serum levels of<br />

adiponectin, leptin, visfatin, and resistin using a Bioplex 200 suspension array system and<br />

Bioplex Pro II wash station (Biorad, Hercules, CA, USA) respectively. Adipokine concentrations<br />

were calculated from the appropriate optimized standard curves using Bio-Plex Manager<br />

software version 6.0 (Biorad). Intra- and interassay coefficients of variation for the Bioplex<br />

assays were both


Chapter 11 Glucocorticoids, adipose tissue & adipokines<br />

study design, no formal power analysis was done. Calculations were done using SPSS 18.0 for<br />

Mac (Chicago, IL, USA). P


Figure 1. Effects of glucocorticoids on adipose tissue function. Prednisolone 30 mg, but not<br />

prednisolone 7.5 mg treatment significantly impaired insulin signaling as measured by insulin-stimulated<br />

phosphorylation of Akt and proline-rich Akt substrate (PRAS)-40 (panel A+B). Both prednisolone<br />

dosages reduced the expression of adipogenesis markers peroxisome proliferator-activated receptor<br />

(PPAR)-γ and krüppel-like factor (KLF)-4 (panel C+D). Adipose triglyceride lipase (ATGL) expression was<br />

reduced by prednisolone 7.5 mg and 30 mg in the fasted state, but no differences were observed during<br />

insulin infusion (panel E). Neither fasting hormone sensitive lipase (HSL) expression (panel F) nor HSL<br />

phosphorylation by insulin was affected by prednisolone treatment (panel G). Adiponectin expression<br />

was reduced by both prednisolone dosages (panel H). Mean±SD is shown. For panel A,B and G: white<br />

bar: pre treatment, fasting; black bar: pre treatment, during insulin infusion; bar with block-pattern:<br />

post treatment, fasting; grey bar: post treatment, during insulin For panel C, D, E, F and H: white bar:<br />

pretreatment, black bar: post treatment. # P=0.07;*P< 0.05;**P0.05).<br />

223<br />

Chapter 11<br />

11


Chapter 11 Glucocorticoids, adipose tissue & adipokines<br />

PRED7.5 mg did not alter the plasma levels of any of these adipokines (P>0.05) (Table 1). No<br />

associations were observed between prednisolone-induced changes in insulin sensitivity and<br />

changes in circulating levels of adipokines (data not shown).<br />

Adipose tissue insulin signaling: Insulin infusion stimulated phosphorylation of PKB/<br />

Akt and its substrate PRAS40 as compared to the fasted state. This effect was reduced by<br />

prednisolone 30 mg treatment (Figure 1A+B). Although a trend was observed for the low<br />

dose group, this effect failed to reach statistical significance (Figure 1A+B).<br />

Adipose tissue markers of adipogenesis: Both low-and high dose prednisolone treatment<br />

decreased the expression of markers of adipose tissue differentiation/adipogenesis PPAR-γ<br />

and KLF-4 (Figure 1C+D).<br />

Adipose tissue lipolytic enzymes: Both prednisolone 7.5 mg and prednisolone 30 mg<br />

reduced the ATGL expression (Figure 1E), but did not alter HSL protein expression (Figure<br />

1F). Insulin tended to lower HSL phosphorylation (P=0.06), which was not altered by low- or<br />

high-dose prednisolone treatment (Figure 1G).<br />

Adipose tissue adiponectin expression: Both prednisolone 7.5 mg and prednisolone 30<br />

mg reduced the expression of adiponectin (P


Impaired insulin action in adipose tissue may adversely affect both glucose and lipid<br />

metabolism. Indeed, an intact insulin signaling pathway is of pivotal importance for insulinstimulated<br />

glucose uptake through recruitment of the glucose transporter (GLUT)-4 to<br />

the plasma membrane enabling glucose transport into the cell [27]. Although the overall<br />

contribution of glucose uptake by adipose tissue in the postprandial state is thought to be<br />

relatively modest [28], it does contribute to glucose tolerance as was elegantly demonstrated<br />

in adipose tissue-specific glut4-/- mice [29]. These mice developed hyperglycemia and were<br />

characterized by liver and skeletal muscle insulin resistance, most likely due to altered<br />

secretion of adipokines [29]. In our study, prednisolone 30 mg altered the secretion of various<br />

adipokines towards a more diabetogenic profile. Thus, plasma concentrations of adiponectin,<br />

which are generally positively associated with insulin sensitivity, showed a tendency toward<br />

reduced levels following high-dose prednisolone treatment, whereas adiponectin protein<br />

expression in SAT was decreased by both prednisolone dosages. The adipokines resistin<br />

and leptin, which are usually increased in insulin resistant states were both increased by<br />

prednisolone 30 mg. Although increased leptin levels has been shown to inhibit food intake<br />

in the CNS, leptin also acts as pro-inflammatory factor in adipose tissue by promoting the<br />

release of pro-inflammatory adipokines. Similarly, resistin has also been demonstrated to<br />

promote circulating pro-inflammatory factors derived from adipose tissue [30].<br />

In addition to promoting glucose uptake in adipose tissue, insulin is a key hormone in the<br />

regulation of lipid metabolism, amongst others by inhibiting adipose tissue lipolysis and<br />

thus decreasing plasma NEFA levels [31]. In contrast to the effects of insulin, GCs increase<br />

fasting lipolysis rates when administered acutely [16, 17]. During more prolonged GC<br />

exposure, however, this lipolytic effect is no longer observed most likely due to compensatory<br />

hyperinsulinemia [18-21]. In our recent study, high-dose prednisolone treatment even<br />

lowered fasting lipolysis [21]. In line with these findings, we observed decreased fasting ATGL<br />

expression, the rate-limiting enzyme in adipose tissue lipolysis, during both low- and highdose<br />

prednisolone treatments, while fasting HSL expression was unchanged.<br />

As stated previously, GCs impaired the suppressive effects of insulin on whole-body lipolysis<br />

[21]. Since both ATGL expression and phosphorylation of HSL during insulin infusion were<br />

unaltered by prednisolone treatment, it is unclear through which mechanisms this effect<br />

occurs. Possibly, the inhibiting effects of insulin on protein kinase A and perilipin expression, a<br />

lipid droplet-associating protein, are reduced by prednisolone treatment, thereby promoting<br />

triglyceride lipolysis [13]. An alternative hypothesis is that the increment in whole-body<br />

lipolysis is mostly derived from VAT, from which no biopsy could be obtained in the present<br />

225<br />

Chapter 11<br />

11


Chapter 11 Glucocorticoids, adipose tissue & adipokines<br />

study design. Importantly, increased lipolysis levels impair glucose homeostasis by reducing<br />

muscle and liver insulin sensitivity [22].<br />

In addition to changing adipose tissue function, GCs are known to alter adipose tissue<br />

distribution during more prolonged treatment by augmenting central fat deposition with<br />

wasting of peripheral fat depots. Although two weeks is a relatively short treatment period,<br />

and no significant alterations were shown in body fat distribution in a previous study [21], we<br />

observed decreased markers of adipogenesis in SAT, compatible with the observed phenotype<br />

after more prolonged GC exposure. The GC-induced reduction in SAT with concomitant<br />

increase in VAT is clinically relevant since VAT is well known to be associated with an untoward<br />

metabolic profile as opposed to SAT and is associated with increased cardiovascular risk [12].<br />

Importantly, a number of effects were already observed during treatment with prednisolone<br />

7.5 mg daily, a low dose commonly used in clinical practice for prolonged periods. Many<br />

patients treated with GCs in clinical practice suffer from systemic inflammatory conditions,<br />

which may also be associated with impaired adipose tissue function [32]. Therefore the<br />

effects of GC treatment on adipose tissue function may be more complex since GCs reduce<br />

systemic inflammation and require further investigation.<br />

In conclusion, this study demonstrates for the first time that prednisolone treatment impaired<br />

SAT function by impairing insulin signaling and reducing adipogenesis in vivo in healthy<br />

humans. In addition, plasma levels of adiponectin, leptin and resistin were changed to a more<br />

diabetogenic profile. Further studies in humans are warranted to study the mechanisms that<br />

may underly the effects of GCs on adipose tissue function.<br />

Funding: This paper was written within the framework of project T1-106 of the Dutch Top<br />

Institute <strong>Pharma</strong>.<br />

226


REFERENCES<br />

1. van Raalte DH, Ouwens DM, Diamant M. Novel insights into glucocorticoid-mediated diabetogenic<br />

effects: towards expansion of therapeutic options? Eur j Clin Invest. 2009;39(2):81-93.<br />

2. McMahon M, Gerich j, Rizza R. Effects of glucocorticoids on carbohydrate metabolism. Diabetes<br />

Metab Rev. 1988;4(1):17-30.<br />

3. Hamel FG, Fawcett j, Bennett RG, Duckworth WC. Control of proteolysis: hormones, nutrients, and<br />

the changing role of the proteasome. Curr Opin Clin Nutr Metab Care. 2004;7(3):255-8.<br />

4. Peckett Aj, Wright DC, Riddell MC. The effects of glucocorticoids on adipose tissue lipid metabolism.<br />

Metabolism. 2011;60(11):1500-10.<br />

5. Rockall AG, Sohaib SA, Evans D, Kaltsas G, Isidori AM, Monson jP, Besser GM, Grossman AB, Reznek<br />

RH. Computed tomography assessment of fat distribution in male and female patients with Cushing’s<br />

syndrome. Eur j Endocrinol. 2003;149(6):561-7.<br />

6. Rockall AG, Sohaib SA, Evans D, Kaltsas G, Isidori AM, Monson jP, Besser GM, Grossman AB, Reznek<br />

RH. Hepatic steatosis in Cushing’s syndrome: a radiological assessment using computed tomography.<br />

Eur j Endocrinol. 2003;149(6):543-8.<br />

7. Dallman MF, la Fleur SE, Pecoraro NC, Gomez F, Houshyar H, Akana SF. Minireview: glucocorticoids-<br />

-food intake, abdominal obesity, and wealthy nations in 2004. Endocrinology. 2004;145(6):2633-8.<br />

8. Fried SK, Russell CD, Grauso NL, Brolin RE. Lipoprotein lipase regulation by insulin and<br />

glucocorticoid in subcutaneous and omental adipose tissues of obese women and men. j Clin Invest.<br />

1993;92(5):2191-8.<br />

9. Ottosson M, Vikman-Adolfsson K, Enerback S, Olivecrona G, Bjorntorp P. The effects of cortisol<br />

on the regulation of lipoprotein lipase activity in human adipose tissue. j Clin Endocrinol Metab.<br />

1994;79(3):820-5.<br />

10. Hauner H, Entenmann G, Wabitsch M, Gaillard D, Ailhaud G, Negrel R, Pfeiffer EF. Promoting effect of<br />

glucocorticoids on the differentiation of human adipocyte precursor cells cultured in a chemically<br />

defined medium. J Clin Invest. 1989;84(5):1663-70.<br />

11. Macfarlane DP, Forbes S, Walker BR. Glucocorticoids and fatty acid metabolism in humans: fuelling<br />

fat redistribution in the metabolic syndrome. j Endocrinol. 2008;197(2):189-204.<br />

12. Ibrahim MM. Subcutaneous and visceral adipose tissue: structural and functional differences. Obes<br />

Rev. 2010;11(1):11-8.<br />

13. Xu C, He j, jiang H, Zu L, Zhai W, Pu S, Xu G. Direct effect of glucocorticoids on lipolysis in adipocytes.<br />

Mol Endocrinol. 2009;23(8):1161-70.<br />

14. Campbell JE, Peckett AJ, D’Souza A M, Hawke TJ, Riddell MC. Adipogenic and lipolytic effects of<br />

chronic glucocorticoid exposure. Am j Physiol Cell Physiol. 2011;300(1):C198-209.<br />

227<br />

Chapter 11<br />

11


Chapter 11 Glucocorticoids, adipose tissue & adipokines<br />

15. Lundgren M, Buren J, Lindgren P, Myrnas T, Ruge T, Eriksson JW. Sex- and depot-specific lipolysis<br />

regulation in human adipocytes: interplay between adrenergic stimulation and glucocorticoids.<br />

Horm Metab Res. 2008;40(12):854-60.<br />

16. Samra jS, Clark ML, Humphreys SM, Macdonald IA, Matthews DR, Frayn KN. Effects of morning rise<br />

in cortisol concentration on regulation of lipolysis in subcutaneous adipose tissue. Am j Physiol.<br />

1996;271(6 Pt 1):E996-1002.<br />

17. Divertie GD, jensen MD, Miles jM. Stimulation of lipolysis in humans by physiological<br />

hypercortisolemia. Diabetes. 1991;40(10):1228-32.<br />

18. Birkenhager JC, Timmermans HA, Lamberts SW. Depressed plasma FFA turnover rate in Cushing’s<br />

syndrome. j Clin Endocrinol Metab. 1976;42(1):28-32.<br />

19. Saunders J, Hall SE, Sonksen PH. Glucose and free fatty acid turnover in Cushing’s syndrome. J<br />

Endocrinol Invest. 1980;3(3):309-11.<br />

20. Miyoshi H, Shulman GI, Peters Ej, Wolfe MH, Elahi D, Wolfe RR. Hormonal control of substrate cycling<br />

in humans. j Clin Invest. 1988;81(5):1545-55.<br />

21. van Raalte DH, Brands M, van der Zijl Nj, Muskiet MH, Pouwels Pj, Ackermans MT, Sauerwein HP,<br />

Serlie Mj, Diamant M. Low-dose glucocorticoid treatment affects multiple aspects of intermediary<br />

metabolism in healthy humans: a randomised controlled trial. Diabetologia. 2011;54(8):2103-12.<br />

22. Boden G. Obesity, insulin resistance and free fatty acids. Curr Opin Endocrinol Diabetes Obes.<br />

2011;18(2):139-43.<br />

23. Fasshauer M, Paschke R. Regulation of adipocytokines and insulin resistance. Diabetologia.<br />

2003;46(12):1594-603.<br />

24. DeFronzo RA, Tobin jD, Andres R. Glucose clamp technique: a method for quantifying insulin<br />

secretion and resistance. Am j Physiol. 1979;237(3):E214-23.<br />

25. Turnbow MA, Keller SR, Rice KM, Garner CW. Dexamethasone down-regulation of insulin receptor<br />

substrate-1 in 3T3-L1 adipocytes. j Biol Chem. 1994;269(4):2516-20.<br />

26. Wamil M, Battle jH, Turban S, Kipari T, Seguret D, de Sousa Peixoto R, Nelson YB, Nowakowska<br />

D, Ferenbach D, Ramage L, Chapman KE, Hughes j, Dunbar DR, Seckl jR, Morton NM. Novel fat<br />

depot-specific mechanisms underlie resistance to visceral obesity and inflammation in 11 betahydroxysteroid<br />

dehydrogenase type 1-deficient mice. Diabetes. 2011;60(4):1158-67.<br />

27. Kanzaki M. Insulin receptor signals regulating GLUT4 translocation and actin dynamics. Endocr j.<br />

2006;53(3):267-93.<br />

28. DeFronzo RA, jacot E, jequier E, Maeder E, Wahren j, Felber jP. The effect of insulin on the disposal<br />

of intravenous glucose. Results from indirect calorimetry and hepatic and femoral venous<br />

catheterization. Diabetes. 1981;30(12):1000-7.<br />

29. Abel ED, Peroni O, Kim jK, Kim YB, Boss O, Hadro E, Minnemann T, Shulman GI, Kahn BB.<br />

Adipose-selective targeting of the GLUT4 gene impairs insulin action in muscle and liver. Nature.<br />

2001;409(6821):729-33.<br />

228


30. Ouchi N, Parker JL, Lugus JJ, Walsh K. Adipokines in inflammation and metabolic disease. Nat Rev<br />

Immunol. 2011;11(2):85-97.<br />

31. Dimitriadis G, Mitrou P, Lambadiari V, Maratou E, Raptis SA. Insulin effects in muscle and adipose<br />

tissue. Diabetes Res Clin Pract. 2011;93 Suppl 1:S52-9.<br />

32. Rho YH, Chung CP, Solus jF, Raggi P, Oeser A, Gebretsadik T, Shintani A, Stein CM. Adipocytokines,<br />

insulin resistance, and coronary atherosclerosis in rheumatoid arthritis. Arthritis Rheum.<br />

2010;62(5):1259-64.<br />

229<br />

Chapter 11<br />

11


Chapter 12<br />

Angiopoietin-Like Protein 4 is Differentially<br />

Regulated by Glucocorticoids and Insulin in<br />

vitro and in vivo in Healthy Humans<br />

D.H. van Raalte*, M. Brands*, M.J. Serlie, K.Mudde, R.Stienstra,<br />

H.P. Sauerwein, S. Kersten and M. Diamant<br />

*Both authors contributed equally<br />

Expl Clin Endocrinol Diabetes. 2012, in press


Chapter 12 Angptl4 in glucocorticoid-induced diabetogenic effects<br />

ABSTRACT<br />

objective: Angiopoietin-like protein 4 (Angptl4) is a circulating inhibitor of plasma<br />

triglyceride clearance via inhibition of lipoprotein lipase. The aim of the present study was<br />

to examine the regulation of Angptl4 by glucocorticoids and insulin in vivo in humans, since<br />

these factors regulate Angptl4 expression in vitro.<br />

Research Design and Methods: In a randomized, placebo-controlled, double-blind, doseresponse<br />

intervention study, 32 healthy males (age: 22±3 years; BMI 22.4±1.7 kg m-2 ) were<br />

allocated to prednisolone 30 mg once daily (n=12), prednisolone 7.5 mg once daily (n=12),<br />

or placebo (n=8) for two weeks. Angptl4 levels and lipid metabolism were measured before<br />

and at 2 weeks of treatment, in the fasted state and during a two-step hyperinsulinemic<br />

clamp. Additionally, human hepatoma cells were treated with dexamethasone and/or<br />

insulin.<br />

Results: Compared to placebo, prednisolone treatment tended to lower fasting Angptl4<br />

levels (P=0.073), raised fasting insulin levels (P=0.0004) and decreased fasting nonesterified<br />

fatty acid concentrations (NEFA) (P=0.017). Insulin infusion reduced Angptl4 levels by 6%<br />

(plasma insulin~200 pmol/l, P=0.006) and 22% (plasma insulin~600 pmol/l, P


The regulation of circulating lipids is tightly controlled. An important endocrine factor involved<br />

in the regulation of circulating lipid levels is angiopoietin-like protein (Angptl) 4. Angptl4 is a<br />

50kD protein secreted by a variety of tissues including liver, white adipose tissue and skeletal<br />

muscle [1-3]. Studies in various animal models have shown that Angptl4 increases plasma<br />

triglyceride levels [4-6], by inhibiting plasma lipoprotein lipase (LPL) activity [7]. Accordingly,<br />

carriers of a rare dysfunctional variant of the Angptl4 gene exhibit low plasma TG levels [8].<br />

Besides raising plasma TG, Angptl4 increases plasma non-esterified fatty acids (NEFA) by<br />

stimulating white adipose tissue lipolysis [6, 9].<br />

Expression of Angptl4 is stimulated by fatty acids in several mouse tissues via activation of<br />

the peroxisome-proliferator activated receptors α, γ and δ [3, 10-12]. Consistent with a role of<br />

PPARs in regulating Angptl4 production in humans, synthetic agonists of PPARα and PPARγ<br />

raise plasma Angptl4 levels in human subjects [3, 4, 13].<br />

Insulin, on the other hand, may suppress Angptl4 levels. In 3T3-L1 adipocytes, insulin<br />

treatment reduced Angptl4 expression, an effect that was attenuated by pharmacological<br />

inhibition of the insulin-signaling cascade and by tumor necrosis factor-α treatment [14].<br />

Recently, a possible role for glucocorticoids (GCs) in the regulation of Angptl4 expression was<br />

suggested. Dexamethasone treatment was found to increase Angptl4 mRNA levels in primary<br />

hepatocytes and adipocytes, and in mouse liver and white adipose tissue. A glucocorticoid<br />

response element (GRE) was identified in the 3′-untranslated region of the Angptl4 gene,<br />

suggesting that Angptl4 represents a direct GC target gene [15]. Interestingly, Angptl4-/- mice<br />

were less susceptible to hypertriglyceridemia and hepatic steatosis following dexamethasone<br />

treatment [15]. Whether GCs are important regulators of Angptl4 levels in humans is presently<br />

unclear.<br />

More recently, we conducted a study in which we observed that short term treatment with both<br />

low- and high-dose prednisolone in healthy men affected glucose metabolism and lipolysis,<br />

resulting in impaired insulin-stimulated suppression of endogenous glucose production<br />

and whole-body lipolysis [16]. Given the recent discovery of the involvement of Angptl4 in<br />

glucocorticoid-induced metabolic deregulation, we additionally determined Angptl4 plasma<br />

concentrations in these individuals before and after prednisolone treatment in samples that<br />

were collected during the study. We hypothesized that prednisolone would increase Angptl4<br />

plasma concentrations and that changes in Angptl4 concentrations would be related to the<br />

observed prednisolone-induced alterations in lipid metabolism. Our experimental design<br />

also permitted to assess the effect of hyperinsulinemia on plasma Angptl4 concentrations.<br />

We hypothesized that insulin infusion would decrease Angptl4 concentrations and that<br />

233<br />

Chapter 12<br />

12


Chapter 12 Angptl4 in glucocorticoid-induced diabetogenic effects<br />

prednisolone would attenuate this effect by inducing insulin resistance. The study was<br />

supported by in vitro experiments using human hepatoma cells.<br />

RESEARCH DESIGN AND METHoDS<br />

Clinical study<br />

Participants: Thirty-two healthy normoglycemic males were included, following a screening<br />

visit during which history was taken and a physical examination, screening blood tests and<br />

an oral glucose tolerance test were performed. Inclusion criteria were: age between 18-35<br />

years and body mass index (BMI) ≤ 25 kg/m2 . Exclusion criteria were any previous or current<br />

illness, use of any medication, first-degree relative with type 2 diabetes, smoking, shift work, a<br />

history of glucocorticoid use, excessive sport activities (i.e. more often than two times/week)<br />

and recent changes in weight. Participants were recruited via local advertisement and mostly<br />

concerned students.<br />

Study design: Details on study design were reported previously [16]. Briefly, in a randomized,<br />

placebo-controlled, double-blind, dose-response intervention trial, 32 healthy volunteers<br />

were allocated to a treatment with prednisolone 30 mg once daily (n=12), prednisolone 7.5<br />

mg once daily (n=12) or placebo (n=8) for a period of two weeks using block randomization<br />

as carried out by the department of Experimental <strong>Pharma</strong>cology of the VU University<br />

Medical Center. Before and at day 14 of treatment, Angptl4 levels, glucose metabolism and<br />

total triglyceride lipolysis were measured in the fasted state and during a hyperinsulinemiceuglycemic<br />

clamp using stable isotopes. The study protocol was approved by a local ethics<br />

committee and was performed in accordance to the principles described in the declaration<br />

of Helsinki. All participating subjects gave their written informed consent prior to screening.<br />

Experimental protocol: Design of the hyperinsulinemic-euglycemic clamp with stable<br />

isotopes is described in detail elsewhere [16]. In short, following an overnight fast and<br />

following measurement of background enrichments, a primed, continuous infusion of isotopes<br />

was started: [6,6-2H ]glucose (prime: 11 mmol/kg; continuous: 0.11 mmol/kg.min) and<br />

2<br />

[1,1,2,3,3,-2H ]glycerol (prime: 1.6 mmol/kg; continuous: 0.11 mmol/kg.min) and continued<br />

5<br />

until the end of the clamp. After a 2-h equilibrium period, blood samples were drawn for<br />

determination of basal glucose concentrations, isotope enrichments, glucoregulatory<br />

hormones, NEFA and Angptl4. Thereafter, a 2-step hyperinsulinemic-euglycemic clamp was<br />

started: step 1 included an infusion of insulin at a rate of 20 mU/m2 .min for 2 hours to assess<br />

hepatic insulin sensitivity; during step 2, insulin was infused at a rate of 60 mU/m2 .min for<br />

234


2 hours to assess peripheral insulin sensitivity. Plasma glucose was kept at 5 mmol/l by a<br />

variable infusion of glucose 20% solution enriched with [6,6-2H ]glucose. After 2 hours of each<br />

2<br />

insulin infusion, blood samples were drawn for the measurement of glucose concentrations,<br />

isotopic enrichments, glucoregulatory hormones, NEFA and Angptl4 (Figure 1).<br />

Figure 1. Design of the two-step hyperinsulinemic clamp using stable isotopes.<br />

Laboratory Analyses: Angptl4 was measured by ELISA as described previously [3]. Briefly,<br />

96-well plates were coated with anti-human Angptl4 polyclonal goat IgG antibody (AF3485,<br />

R&D Systems) and incubated overnight at 4°C. Plates were washed extensively between each<br />

step. After blocking, 100 μl of 20-fold diluted human plasma was applied, followed by 2 h<br />

incubation at room temperature. A standard curve was prepared using recombinant human<br />

Angptl4 (3485-AN, R&D Systems) at 0.3 to 2.1 ng/well. Next, 100 μl of diluted biotinylated<br />

anti-human Angptl4 polyclonal goat IgG antibody (BAF3485, R&D Systems) was added for<br />

2 h, followed by addition of streptavidin-conjugated horseradish peroxidase for 20 min, and<br />

tetramethyl benzidine substrate reagent for 6 min. The reaction was stopped by addition of<br />

50 μl of 10% H2SO4, and the absorbance was measured at 450 nm. The intra-assay coefficient<br />

of variation (CV) for the assay was determined at 7%. Plasma glucose concentrations were<br />

measured with the glucose oxidase method using a Biosen C-line plus glucose analyzer (EKF<br />

Diagnostics, Barleben/Magedeburg, Germany). Insulin was determined on an Immulite<br />

2000 system (Diagnostic Products Corporation, Los Angeles, CA) with a chemiluminescent<br />

immunometric assay. Plasma NEFA concentrations were measured with an enzymatic<br />

colorimetric method (NEFA-C test kit; Wako Chemicals GmbH, Neuss, Germany). [6,6-2H ] 2<br />

Glucose and [1,1,2,3,3,-2H ]glycerol enrichment were measured with gas chromatography-<br />

5<br />

mass spectrometry (GC-MS) as described in detail elsewhere [17, 18].<br />

Calculations: Endogenous glucose production (EGP) and the peripheral uptake of glucose<br />

(Rate of disappearance, Rd) were calculated using modified versions of the Steele equations<br />

for the non-steady state [19, 20]. Lipolysis (glycerol turnover) was computed using formulas<br />

for steady state kinetics adapted for stable isotopes [17].<br />

235<br />

Chapter 12<br />

12


Chapter 12 Angptl4 in glucocorticoid-induced diabetogenic effects<br />

Statistical analyses: Data are presented as mean values ± standard deviation (SD), or<br />

as median (interquartile range) in case of skewed distribution. To assess the effects of<br />

prednisolone treatment on various parameters, absolute changes from baseline (on treatment<br />

value minus pre treatment value) were computed for each treatment arm. Significance was<br />

tested using the Kruskal-Wallis test. Non-parametric analysis was chosen due to the relatively<br />

small number of participants and the uneven group sizes (P1 value in Table 2). Only in case<br />

of a significant finding, prednisolone 7.5 mg and prednisolone 30 mg were compared against<br />

placebo by posthoc testing, using the Mann-Whitney U test (P2 and P3 respectively in Table<br />

2). To correct for multiple testing, Bonferroni correction was applied. The effects of insulin<br />

infusion on plasma Angptl4 levels prior to PRED treatment was tested with the paired t-test.<br />

Correlations between Angptl4 levels and parameters of glucose and lipid metabolism were<br />

calculated by regression analysis adjusting for age, BMI [21] and insulin concentrations.<br />

Since the measurement of Angptl4 was not a primary endpoint of the original study, no formal<br />

power calculation was performed for this parameter. However, our sample size determined<br />

for the primary endpoints of the original protocol would provide a power of 82% with an<br />

estimated PRED-induced increase of 3 ng/mL in plasma Angptl4 plasma concentrations,<br />

a standard deviation of 30% and alpha set at 0.05, which we considered sufficient. The<br />

estimated changes were based on other interventions that altered Angptl4 concentrations,<br />

including fenofibrate treatment and caloric restriction [3]. All statistical analyses were run<br />

on SPSS 18.0 for Mac OS X (SPSS, Chicago, IL, USA). All statistical tests were conducted at a<br />

significance level of 0.05 (two-sided).<br />

In vitro study<br />

Cell culture: HepG2 cells were cultured in DMEM containing 10% fetal calf serum, 100U/<br />

mL penicillin and 100 µg/mL streptomycin. Cells were plated in 12-well plates at density<br />

of 250000 cells per well. Medium was switched to DMEM without serum one hour prior to<br />

hormone incubations. Cells were treated with dexamethasone (Sigma, Zwijndrecht, NL) and/<br />

or insulin at the indicated concentrations for 24 or 48 hours. Both Angptl4 mRNA expression<br />

and protein levels in the medium were measured. Differences were evaluated using Student’s<br />

t-test.<br />

RESULTS<br />

Baseline characteristics and anthropometrics: Baseline characteristics of the participants<br />

are presented in Table 1. No significant differences were observed between the groups at<br />

baseline. BMI was not changed by either treatment regimen (Table 2) [16].<br />

236


Table 1. Subject characteristics at inclusion.<br />

Placebo Prednisolone<br />

7.5 mg 30 mg P<br />

N 8 12 12<br />

Age (y) 22±3 22±2 22±3 0.740<br />

Weight (kg) 76.9±8.7 74.8±7.2 77.0±5.4 0.250<br />

Height (cm) 184±5.6 183±7.6 186±4 0.641<br />

BMI (kg m-2 ) 22.7±2.1 22.2±1.9 22.4±1.2 0.906<br />

Lean body mass (%) 78.6±7.9 82.1±3.9 80.7±2.5 0.731<br />

Fat mass (%) 16.9±4.7 15.9±2.8 14.8±3.5 0.531<br />

Systolic blood pressure (mmHg) 118±11 125±9 127±9 0.172<br />

Diastolic blood pressure (mmHg) 74±14 72±14 79±13 0.392<br />

Fasting plasma glucose (mmol/l) 5.1±0.3 5.0±0.2 5.0±0.4 0.899<br />

2-hr glucose OGTT (mmol/l) 4.2±0.8 4.7±0.9 4.2±1.1 0.407<br />

Mean±SD are provided. Abbreviations: BMI: body mass index; 2-hr glucose OGTT: plasma glucose<br />

concentrations 2 h after ingestion of 75 g glucose during an oral glucose tolerance test.<br />

Figure 2. Plasma Angptl4 levels before and at 2 weeks treatment with study medication. Angptl4 levels<br />

in the fasted state are depicted for placebo (A), prednisolone 7.5 mg daily (B) and prednisolone 30 mg<br />

daily (C). Median changes from baseline are provided in D (Box-and-Whisker plots; min-max). Prior<br />

to treatment, insulin infusion dose-dependently decreased Angptl4 levels (E). Fasting values were set<br />

at 100% (white bar), Angptl4 levels during step 1 of the clamp (200 pmol/l) are depicted in the grey<br />

bar as % of baseline, and Angptl4 levels during step 2 of the clamp (600 pmol/l) are depicted in the<br />

black bar as % of baseline. The effects of prednisolone treatment on Angptl4 concentrations during<br />

insulin infusion are shown in panel F and G. Panel F shows Angptl4 concentrations during low-dose<br />

insulin infusion, expressed as percentage change from fasting concentrations. White bar represents pretreatment<br />

values and black bar post-treatment values. Fasting values were set at 100%. Panel G shows<br />

Angptl4 concentrations during high-dose insulin infusion, expressed as percentage change from fasting<br />

concentrations. White bar represents pre-treatment values and black bar post-treatment values. Fasting<br />

values were set at 100%. Median and SD are provided in panel (E-G). † p=0.073,*p


Chapter 12 Angptl4 in glucocorticoid-induced diabetogenic effects<br />

Angptl4 levels: Prior to treatment, median Angptl4 levels were 10.8 (6.4-14.0) ng/mL. As<br />

observed previously [3], Angptl4 levels showed considerable inter-individual variability<br />

(Figure 2A-C). At baseline, plasma Angptl4 levels were not related to fasting glucose, insulin,<br />

NEFA and triglyceride levels (data not shown). High-dose prednisolone treatment tended to<br />

reduce plasma Angptl4 levels in the fasted state (Figure 2A-D) (P=0.073). Insulin infusion<br />

dose-dependently decreased plasma Angptl4 levels to 94% and 78% of fasting concentrations<br />

at insulin levels of 171 (153-183) pmol/l and 534 (485-564) pmol/l, respectively (Figure<br />

2E). During low-dose insulin infusion (step 1 of the clamp), Angptl4 levels, expressed as %<br />

of fasting values, were not altered by either dosages of prednisolone (Figure 2F). However,<br />

during high-dose insulin infusion (step 2 of the clamp), insulin-mediated suppression of<br />

Angptl4 levels was blunted by prednisolone treatment (P=0.032) (Figure 2G). This effect<br />

seemed driven by the prednisolone 30 mg arm, but did not reach statistical significance in<br />

posthoc testing (data not shown). In supplemental Table 1, the absolute values of plasma<br />

Angptl4 levels during insulin infusion are depicted.<br />

Prednisolone effects on glucose and lipid metabolism: As published previously [16],<br />

prednisolone treatment dose-dependently impaired glucose and lipid metabolism primarily<br />

by interfering with the metabolic actions of insulin. As such, insulin-stimulated suppression<br />

of EGP and lipolysis and insulin-stimulated glucose disposal were decreased in a dosedependent<br />

manner. In addition, prednisolone 30 mg, but not prednisolone 7.5 mg, increased<br />

fasting glucose, insulin and triglyceride levels (Table 2).<br />

Associations between prednisolone-induced metabolic changes and Angptl4 levels:<br />

Since NEFA are potent activators of Angptl4 expression and plasma NEFAs were shown to<br />

positively correlate with plasma Angptl4 levels in large cohort studies [22], the reduction in<br />

plasma Angptl4 levels by prednisolone and insulin may be a consequence of changes in plasma<br />

NEFA levels, which were lowered by prednisolone and insulin at baseline and during the<br />

clamp, respectively. Indeed, prednisolone-induced changes in fasting NEFA levels correlated<br />

positively with prednisolone-induced alterations in fasting Angptl4 levels (β=0.444; P=0.011;<br />

R2 =0.329). Changes in plasma NEFA levels by insulin infusion, however, were not related to<br />

alterations in Angptl4 levels. Prednisolone-induced changes in plasma Angptl4 levels in the<br />

fasted state were not related to changes in fasting glucose, insulin or triglyceride levels (data<br />

not shown). Furthermore, prednisolone-induced changes in Angptl4 levels during low-and<br />

high-dose insulin infusion were not associated with changes in whole-body lipolysis and Rd<br />

(data not shown).<br />

238


Table 2. Parameters of glucose metabolism and lipolysis before and during treatment with prednisolone 30 mg once daily, prednisolone 7.5 mg once<br />

daily or placebo for a period of two weeks.<br />

Placebo Prednisolone 7.5 mg Prednisolone 30 mg P1 P2 P3<br />

Baseline On-treatment Baseline On-treatment Baseline On-treatment<br />

BMI (kg m-2 ) 23.0±2.3 22.8±2.2 22.3±2.0 22.3±1.8 22.6±1.1 22.6±1.1 0.449 NA NA<br />

FPG (mmol/l) 5.0±0.2 4.8±0.3 4.9±0.2 5.1±0.3 4.7±0.2 5.4±0.3 < 0.0001 0.06 < 0.001<br />

FPI (pmol/l) 30 (16-51) 28 (


Chapter 12 Angptl4 in glucocorticoid-induced diabetogenic effects<br />

Figure 3. Insulin and dexamethasone differentially regulate Angptl4 mRNA and secretion in hepatocytes<br />

in vitro. Human HepG2 hepatoma cells were treated with indicated concentrations of insulin (A)<br />

or dexamethasone (B). Relative changes in Angptl4 mRNA expression (left panel) and the absolute<br />

concentration of Angptl4 in the culture medium (right panel) were measured. C) Human HepG2 hepatoma<br />

cells were treated with indicated concentrations of dexamethasone in the presence or absence of insulin.<br />

Means and SD are provided. *p


is under control of GCs and insulin in healthy humans. Accordingly, it can be hypothesized that<br />

specific metabolic actions of GCs and insulin may be partially accounted for via regulation of<br />

Angptl4.<br />

Recently, it was shown that the Angptl4 gene contains a GRE in its 3’-untranslated region, and<br />

is a direct transcriptional target of GCs in cultured hepatocytes and adipocytes, as well as in<br />

mouse liver and white adipose tissue [15]. Upregulation of Angptl4 mRNA by GCs seemingly<br />

conflicts with data showing that cortisol markedly stimulates LPL activity in human adipose<br />

tissue in vitro [23]. Interestingly, Angptl4-/- mice were more resistant to dexamethasoneinduced<br />

hypertriglyceridemia compared to their wild type littermates, leading the authors<br />

to suggest a role for Angptl4 in the metabolic effects of dexamethasone treatment [15]. Using<br />

human HepG2 cells, we confirmed that dexamethasone increases Angptl4 mRNA expression<br />

as well as protein secretion.<br />

Surprisingly, in the in vivo part of this study, rather than raising fasting Angptl4 levels,<br />

high-dose prednisolone showed a tendency towards lowering Angptl4 levels. It is unlikely<br />

that this reduction is consequential to a direct effect of prednisolone treatment. High-dose<br />

prednisolone treatment induced several other metabolic effects that could have modulated<br />

fasting Angptl4 concentrations. Importantly, prednisolone 30 mg daily induced insulin<br />

resistance, fasting hyperinsulinemia and decreased fasting NEFA concentrations. Since<br />

plasma NEFA concentrations augment Angptl4 concentrations both in vitro and in vivo [1-3],<br />

decreased fasting NEFA concentrations could have therefore led to a net reduction of plasma<br />

Angptl4 concentrations. In support of this notion, in regression analysis, prednisoloneinduced<br />

changes in NEFA independently predicted concomitant changes in Angptl4,<br />

whereas prednisolone treatment was not a significant predictor in this model. We show<br />

that insulin decreases Angptl4 concentrations and the observed increase in fasting insulin<br />

could also have contributed to the net reduction in fasting Angptl4 concentrations following<br />

glucocorticoid treatment. In addition, other, yet unknown, regulators that were not measured<br />

in the present study could have modulated fasting Angptl4 concentrations upon treatment<br />

with prednisolone. It should be emphasized that many aspects of the regulation of Angptl4<br />

concentrations in humans are presently unclear.<br />

Furthermore, we did not find a significant relationship between prednisolone-induced<br />

changes in Angptl4 levels and changes in adipose tissue lipolysis and triglyceride levels, both<br />

in the fasting state and during insulin infusion, suggesting that Angptl4 may not play a role in<br />

the metabolic effects of glucocorticoid treatment.<br />

241<br />

Chapter 12<br />

12


Chapter 12 Angptl4 in glucocorticoid-induced diabetogenic effects<br />

In addition to regulation by NEFA and GCs, Angptl4 is governed by insulin. In our study,<br />

insulin infusion dose-dependently reduced plasma Angptl4 levels. Since insulin infusion<br />

also decreased NEFA levels, it is impossible to conclude whether the decrease in Angptl4 is<br />

due to a direct effect of insulin on Angptl4 levels and/or an indirect effect through reduction<br />

of circulating NEFA. Again using HepG2 cells, it was shown that insulin dose-dependently<br />

reduced expression and secretion of Angptl4, pointing to a direct effect of insulin. Previously,<br />

insulin was already shown to inhibit Angptl4 expression in 3T3-L1 adipocytes [14]. The<br />

mechanism by which insulin regulates Angptl4 remains presently unclear.<br />

Suppression of Angptl4 production in adipocytes and hepatocytes, which likely represent the<br />

primary sites of Angptl4 production in humans [13], may in part explain the stimulatory effect<br />

of insulin on adipose LPL and on postprandial clearance of triglyceride-rich lipoproteins [24].<br />

Conversely, during fasting, the lack of insulin-mediated suppression of Angptl4 production<br />

may explain the decreased LPL activity [25]. Studies are underway using Angptl4-/- mice to<br />

verify the role of Angptl4 in regulation of LPL activity in the feeding-fasting cycle.<br />

The attenuating effect of high-dose prednisolone on the insulin-mediated decrease in Angptl4<br />

levels suggests an interaction between insulin and GCs in regulation of Angptl4 in vivo.<br />

Similarly, in HepG2 cells, dexamethasone prevented the reduction in Angptl4 secretion by<br />

insulin. Considering the low NEFA levels during insulin infusion and given the fact that the<br />

interaction was also observed in vitro, the attenuating effect of GCs is likely independent of<br />

changes in NEFA levels. Conceivably, GCs may interfere with insulin signaling and thereby<br />

reduce the effects of insulin. However, we did not observe an association between changes<br />

in Angptl4 levels and changes in insulin sensitivity, including EGP and Rd, suggesting<br />

independence of changes in insulin signaling.<br />

We conclude that in addition to regulation by NEFAs, plasma Angptl4 is regulated by GCs<br />

and insulin, albeit in opposing ways. Whereas GCs enhance Angptl4 expression and secretion,<br />

both are reduced by insulin. Treatment with GCs attenuated the insulin-mediated decrease in<br />

Angptl4. Determination of the direct independent effect of these hormones on Angptl4 levels<br />

in humans is complicated by their interdependence. Finally, our data suggest that Angtpl4<br />

may not be an important regulator in GC-mediated changes in lipid metabolism in humans<br />

but may contribute to insulin-mediated changes in lipid metabolism.<br />

Funding: This paper was written within the framework of project T1-106 of the Dutch Top<br />

Institute <strong>Pharma</strong>.<br />

242


REFERENCES<br />

1. Staiger H, Haas C, Machann j, Werner R, Weisser M, Schick F, Machicao F, Stefan N, Fritsche A, Haring<br />

HU. Muscle-derived angiopoietin-like protein 4 is induced by fatty acids via peroxisome proliferatoractivated<br />

receptor (PPAR)-delta and is of metabolic relevance in humans. Diabetes. 2009;58(3):579-<br />

89.<br />

2. Lichtenstein L, Kersten S. Modulation of plasma TG lipolysis by Angiopoietin-like proteins and<br />

GPIHBP1. Biochim Biophys Acta. 2010;1801(4):415-20.<br />

3. Kersten S, Lichtenstein L, Steenbergen E, Mudde K, Hendriks HF, Hesselink MK, Schrauwen P, Muller<br />

M. Caloric restriction and exercise increase plasma ANGPTL4 levels in humans via elevated free fatty<br />

acids. Arterioscler Thromb Vasc Biol. 2009;29(6):969-74.<br />

4. Xu A, Lam MC, Chan KW, Wang Y, Zhang j, Hoo RL, Xu jY, Chen B, Chow WS, Tso AW, Lam KS.<br />

Angiopoietin-like protein 4 decreases blood glucose and improves glucose tolerance but induces<br />

hyperlipidemia and hepatic steatosis in mice. Proc Natl Acad Sci U S A. 2005;102(17):6086-91.<br />

5. Koster A, Chao YB, Mosior M, Ford A, Gonzalez-DeWhitt PA, Hale jE, Li D, Qiu Y, Fraser CC, Yang<br />

DD, Heuer jG, jaskunas SR, Eacho P. Transgenic angiopoietin-like (angptl)4 overexpression and<br />

targeted disruption of angptl4 and angptl3: regulation of triglyceride metabolism. Endocrinology.<br />

2005;146(11):4943-50.<br />

6. Mandard S, Zandbergen F, van Straten E, Wahli W, Kuipers F, Muller M, Kersten S. The fasting-induced<br />

adipose factor/angiopoietin-like protein 4 is physically associated with lipoproteins and governs<br />

plasma lipid levels and adiposity. j Biol Chem. 2006;281(2):934-44.<br />

7. Yoshida K, Shimizugawa T, Ono M, Furukawa H. Angiopoietin-like protein 4 is a potent hyperlipidemiainducing<br />

factor in mice and inhibitor of lipoprotein lipase. j Lipid Res. 2002;43(11):1770-2.<br />

8. Romeo S, Pennacchio LA, Fu Y, Boerwinkle E, Tybjaerg-Hansen A, Hobbs HH, Cohen jC. Populationbased<br />

resequencing of ANGPTL4 uncovers variations that reduce triglycerides and increase HDL.<br />

Nat Genet. 2007;39(4):513-6.<br />

9. Sanderson LM, Degenhardt T, Koppen A, Kalkhoven E, Desvergne B, Muller M, Kersten S. Peroxisome<br />

proliferator-activated receptor beta/delta (PPARbeta/delta) but not PPARalpha serves as a plasma<br />

free fatty acid sensor in liver. Mol Cell Biol. 2009;29(23):6257-67.<br />

10. Georgiadi A, Lichtenstein L, Degenhardt T, Boekschoten MV, van Bilsen M, Desvergne B, Muller M,<br />

Kersten S. Induction of cardiac Angptl4 by dietary fatty acids is mediated by peroxisome proliferatoractivated<br />

receptor beta/delta and protects against fatty acid-induced oxidative stress. Circ Res.<br />

2010;106(11):1712-21.<br />

11. Kersten S, Mandard S, Tan NS, Escher P, Metzger D, Chambon P, Gonzalez Fj, Desvergne B, Wahli<br />

W. Characterization of the fasting-induced adipose factor FIAF, a novel peroxisome proliferatoractivated<br />

receptor target gene. j Biol Chem. 2000;275(37):28488-93.<br />

12. Yoon jC, Chickering TW, Rosen ED, Dussault B, Qin Y, Soukas A, Friedman jM, Holmes WE, Spiegelman<br />

BM. Peroxisome proliferator-activated receptor gamma target gene encoding a novel angiopoietinrelated<br />

protein associated with adipose differentiation. Mol Cell Biol. 2000;20(14):5343-9.<br />

243<br />

Chapter 12<br />

12


Chapter 12 Angptl4 in glucocorticoid-induced diabetogenic effects<br />

13. Mandard S, Zandbergen F, Tan NS, Escher P, Patsouris D, Koenig W, Kleemann R, Bakker A, Veenman<br />

F, Wahli W, Muller M, Kersten S. The direct peroxisome proliferator-activated receptor target fastinginduced<br />

adipose factor (FIAF/PGAR/ANGPTL4) is present in blood plasma as a truncated protein<br />

that is increased by fenofibrate treatment. J Biol Chem. 2004;279(33):34411-20.<br />

14. Yamada T, Ozaki N, Kato Y, Miura Y, Oiso Y. Insulin downregulates angiopoietin-like protein 4 mRNA<br />

in 3T3-L1 adipocytes. Biochem Biophys Res Commun. 2006;347(4):1138-44.<br />

15. Koliwad SK, Kuo T, Shipp LE, Gray NE, Backhed F, So AY, Farese RV, jr., Wang jC. Angiopoietin-like 4<br />

(ANGPTL4, fasting-induced adipose factor) is a direct glucocorticoid receptor target and participates<br />

in glucocorticoid-regulated triglyceride metabolism. j Biol Chem. 2009;284(38):25593-601.<br />

16. van Raalte DH, Brands M, van der Zijl Nj, Muskiet MH, Pouwels Pj, Ackermans MT, Sauerwein HP,<br />

Serlie Mj, Diamant M. Low-dose glucocorticoid treatment affects multiple aspects of intermediary<br />

metabolism in healthy humans: a randomised controlled trial. Diabetologia. 2011.<br />

17. Ackermans MT, Ruiter AF, Endert E. Determination of glycerol concentrations and glycerol isotopic<br />

enrichments in human plasma by gas chromatography/mass spectrometry. Anal Biochem.<br />

1998;258(1):80-6.<br />

18. Ackermans MT, Pereira Arias AM, Bisschop PH, Endert E, Sauerwein HP, Romijn JA. The quantification<br />

of gluconeogenesis in healthy men by (2)H2O and [2-(13)C]glycerol yields different results: rates of<br />

gluconeogenesis in healthy men measured with (2)H2O are higher than those measured with [2-<br />

(13)C]glycerol. j Clin Endocrinol Metab. 2001;86(5):2220-6.<br />

19. Finegood DT, Bergman RN, Vranic M. Estimation of endogenous glucose production during<br />

hyperinsulinemic-euglycemic glucose clamps. Comparison of unlabeled and labeled exogenous<br />

glucose infusates. Diabetes. 1987;36(8):914-24.<br />

20. Steele R. Influences of glucose loading and of injected insulin on hepatic glucose output. Ann N Y<br />

Acad Sci. 1959;82:420-30.<br />

21. Smart-Halajko MC, Robciuc MR, Cooper jA, jauhiainen M, Kumari M, Kivimaki M, Khaw KT,<br />

Boekholdt SM, Wareham Nj, Gaunt TR, Day IN, Braund PS, Nelson CP, Hall AS, Samani Nj, Humphries<br />

SE, Ehnholm C, Talmud Pj. The relationship between plasma angiopoietin-like protein 4 levels,<br />

angiopoietin-like protein 4 genotype, and coronary heart disease risk. Arterioscler Thromb Vasc<br />

Biol. 2010;30(11):2277-82.<br />

22. Robciuc MR, Tahvanainen E, jauhiainen M, Ehnholm C. Quantitation of serum angiopoietin-like<br />

proteins 3 and 4 in a Finnish population sample. j Lipid Res. 2010;51(4):824-31.<br />

23. Ottosson M, Vikman-Adolfsson K, Enerback S, Olivecrona G, Bjorntorp P. The effects of cortisol<br />

on the regulation of lipoprotein lipase activity in human adipose tissue. j Clin Endocrinol Metab.<br />

1994;79(3):820-5.<br />

24. Wang H, Eckel RH. Lipoprotein lipase: from gene to obesity. Am j Physiol Endocrinol Metab.<br />

2009;297(2):E271-88.<br />

25. Bergo M, Wu G, Ruge T, Olivecrona T. Down-regulation of adipose tissue lipoprotein lipase<br />

during fasting requires that a gene, separate from the lipase gene, is switched on. j Biol Chem.<br />

2002;277(14):11927-32.<br />

244


SUPPLEMENTAL MATERIALS<br />

Supplemental Table 1. Effect of study medication on absolute plasma Angptl4 levels in the fasted state and during insulin infusion.<br />

Placebo prednisolone 7.5 mg prednisolone 30 mg<br />

Baseline On-treatment Baseline On-treatment Baseline On-treatment<br />

9.5 (8.0-23.6) 11.8 (8.0-18.5) 9.0 (6.9-13.2) 10.8 (6.3-13.4) 8.6 (5.4-19.8) 6.6 (5.9-16.0)<br />

9.1 (7.5-19.2) 10.3 (8.2-17.1) 8.7 (7.0-12.6) 12.0 (7.9-13.5) 9.3 (4.9-17.9) 6.6 (5.1-17.5)<br />

7.8 (5.9-16.9) 8.7 (7.2-16.1) 7.3 (5.8-11.1) 9.2 (7.1-11.6) 5.9 (3.8-14.2) 7.4 (6.2-18.5)<br />

Angptl4 fasting<br />

(ng/mL)<br />

Angptl4 clamp 1<br />

(ng/mL)<br />

Angptl4 clamp 2<br />

(ng/mL)<br />

245<br />

Chapter 12<br />

12


PART<br />

Metabolic Effects of Glucocorticoids and<br />

Interaction with Systemic Inflammation<br />

IV


Chapter 13<br />

Metabolic Effects of High-Dose Prednisolone<br />

Treatment In Early Rheumatoid Arthritis<br />

Balance Between Diabetogenic Effects and Inflammation<br />

Reduction<br />

D. den Uyl, D.H. van Raalte, M.T. Nurmohamed, W.F. Lems, J.W.J.<br />

Bijlsma, J.N. Hoes, B.A.C. Dijkmans and M. Diamant<br />

Arthritis Rheum. 2012;64(3):936-46


Chapter 13 Glucocorticoids in acute rheumatoid arthritis<br />

ABSTRACT<br />

objective: To investigate the dose-related effects of glucocorticoid (GC) treatment on<br />

glucose tolerance, beta-cell function and insulin sensitivity in patients with early active<br />

rheumatoid arthritis (RA).<br />

Methods: A randomized controlled, single-blind trial was conducted in 41 patients with<br />

early active RA. At the beginning of the trial patients had not been treated for their RA and<br />

were randomized to receive prednisolone 60mg daily or prednisolone 30mg daily. Before<br />

and at the end of 1 week of treatment, a frequently-sampled oral glucose tolerance test<br />

was performed. The area under glucose curve (AUC ) was calculated. In addition, beta-cell<br />

G<br />

function and insulin sensitivity parameters were computed.<br />

Results. Patients (age 55.5 ± 14.8 years and 54.2 ± 12.6 years in the prednisolone 60mg<br />

daily and prednisolone 30mg daily groups, respectively; body mass index 24.5 ± 4.1 kg/m2 and 25.4 ± 4.2 kg/m2 , respectively) had active disease at baseline (Disease Activity Score in<br />

44 joints 4.1 ± 0.7 and 4.0 ± 0.8, respectively; C-reactive protein (CRP): 14 (6-34) mg/liter<br />

and 19 (3-39) mg/liter respectively). In addition, 56% of the patients had impaired glucose<br />

tolerance, and 7% were found to have previously unrecognized type 2 diabetes mellitus<br />

(T2DM). Associations of the AUC with erythrocyte sedimentation rate (β=2.430 [95%<br />

G<br />

confidence interval 0.179-4.681], P=0.04) and with CRP (β=2.358 [95% confidence interval<br />

0.210-4.506], P=0.03) were demonstrated. Treatment with prednisolone at both dosages<br />

reduced CRP levels significantly. The incidence of T2DM increased to 24% (P


INTRoDUC<strong>TI</strong>oN<br />

Patients with rheumatoid arthritis (RA) are at increased risk of developing cardiovascular<br />

disease (CVD) [1]. Defects in glucose metabolism may contribute to this increased CVD risk [2].<br />

Two main determinants of glucose metabolism are insulin sensitivity and beta-cell function.<br />

Insulin sensitivity refers to the ability of insulin to reduce plasma glucose by stimulating<br />

its uptake in insulin-sensitive tissues and by suppressing its production in the liver. Betacell<br />

function is commonly referred to as the ability of the pancreatic beta cell to secrete the<br />

appropriate amount of insulin to maintain euglycemia. These processes are closely related: if<br />

insulin sensitivity decreases, a healthy beta cell will respond by augmenting insulin secretion,<br />

to prevent hyperglycemia [3]. In RA patients, impaired insulin sensitivity, as assessed in the<br />

fasting state using the homeostatic model assessment of insulin resistance (HOMA-IR), has<br />

been reported, and this was associated with measures of disease activity and plasma levels<br />

of inflammatory proteins, including C-reactive protein (CRP), tumor necrosis factor α and<br />

interleukin-6 [4, 5].<br />

Glucocorticoids (GCs) are commonly used to treat RA [6]. However, the metabolic effects of GCs<br />

in RA patients are uncertain. On the one hand, short-term clinical trials in healthy individuals<br />

have shown that GCs deteriorate glucose metabolism by reducing hepatic and peripheral<br />

insulin sensitivity [7] and by impairing beta-cell function [8]. In population-based studies,<br />

GC use was associated, in a cumulative dose-dependent manner, with incident diabetes [9]<br />

and need for blood-glucose lowering treatment [10]. In cross-sectional studies in RA patients,<br />

GC exposure was related to impaired fasting insulin sensitivity [11], and tended to predict<br />

development of type 2 diabetes (T2DM) [12, 13]. On the other hand, the use of GCs in chronic<br />

inflammatory conditions may improve glucose tolerance via anti-inflammatory and disease<br />

modifying effects, as has been demonstrated in a number of short-term studies [14, 15].<br />

Previous studies of the effects of GC treatment on parameters of beta-cell function and insulin<br />

sensitivity in RA patients relied solely on measures obtained in the fasting state (i.e., HOMA<br />

of beta-cell function (HOMA-B) and HOMA-IR, respectively) and yielded conflicting results<br />

(for review, see [2]). As mentioned above, since insulin sensitivity and insulin secretion are<br />

interrelated, the use of these HOMA formulas, both of which are based on fasting plasma<br />

glucose and insulin levels, may not be appropriate for discerning changes in insulin sensitivity<br />

and changes in insulin secretion [16]. In addition, fasting parameters provide no information<br />

of the stimulated state. From dynamic tests, such as the frequently-sampled oral glucose<br />

tolerance test (OGTT), indices of glucose-stimulated beta-cell function and postload insulin<br />

sensitivity may be calculated, providing more detailed information on glucose metabolism.<br />

251<br />

Chapter 13<br />

13


Chapter 13 Glucocorticoids in acute rheumatoid arthritis<br />

Therefore, in the present randomized clinical study, we used frequently-sampled OGTTs<br />

to assess the effects of 2 different doses prednisolone, i.e., 60 mg daily and 30 mg daily,<br />

administered for 1 week, on glucose tolerance, beta-cell function and insulin sensitivity in<br />

recently diagnosed untreated RA patients.<br />

RESEARCH DESIGN AND METHoDS<br />

Participants: Patients with early RA were recruited between March 2008 and july 2010 in<br />

three clinics in Amsterdam and Hoorn, the Netherlands. Inclusion criteria were: RA (according<br />

to the revised American College of Rheumatology classification criteria [17]), age ≥ 18 years,<br />

disease duration < 2 years and currently active disease. The latter was defined as the presence<br />

of 6 or more painful and swollen joints, an erythrocyte sedimentation rate (ESR) of ≥ 28<br />

mm/h and/or a global health score of ≥ 20mm (on a visual analogue scale 0-100). Exclusion<br />

criteria were: previous treatment with GCs, DMARDs (other than hydroxychloroquine (HCQ)),<br />

known T2DM, heart failure (i.e. New York Heart Association class 3 and -4 [18]), uncontrolled<br />

hypertension, ALAT/ASAT > 3 times normal values, reduced renal function (serum creatinine<br />

> 150 micromol), contra-indications for GC use, indications of probable tuberculosis and<br />

pregnancy or wish to conceive during the study. Concomitant treatment with nonsteroidal<br />

anti-inflammatory drugs (NSAIDs) during the study was permitted. The study was approved<br />

by an independent ethics committee and was conducted in accordance with the Declaration of<br />

Helsinki. All participants provided written informed consent before participation.<br />

Study design: This study was a randomized, active-controlled, single blind, multicenter trial<br />

and a substudy of a larger study comparing two treatment schedules for the treatment of<br />

early RA (www.controlled-trials.com; ISRCTN55552928). Following assessment of eligibility,<br />

participants were randomized to receive either prednisolone 60 mg once daily (n=21)<br />

or prednisolone 30mg once daily (n=20). On day 0 and day 7, a frequently-sampled OGTT<br />

was performed to measure glucose tolerance, parameters of beta-cell function and insulin<br />

sensitivity. In addition, CRP levels were determined on both study days. On day 7, study<br />

medication was taken 2 hours before the start of the OGTT [19].<br />

Screening visit: At baseline, demographics were recorded and body weight, body mass<br />

index (BMI), waist circumference and blood pressure were determined. Disease activity was<br />

measured by the 44-joint Disease Activity Score (DAS44 [20]). In addition, screening blood<br />

tests were performed, including measurement of ESR, CRP, rheumatoid factor (RF) and anticyclic<br />

citrullinated peptide (anti-CCP).<br />

252


oral glucose tolerance test: The patients visited the research unit at 08.00 AM following an<br />

overnight fast of 10 hours. To obtain venous blood samples, a cannula was inserted in a vein<br />

within the fold of the elbow or in a wrist vein. Patients ingested 75 gram glucose solution,<br />

following which blood was withdrawn for the determination of plasma glucose, insulin and<br />

C-peptide levels at time points 0, 10, 20, 30, 60, 90 and 120 min. C-peptide is secreted by the<br />

pancreatic beta-cell at an equimolar rate with insulin, but is cleared from the circulation by<br />

urinary excretion, whereas insulin clearance is also strongly determined by the liver. Since<br />

insulin clearance may vary considerably between subjects [21], plasma C-peptide levels may<br />

provide more accurate information on beta-cell function than determination of peripheral<br />

insulin levels.<br />

Laboratory measurements: Plasma glucose concentrations were determined using a<br />

hexokinase method (Gluco-quant, Roche Diagnostics). Insulin and C-peptide levels were<br />

determined using an immunometric assay (ADVIA Centaur immunoassay system, Siemens<br />

Medical Solutions Diagnostics). CRP concentration was determined by Immuno turbidometric<br />

method (Roche diagnostics). Anti-CCP levels were determined by second-generation anti-CCP<br />

enzyme-linked immunosorbent assay (ELISA;Axis Shield). The anti-CCP test was performed<br />

according to manufactures instructions with a cut-off level for positivity set at 5 Arbitrary<br />

Units/ml. IgM-RF was measured with an in-house ELISA. The cut-of level was set at 30 IU,<br />

determined on the basis of receiver operator characteristics curves as described previously<br />

[22].<br />

Data analysis: Glucose tolerance state was assessed according to the American Diabetes<br />

Association guidelines [23]. Normal glucose metabolism was defined as fasting plasma<br />

glucose < 5.6 mmol/L and a glucose level < 7.8 mmol/L 2 h following a 75 g OGTT. Impaired<br />

glucose metabolism was defined as FPG between 5.6 – 6.9 mmol/L, or a 2 h OGTT glucose value<br />

between 7.8 mmol/L and 11.0 mmol/L. T2DM was diagnosed with a FPG > 6.9 mmol/L or a<br />

2 h OGTT glucose value >11.0 mmol/L [23]. Area under the 2 h glucose (AUC ), insulin (AUC )<br />

G I<br />

and C-peptide (AUC ) curves were determined by the trapezoidal rule. Insulin sensitivity<br />

CP<br />

during the OGTT was estimated by oral glucose insulin sensitivity (OGIS) index [24] and by<br />

homeostatic model assessment (HOMA-IR) in the fasted state [25]. We calculated empirical<br />

measures of beta-cell function, including total secretion C-peptide secretion divided by<br />

glucose levels (AUC /AUC ratio) and the insulinogenic index (IGI), i.e., (insulin -insulin )/<br />

CP G t=30 t=0<br />

(gluc -gluc ), a marker for early phase insulin secretion. Fasting beta-cell function was<br />

t=30 t=0<br />

measured by HOMA-B [25].<br />

253<br />

Chapter 13<br />

13


Chapter 13 Glucocorticoids in acute rheumatoid arthritis<br />

Statistical analysis: Data are presented as mean ± SD or when distribution was skewed, as the<br />

median and interquartile range (IQR). Differences between the two prednisolone dose groups<br />

were analyzed using the independent-samples t-test or non-parametric Mann-Whitney U test,<br />

as appropriate. Within-group changes were tested using the paired-samples t-test or the nonparametric<br />

Wilcoxon’s signed rank test. The effect of treatment on glucose tolerance state<br />

was analyzed by the Chi-square test. Multivariate regression analysis was used to evaluate the<br />

relation between disease characteristics and activity, and metabolic parameters at baseline,<br />

with correction for age, gender and BMI. Similarly, determinants of prednisolone-induced<br />

changes on AUC , beta-cell function and insulin sensitivity were assessed in multivariate<br />

G<br />

linear regression analyses, with correction for age, gender and BMI. Multivariate logistic<br />

regression analyses were used to associate disease characteristics with progression of<br />

glucose state, also with correction for age, gender and BMI. Pooled analysis was performed<br />

since treatment group was not an effect modifier in any of the tested parameters (this was<br />

evaluated by adding treatment group as an interaction term using multivariate regression<br />

analysis, P-value > 0.1). Correlation analyses included calculation of 95% confidence intervals<br />

(%95 CIs). All statistical analyses were run on SPSS for Windows version 15.0 (SPSS, Chicago,<br />

IL, USA). A P < 0.05 was considered statistically significant.<br />

RESULTS<br />

Characteristics of the study patients: Forty-one consecutive subjects with recent-onset<br />

RA were enrolled. Baseline characteristics are shown in Table 1. Patients were middle-aged<br />

and were not obese. The majority was female, and the median disease duration was 21<br />

weeks. Based on the OGTT at screening, T2DM was diagnosed in 3 patients (7%), and 23<br />

patients (56%) were classified as having impaired glucose metabolism. None of the baseline<br />

characteristics differed significantly between the 2 prednisolone dose groups (Table 1).<br />

Inflammation profiles: Patients had active disease as indicated by DAS44, ESR and CRP values<br />

(Table 1). In multivariate analysis, ESR and CRP plasma levels were positively correlated with<br />

AUC (for ESR, β=2.430 [95% CI 0.179-4.681], P=0.04; for CRP, β=2.358 [95% CI 0.210-4.506],<br />

G<br />

P=0.03). Correcting for ethnicity, NSAID or HCQ usage did not alter the associations (data<br />

not shown). During treatment with prednisolone 60 m daily and with 30 mg daily, plasma<br />

CRP levels decreased to a similar extent, i.e. by 84% (to a median of 2.5 (2.0-2.7) mg/liter;<br />

P


Table 1 Baseline characteristics of the RA study population.<br />

Prednisolone 60 mg<br />

(N=21)<br />

Prednisolone 30 mg<br />

(N=20)<br />

Age (years) 55.5 ±14.8 54.2 ±12.6<br />

Female (%) 62 60<br />

Nonwhite ethnicity (N) 2 0<br />

Symptom duration (weeks) 15 (10-30) 24 (12-52)<br />

DAS44 (no dimension) 4.1±0.7 4.0 ±0.8<br />

ESR (mm/hour) 28 (18-63) 21 (12-44)<br />

CRP (mg/liter) 14 (6-34) 19 (3-39)<br />

RF positive (%) 71 50<br />

Anti-CCP positive (%) 81 55<br />

BMI (kg/m 2 ) 24.5 ±4.1 25.4 ±4.2<br />

Waist circumference (cm) 89 ±15 92 ±13<br />

Current smoking (%) 19 25<br />

Impaired glucose tolerance (N;(%)) 12 (57) 11 (55)<br />

Type 2 diabetes mellitus no (%) 3 (14) 0 (0)<br />

Prior HCQ treatment (N) 1 0<br />

Concurrent NSAID treatment (N) 21 19<br />

Data are mean±SD or median (interquartile range). There were no statistically significant differences<br />

between the 2 treatment groups, as assessed by independent t-test, Mann-Whitney U test, or chi-square<br />

test. RA= rheumatoid arthritis; IQR= interquartile range; DAS44= disease activity score 44 joint count;<br />

ESR= erythrocyte sedimentation rate; CRP= C-reactive protein; RF= rheumatoid factor; anti-CCP= anticyclic<br />

citrullinated peptide; BMI= body mass index; HCQ= hydrochloroquine; NSAID= nonsteroidal antiinflammatory<br />

drug.<br />

Figure 1. oral glucose tolerance test results before treatment (squares connected by solid lines)<br />

and on day 7 of treatment with prednisolone 60 mg daily (circles connected by dashed lines). A,<br />

Glucose concentrations. B, Insulin concentrations. C, C-peptide concentrations. No significant changes<br />

were induced by treatment with prednisolone 60 mg/day. Values are the mean ± SD.<br />

255<br />

Chapter 13<br />

13


Chapter 13 Glucocorticoids in acute rheumatoid arthritis<br />

Table 2. Parameters of beta-cell function and insulin sensitivity during the oral glucose tolerance<br />

test, before and after 7 days prednisolone treatment.<br />

Fasting glucose<br />

(mmol/l)<br />

Fasting insulin<br />

(pmol/l)<br />

Fasting C-peptide<br />

(nmol/l)<br />

AUC G<br />

(mmol/L.120min)<br />

AUC I<br />

(nmol/L.120min)<br />

AUC CP<br />

(nmol/L.120min)<br />

IGI<br />

(no dimension)<br />

AUC CP /AUC G<br />

(no dimensions)<br />

OGIS<br />

(ml/min.m 2 )<br />

HOMA-IR<br />

(no dimension)<br />

HOMA-B<br />

(%)<br />

256<br />

Prednisolone 60 mg/day Prednisolone 30 mg/day<br />

Pre- On-treatment P† Pre- On-treatment P†<br />

treatmenttreatment<br />

5.5±0.7 5.3±0.9 NS 5.6±0.5 5.3±0.6 NS<br />

43 (34-69) 53 (37-77) NS 36 (30-113) 68 (44-129) 0.004<br />

0.5 (0.4-0.7) 0.8 (0.7-1.0) 0.001 0.5 (0.4-0.9) 0.9 (0.6-1.1) 0.001<br />

1051±237 1077±341 NS 1050±172 1031±260 NS<br />

48 (35-74) 46 (32-83) NS 50 (28-95) 63 (33-90) NS<br />

304±92 355±141 NS 339±172 389±180 NS<br />

73 (46-161) 95 (37-284) 0.02 95 (58-148) 127 (65-236) 0.04<br />

0.3 (0.2-0.4) 0.3 (0.2-0.5) NS 0.3 (0.2-0.4) 0.4 (0.2-0.5) 0.02<br />

366 ±64.2 365 ±71.4 NS 358±68 363±68 NS<br />

1.0 (0.7-1.5) 1.1 (0.8-1.6) NS 0.8 (0.7-2.5) 1.5 (1.0-2.8) 0.005<br />

84 (60-113) 104 (76-124) NS 75 (61-122) 119 (77-169) 0.001<br />

Data are mean±SD or median (interquartile range). There were no statistically significant differences<br />

between the 2 treatment groups in the degree of change, as assessed by independent t-test or Mann-<br />

Whitney U test. NS= not significant; AUC = area under the C-peptide curve; AUC = area under the<br />

CP G<br />

glucose curve; AUC = area under the insulin curve; IGI= insulinogenic index; OGIS= oral glucose insulin<br />

I<br />

sensitivity index; HOMA-IR= homeostatic model assessment of insulin resistance; HOMA-B= HOMA of<br />

beta-cell function. † By paired-samples t-test or Wilcoxon’s signed rank test.<br />

Glucose profiles and glucose tolerance: Fasting glucose levels were not altered following<br />

prednisolone treatment (Table 2). Similarly, AUC was not changed following treatment with<br />

G<br />

prednisolone at either 60 mg/day or 30 mg/day (Figure 1A and 2A, Table 2); however, at<br />

the individual level, there was a large variation in response to prednisolone treatment. Seven<br />

patients with impaired glucose metabolism at baseline were classified as having T2DM after<br />

prednisolone treatment, resulting in an increase in the number with T2DM from 3 (7%)<br />

to 10 (24%) (P


(4 in the 30 mg/day group and 5 in the 60 mg/day group). The number of patients who were<br />

first classified as having T2DM was equally divided between the 2 groups (data not shown).<br />

Patients were classified as progressor if the glucose state deteriorated with prednisolone<br />

treatment and as nonprogressor if the glucose state remained unaltered or showed<br />

improvement. Characteristics of the progressor and nonprogressor groups are shown in<br />

Table 3. Disease duration was significantly associated with deterioration of the glucose state<br />

(OR=1.068 [95% CI 1.017-1.122], p=0.009).<br />

Table 3. Disease and anthropometric characteristics of glucometabolic state progressors and<br />

nonprogressors.<br />

Progressors<br />

(n=13)<br />

Nonprogressors<br />

(n=28)<br />

Age (years) 57 ±10.8 53.8 ±14.8<br />

Female (%) 46 68<br />

Non white ethnicity (N) 7 4<br />

Symptom duration (weeks) 36 (12-74)† 16 (8-24)<br />

DAS44 (no dimension) 4.0 ±0.8 4.0 ±0.7<br />

ESR (mm/hour) 33 (16-55) 27 (13-47)<br />

CRP (mg/l) 17 (9-27) 12 (4-39)<br />

RF positive (%) 54 64<br />

Anti-CCP positive (%) 54 75<br />

BMI (kg/m2 ) 25.0 ±4.5 24.9 ±3.9<br />

Waist circumference (cm) 93 ±13 89 ±14<br />

Current smoker (%) 23 25<br />

Prior HCQ treatment (N) 0 1<br />

Concurrent NSAIDs treatment (N) 13 27<br />

Fasting glucose (mmol/l) 5.5 ±0.5 5.5 ±0.7<br />

Fasting insulin (pmol/l) 43 (33-89) 36 (29-74)<br />

Fasting C-peptide (nmol/l) 0.5 (0.3-0.8) 0.6 (0.4-0.8)<br />

HOMA-IR (No dimension) 1.0 (0.7-2.0) 0.8 (0.6-1.8)<br />

HOMA-B (No dimension) 81 (64-120) 82 (60-114)<br />

Data are mean±SD or median (interquartile range). HOMA-IR= homeostatic model assessment of insulin<br />

resistance; HOMA-B= HOMA of the beta cell function. † P=0.04 versus nonprogressors by Mann-Whitney<br />

U test.<br />

Insulin and C-peptide responses during the oGTT: Fasting insulin levels were similar<br />

in the 2 groups prior to treatment. AUC and AUC were not different between the groups<br />

I CP<br />

at baseline (Table 2). Fasting insulin levels were increased following treatment with<br />

prednisolone 30 mg/day, but not with prednisolone 60 mg/day, whereas fasting C-peptide<br />

257<br />

Chapter 13<br />

13


Chapter 13 Glucocorticoids in acute rheumatoid arthritis<br />

levels were increased significantly following prednisolone treatment at either dosage (Table<br />

2). Treatment with prednisolone at either dosage did not significantly affect postload insulin<br />

and C-peptide concentrations (Figure 1B-C, Figure 2B-C, Table 2).<br />

Figure 2: oral glucose tolerance test results before treatment (squares connected by solid lines)<br />

and on day 7 of treatment with prednisolone 30mg daily (circles connected by dashed lines). A,<br />

Glucose concentrations. B, Insulin concentrations. C, C-peptide concentrations. No significant changes<br />

were induced by treatment with prednisolone 30 mg/day. Values are the mean ± SD.<br />

Parameters of beta-cell function: The IGI, as an index for early insulin secretion, increased<br />

during both prednisolone 60 mg/day (P=0.02) and prednisolone 30 mg/day (P=0.04) (Table<br />

2). The ratio of C-peptide levels to glucose levels during the entire OGTT was enhanced<br />

significantly by prednisolone 30 mg/dasy and a near-significant enhancement was observed<br />

with prednisolone 60 mg/day (P=0.07) (Table 2). The amount of increase in parameters of<br />

beta-cell function did not differ between the treatment groups (Table 2).<br />

Insulin sensitivity: Fasting insulin sensitivity (HOMA-IR) was not changed by prednisolone<br />

60 mg/day, but was decreased by prednisolone 30 mg/day (P=0.005). Insulin sensitivity<br />

during the OGTT (OGIS) was not changed by treatment with prednisolone at either 60 mg/<br />

day or 30 mg/day. The degree of change in parameters of insulin sensitivity did not differ<br />

between treatment groups (Table 2).<br />

Determinants of prednisolone-induced changes: Multivariate analysis revealed that<br />

disease duration was positively associated with prednisolone-induced changes in the AUCG (b=3.626 [95% CI 1.077-6.174], P=0.007). Correcting for ethnicity, NSAID or HCQ usage<br />

did not alter this relation (data not shown). No associations of disease characteristics with<br />

changes in IGI or AUC /AUC ratio were identified (data not shown).<br />

CP G<br />

258


DISCUSSIoN<br />

The main findings of the present study are that glucose metabolism is frequently impaired<br />

in patients with early and active RA, probably, at least partly related to inflammation, and<br />

that short-term treatment with high-dose prednisolone does not further deteriorate glycemic<br />

control, as determined in the fasted state and following an oral glucose load. There was,<br />

however, large variability among subjects, with some patients showing improvement, and<br />

others showing deterioration in glucose tolerance. Overall, an increase in patients classified as<br />

having T2DM was observed following 1-week prednisolone treatment. Interestingly, though,<br />

in 9 patients with impaired glucose metabolism at baseline, glucose metabolism reverted to<br />

normal after short-term GC exposure.<br />

The results of this study are clinically important, since GCs are frequently used as<br />

immunosuppressive and anti-inflammatory drugs in the treatment of RA and other<br />

inflammatory rheumatic diseases, such as systemic lupus erythematosus (SLE), polymyalgia<br />

rheumatica, polymyositis and Wegener’s disease, showing powerful effects on disease activity<br />

and radiological progression. However, despite compelling evidence showing the effectiveness<br />

of high-dose GCs in the treatment of early and active RA, rheumatologists are reluctant to<br />

prescribe comparably high dosages, due to fear of serious prednisolone-associated adverse<br />

events, including the development of hyperglycemia and diabetes [26].<br />

Treatment with prednisolone 30 mg daily increased fasting insulin levels in the present study,<br />

but this was not found after treatment with prednisolone 60 mg. Insulin levels are known<br />

to have higher between and within-subject variability than C-peptide levels, as shown in<br />

reproducibility studies [27], which could have contributed to this unexpected finding. Fasting<br />

C-peptide levels, which indicate fasting insulin secretion and are known to be less affected<br />

by high variability, increased significantly in both groups. Furthermore, since clearance of<br />

C-peptide is primarily by the kidney and hepatic clearance is negligible [21], the differences<br />

in fasting insulin levels might be due to altered hepatic clearance of insulin.<br />

In a previous study of healthy individuals performed by our group, high-dose prednisolone<br />

treatment impaired both beta-cell function and insulin sensitivity and deteriorated glucose<br />

tolerance [8]. However, approximately half of the present population, i.e. RA patients with<br />

active disease, had glucometabolic disturbances at baseline: 56% were classified as having<br />

impaired glucose metabolism and 7% fulfilled the criteria of overt T2DM (an increased<br />

percentage as compared to the normal population). This finding is in accordance with the<br />

results of earlier studies in patients with active chronic RA, who showed impaired handling of<br />

intravenously administered glucose compared with matched controls [15, 28].<br />

259<br />

Chapter 13<br />

13


Chapter 13 Glucocorticoids in acute rheumatoid arthritis<br />

Impaired glucose tolerance in RA patients has been mainly attributed to the pro-inflammatory<br />

state. Indeed, in the present study, we demonstrated for the first time that baseline ESR<br />

and plasma CRP levels were positively associated with postload glucose levels. Preclinical<br />

studies suggest that inflammation may impair glucose tolerance both by reducing insulin<br />

sensitivity and by impairing beta-cell function. Several pro-inflammatory cytokines, including<br />

interleukin-6 and tumor necrosis factor α, are known to interfere with the insulin-receptor<br />

signaling cascade resulting in impaired glucose uptake in insulin sensitive tissues [29, 30].<br />

The acute-phase protein CRP was similarly shown to induce insulin resistance by impairment<br />

of insulin signaling in an animal model [31]. Importantly, pro-inflammatory cytokines have<br />

recently been shown to induce beta-cell dysfunction [32]. In addition, chronic inflammation<br />

may reduce beta-cell mass by stimulating apoptosis of beta-cells [33].<br />

While on average no significant effects on fasting and postload glucose were observed, an<br />

increased number of patients met the criteria for T2DM after 1-week treatment with highdose<br />

GCs (with percentage increasing from 7% to 24%). Most of these patients (7 of 10)<br />

had impaired glucose metabolism prior to treatment. Interestingly, 9 patients with impaired<br />

glucose metabolism at baseline were classified as having normal glucose metabolism after<br />

prednisolone treatment, with the numbers equally distributed between the 2 treatment<br />

groups. Although our study was not designed to identify predictors of deterioration of<br />

the glucometabolic state, we found that patients who progressed from normal glucose<br />

metabolism to impaired glucose metabolism or T2DM, or from impaired glucose metabolism<br />

to type 2 DM, had significantly longer disease duration. It is important to note that the use of<br />

the OGTT is hampered by its high within-subject variability [34]. Accordingly, this study does<br />

not allow to distinguish the relative contributions to the observed changes in classification of<br />

the glucometabolic state upon retesting that were attributed to prednisolone treatment per<br />

se, versus the within-subject variability of the OGTT method.<br />

Patients with longest disease duration in the present study not only had the highest<br />

postload glucose levels after treatment, but were also at higher risk of progression in their<br />

glucometabolic classification. These results confirm the findings from preclinical studies<br />

which showed that prolonged inflammation has a significant negative impact on beta-cell<br />

function [32]. Although the disease duration in all patients in our study is relatively short,<br />

plasma CRP levels have been shown to be increased years before the first clinical symptoms of<br />

RA [35], and a cumulative effect of systemic inflammation can be expected. While we observed<br />

that short-term treatment with high-dose prednisolone does not further deteriorate overall<br />

glycemic control, an additional effect of GCs might further disrupt already impaired beta<br />

cells in patients with long disease duration. These observations suggest that treatment in<br />

260


RA patients should be started early in the course of the disease, not only to slow down the<br />

radiological progression of the disease [36], but also to limit the development of comorbid<br />

conditions such as metabolic and cardiovascular disease.<br />

Another potential explanation for the present findings is the possible contribution of<br />

interindividual responses to GC treatment, due to genetic variation in the glucocorticoid<br />

receptor (GR) [37]. Given these differential metabolic responses to high dose prednisolone,<br />

further characterization of progressors versus nonprogressors might better identify<br />

susceptible subjects. In clinical practice, regularly follow-up measurement of plasma glucose<br />

levels and glycosylated hemoglobin levels during GC treatment is recommended.<br />

It was of interest that no dose-dependent effects of prednisolone with regard to the effects on<br />

glucose tolerance, beta-cell function and insulin sensitivity were demonstrated in the present<br />

study. Results of previous studies indicated that the effects of GCs on glucose metabolism were<br />

dose-dependent [10, 11]. However, both dosages of prednisolone used in the present study<br />

are considered to be high [38], and although precise data on a dose-dependent occupation of<br />

the GR is lacking, we hypothesize that similar receptor saturation might explain the fact that<br />

we showed no differences between the 2 treatment groups.<br />

This study has a number of limitations. First, it provides no information regarding the longterm<br />

effects of high-dose GCs on glucose metabolism. However, in clinical practice, the GC<br />

dosage is rapidly tapered and patients are at low maintenance dose levels within 2 months.<br />

Whether the effects shown in this study will be sustained in the long term needs to be<br />

determined.<br />

Second, we used surrogate measures for beta-cell function and insulin sensitivity, as<br />

derived from the OGTT. The ‘gold’ standards for measurement of beta-cell function and<br />

insulin sensitivity are the hyperglycemic clamp and the hyperinsulinemic-euglycemic<br />

clamp procedures, respectively. These tests are rather laborious and were considered too<br />

demanding in this population. In addition, the measures derived from the OGTT as presented<br />

in the manuscript are well validated against the clamp methods [24] and suitable for followup<br />

measurements. Another advantage of the OGTT is that glucose tolerance and parameters<br />

of both beta-cell function and insulin sensitivity can be derived from a single test.<br />

Finally, we measured whole-body insulin sensitivity by HOMA-IR and OGIS, which does not<br />

allow to tease out the various influences of prednisolone on different tissues. Using stable<br />

isotopes, it was shown that prednisolone treatment may induce insulin resistance to a<br />

261<br />

Chapter 13<br />

13


Chapter 13 Glucocorticoids in acute rheumatoid arthritis<br />

different extent in the various insulin-sensitive tissues, including liver, skeletal muscle and<br />

adipose tissue [39]. In our study we were unable to address this topic.<br />

We conclude that short-term treatment with prednisolone at 60mg daily or 30mg daily<br />

did not cause deterioration of the glucometabolic state in patients with active RA. Our data<br />

suggest that there is a balance in the diabetogenic effects and anti-inflammatory effects of GC<br />

treatment in early active RA patients, making short-term exposure to high-dose prednisolone<br />

a safe treatment option for most patients, from a metabolic viewpoint. Due to individual<br />

differences, however, regular glucose monitoring is recommended.<br />

Acknowledgements<br />

This research was performed within the framework of project T1-106 of the Dutch Top<br />

Institute <strong>Pharma</strong>.<br />

262


REFERENCES<br />

1. Peters Mj, van Halm VP, Voskuyl AE, Smulders YM, Boers M, Lems WF, Visser M, Stehouwer CD,<br />

Dekker jM, Nijpels G, Heine R, Dijkmans BA, Nurmohamed MT. Does rheumatoid arthritis equal<br />

diabetes mellitus as an independent risk factor for cardiovascular disease? A prospective study.<br />

Arthritis Rheum. 2009;61(11):1571-9.<br />

2. Wasko MC, Kay j, Hsia EC, Rahman MU. Diabetes mellitus and insulin resistance in patients with<br />

rheumatoid arthritis: risk reduction in a chronic inflammatory disease. Arthritis Care Res (Hoboken).<br />

2011;63(4):512-21.<br />

3. Weyer C, Bogardus C, Mott DM, Pratley RE. The natural history of insulin secretory dysfunction and<br />

insulin resistance in the pathogenesis of type 2 diabetes mellitus. j Clin Invest. 1999;104(6):787-94.<br />

4. Chung CP, Oeser A, Solus jF, Gebretsadik T, Shintani A, Avalos I, Sokka T, Raggi P, Pincus T, Stein CM.<br />

Inflammation-associated insulin resistance: differential effects in rheumatoid arthritis and systemic<br />

lupus erythematosus define potential mechanisms. Arthritis Rheum. 2008;58(7):2105-12.<br />

5. Dessein PH, joffe BI. Insulin resistance and impaired beta cell function in rheumatoid arthritis.<br />

Arthritis Rheum. 2006;54(9):2765-75.<br />

6. Gorter SL, Bijlsma jW, Cutolo M, Gomez-Reino j, Kouloumas M, Smolen jS, Landewe R. Current<br />

evidence for the management of rheumatoid arthritis with glucocorticoids: a systematic literature<br />

review informing the EULAR recommendations for the management of rheumatoid arthritis. Ann<br />

Rheum Dis. 2010;69(6):1010-4.<br />

7. van Raalte DH, Ouwens DM, Diamant M. Novel insights into glucocorticoid-mediated diabetogenic<br />

effects: towards expansion of therapeutic options? Eur j Clin Invest. 2009;39(2):81-93.<br />

8. van Raalte DH, Nofrate V, Bunck MC, van Iersel T, Elassaiss Schaap j, Nassander UK, Heine Rj, Mari<br />

A, Dokter WH, Diamant M. Acute and 2-week exposure to prednisolone impair different aspects of<br />

beta-cell function in healthy men. Eur j Endocrinol. 2010;162(4):729-35.<br />

9. Gulliford MC, Charlton j, Latinovic R. Risk of diabetes associated with prescribed glucocorticoids in<br />

a large population. Diabetes Care. 2006;29(12):2728-9.<br />

10. [10] Gurwitz jH, Bohn RL, Glynn Rj, Monane M, Mogun H, Avorn j. Glucocorticoids and the risk for<br />

initiation of hypoglycemic therapy. Arch Intern Med. 1994;154(1):97-101.<br />

11. Dessein PH, joffe BI, Stanwix AE, Christian BF, Veller M. Glucocorticoids and insulin sensitivity in<br />

rheumatoid arthritis. j Rheumatol. 2004;31(5):867-74.<br />

12. van Tuyl LH, Boers M, Lems WF, Landewe RB, Han H, van der Linden S, van de Laar M, Westhovens<br />

R, van Denderen jC, Westedt ML, Peeters Aj, jacobs P, Huizinga TW, van de Brink H, Dijkmans BA,<br />

Voskuyl AE. Survival, comorbidities and joint damage 11 years after the COBRA combination therapy<br />

trial in early rheumatoid arthritis. Ann Rheum Dis. 2010;69(5):807-12.<br />

13. Wolfe F, Michaud K. Severe rheumatoid arthritis (RA), worse outcomes, comorbid illness,<br />

and sociodemographic disadvantage characterize ra patients with fibromyalgia. J Rheumatol.<br />

2004;31(4):695-700.<br />

263<br />

Chapter 13<br />

13


Chapter 13 Glucocorticoids in acute rheumatoid arthritis<br />

14. Hallgren R, Berne C. Glucose intolerance in patients with chronic inflammatory diseases is<br />

normalized by glucocorticoids. Acta Med Scand. 1983;213(5):351-5.<br />

15. Svenson KL, Lundqvist G, Wide L, Hallgren R. Impaired glucose handling in active rheumatoid<br />

arthritis: effects of corticosteroids and antirheumatic treatment. Metabolism. 1987;36(10):944-8.<br />

16. Wallace TM, Levy jC, Matthews DR. Use and abuse of HOMA modeling. Diabetes Care.<br />

2004;27(6):1487-95.<br />

17. Arnett FC, Edworthy SM, Bloch DA, McShane Dj, Fries jF, Cooper NS, Healey LA, Kaplan SR, Liang MH,<br />

Luthra HS, et al. The American Rheumatism Association 1987 revised criteria for the classification<br />

of rheumatoid arthritis. Arthritis Rheum. 1988;31(3):315-24.<br />

18. The Criteria Committee of the New York Heart Association. Nomenclature and Criteria for Diagnosis<br />

of Diseases of the Heart and Great Vessels. 9th ed. Boston: Little, Brown & Co; 1994: 253-6.<br />

19. Boers M, Verhoeven AC, Markusse HM, van de Laar MA, Westhovens R, van Denderen jC, van<br />

Zeben D, Dijkmans BA, Peeters Aj, jacobs P, van den Brink HR, Schouten Hj, van der Heijde DM,<br />

Boonen A, van der Linden S. Randomised comparison of combined step-down prednisolone,<br />

methotrexate and sulphasalazine with sulphasalazine alone in early rheumatoid arthritis. Lancet.<br />

1997;350(9074):309-18.<br />

20. van der Heijde DM, van ‘t Hof MA, van Riel PL, Theunisse LA, Lubberts EW, van Leeuwen MA, van<br />

Rijswijk MH, van de Putte LB. judging disease activity in clinical practice in rheumatoid arthritis:<br />

first step in the development of a disease activity score. Ann Rheum Dis. 1990;49(11):916-20.<br />

21. Kotronen A, Vehkavaara S, Seppala-Lindroos A, Bergholm R, Yki-jarvinen H. Effect of liver fat on<br />

insulin clearance. Am j Physiol Endocrinol Metab. 2007;293(6):E1709-15.<br />

22. Nielen MM, van Schaardenburg D, Reesink HW, van de Stadt Rj, van der Horst-Bruinsma IE, de<br />

Koning MH, Habibuw MR, Vandenbroucke JP, Dijkmans BA. Specific autoantibodies precede the<br />

symptoms of rheumatoid arthritis: a study of serial measurements in blood donors. Arthritis<br />

Rheum. 2004;50(2):380-6.<br />

23. American Diabetes A. Standards of medical care in diabetes--2011. Diabetes Care. 2011;34 Suppl<br />

1:S11-61.<br />

24. Mari A, Ferrannini E. Beta-cell function assessment from modelling of oral tests: an effective<br />

approach. Diabetes Obes Metab. 2008;10 Suppl 4:77-87.<br />

25. Levy jC, Matthews DR, Hermans MP. Correct homeostasis model assessment (HOMA) evaluation<br />

uses the computer program. Diabetes Care. 1998;21(12):2191-2.<br />

26. van Tuyl LH, Plass AM, Lems WF, Voskuyl AE, Dijkmans BA, Boers M. Why are Dutch rheumatologists<br />

reluctant to use the COBRA treatment strategy in early rheumatoid arthritis? Ann Rheum Dis.<br />

2007;66(7):974-6.<br />

27. Mooy jM, Grootenhuis PA, de Vries H, Kostense Pj, Popp-Snijders C, Bouter LM, Heine Rj. Intraindividual<br />

variation of glucose, specific insulin and proinsulin concentrations measured by two<br />

oral glucose tolerance tests in a general Caucasian population: the Hoorn Study. Diabetologia.<br />

1996;39(3):298-305.<br />

264


28. Svenson KL, Lundqvist G, Wide L, Hallgren R. Impaired glucose handling in active rheumatoid<br />

arthritis: relationship to the secretion of insulin and counter-regulatory hormones. Metabolism.<br />

1987;36(10):940-3.<br />

29. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444(7121):860-7.<br />

30. Nieto-Vazquez I, Fernandez-Veledo S, Kramer DK, Vila-Bedmar R, Garcia-Guerra L, Lorenzo M. Insulin<br />

resistance associated to obesity: the link TNF-alpha. Arch Physiol Biochem. 2008;114(3):183-94.<br />

31. Xi L, Xiao C, Bandsma RH, Naples M, Adeli K, Lewis GF. C-reactive protein impairs hepatic insulin<br />

sensitivity and insulin signaling in rats: role of mitogen-activated protein kinases. Hepatology.<br />

2011;53(1):127-35.<br />

32. Donath MY, Boni-Schnetzler M, Ellingsgaard H, Ehses JA. Islet inflammation impairs the pancreatic<br />

beta-cell in type 2 diabetes. Physiology (Bethesda). 2009;24:325-31.<br />

33. Gurzov EN, Germano CM, Cunha DA, Ortis F, Vanderwinden jM, Marchetti P, Zhang L, Eizirik DL.<br />

p53 up-regulated modulator of apoptosis (PUMA) activation contributes to pancreatic beta-cell<br />

apoptosis induced by proinflammatory cytokines and endoplasmic reticulum stress. J Biol Chem.<br />

2010;285(26):19910-20.<br />

34. Owens DR, Wragg KG, Biggs PI, Luzio S, Davies Cj, jones MK. The reproducibility of serial meal and<br />

oral glucose tolerance tests in normal subjects. Diabete Metab. 1981;7(1):25-33.<br />

35. Nielen MM, van Schaardenburg D, Reesink HW, Twisk jW, van de Stadt Rj, van der Horst-Bruinsma<br />

IE, de Gast T, Habibuw MR, Vandenbroucke jP, Dijkmans BA. Increased levels of C-reactive<br />

protein in serum from blood donors before the onset of rheumatoid arthritis. Arthritis Rheum.<br />

2004;50(8):2423-7.<br />

36. Bukhari MA, Wiles NJ, Lunt M, Harrison BJ, Scott DG, Symmons DP, Silman AJ. Influence of diseasemodifying<br />

therapy on radiographic outcome in inflammatory polyarthritis at five years: results from<br />

a large observational inception study. Arthritis Rheum. 2003;48(1):46-53.<br />

37. van Rossum EF, Lamberts SW. Polymorphisms in the glucocorticoid receptor gene and their<br />

associations with metabolic parameters and body composition. Recent Prog Horm Res. 2004;59:333-<br />

57.<br />

38. Buttgereit F, da Silva jA, Boers M, Burmester GR, Cutolo M, jacobs j, Kirwan j, Kohler L, Van Riel<br />

P, Vischer T, Bijlsma jW. Standardised nomenclature for glucocorticoid dosages and glucocorticoid<br />

treatment regimens: current questions and tentative answers in rheumatology. Ann Rheum Dis.<br />

2002;61(8):718-22.<br />

39. van Raalte DH, Brands M, van der Zijl Nj, Muskiet MH, Pouwels Pj, Ackermans MT, Sauerwein HP,<br />

Serlie Mj, Diamant M. Low-dose glucocorticoid treatment affects multiple aspects of intermediary<br />

metabolism in healthy humans: a randomised controlled trial. Diabetologia. 2011;54(8):2103-12.<br />

265<br />

Chapter 13<br />

13


Chapter 14<br />

Glucose Tolerance, Insulin Sensitivity and<br />

Beta-Cell Function in Rheumatoid Arthritis<br />

Patients Treated With or Without Low-to-<br />

Medium Dose Glucocorticoids.<br />

J.N. Hoes, M.C. van der Goes, D.H. van Raalte, N.J. van der Zijl, D.<br />

den Uyl, W.F. Lems, F.P.G.J. Lafeber, J.W.G. Jacobs, P.M.J. Welsing, M.<br />

Diamant and J.W.J. Bijlsma<br />

Ann Rheum Dis. 2011;70(11):1887-94


Chapter 14 Glucocorticoids in chronic rheumatoid arthritis<br />

ABSTRACT<br />

objective: To compare glucose tolerance and parameters of insulin sensitivity and betacell<br />

function between chronic glucocorticoid (GC)-using and GC-naive RA patients.<br />

Research design and methods: Frequently-sampled 75-g oral glucose tolerance tests were<br />

performed in 58 chronic GC-using and 82 GC-naive RA patients with established disease,<br />

known type 2 diabetes mellitus (T2DM) precluded participation, and 50 control subjects<br />

of comparable age with normal glucose tolerance. The associations between cumulative<br />

GC dose and disease characteristics and glucose tolerance state, insulin sensitivity and<br />

beta-cell function were tested using multivariate linear and logistic regression models,<br />

correcting for patient characteristics.<br />

Results: Glucose tolerance state, insulin sensitivity and beta-cell function did not differ<br />

between both RA populations; we detected de novo T2DM in 11% and impaired glucose<br />

metabolism in 35% of RA patients. Within RA patients, cumulative GC dose was associated<br />

with T2DM, which seemed mostly driven by the effects of cumulative GC dose on insulin<br />

resistance; however, the association decreased when corrected for current disease activity.<br />

RA patients had decreased insulin sensitivity and impaired beta-cell function compared to<br />

controls, and multivariate regression analyses showed a negative association between the<br />

presence of RA and insulin sensitivity.<br />

Conclusions: GC-using and GC-naive RA patients had comparable metabolic parameters,<br />

and had decreased insulin sensitivity and beta-cell function as compared to healthy controls.<br />

Although cumulative GC dose was shown to have a negative impact on glucose tolerance<br />

state and insulin sensitivity, confounding by indication remains the main challenge in this<br />

cross-sectional analysis.<br />

268


INTRoDUC<strong>TI</strong>oN<br />

Rheumatoid arthritis (RA) patients are at increased risk to develop cardiovascular disease,<br />

comparable to the risks observed in subjects with type 2 diabetes mellitus (T2DM) [1].<br />

Additional impairment in glucose metabolism may contribute significantly to the accelerated<br />

atherogenesis in RA patients [2]. Fasting and postprandial glucose metabolism are<br />

determined by beta-cell function (insulin production) and by the peripheral effects of insulin<br />

(insulin sensitivity) aimed to increase glucose uptake in skeletal muscle and to decrease<br />

glucose production by the liver. Glucocorticoids (GCs) impair hepatic and peripheral insulin<br />

sensitivity and induce beta-cell dysfunction, however, the precise underlying mechanisms<br />

are still being investigated [3, 4]. Previously, RA patients were shown to have impaired<br />

fasting insulin sensitivity (homeostatic model assessment (HOMA)-IR) and fasting beta-cell<br />

function (HOMA-B), which correlated with disease activity and markers of inflammation [5-<br />

7]. Consequently, prevalent diabetes was estimated to be up to 15-19% in RA patients [8, 9],<br />

an increased number as compared to the estimated T2DM prevalence of 4-8% in middle-aged<br />

men and women in the general population [10].<br />

The role of GCs in glucose intolerance in RA patients has been one of paradox. On the one<br />

hand, in animal models and in short-term clinical trials in healthy subjects, GCs were shown<br />

to deteriorate glucose metabolism by impairing hepatic and peripheral insulin sensitivity<br />

and by inducing beta-cell dysfunction [11]. In retrospective, population-based studies, GC<br />

therapy was associated with incident diabetes [12], and the need for blood-glucose lowering<br />

treatment in a cumulative dose-dependent way [13]. In retrospective studies in RA patients,<br />

GC exposure was shown to correlate with insulin resistance [14] and to predict diabetes<br />

[15]. On the other hand, the use of GCs in chronic inflammatory states may improve glucose<br />

tolerance by their anti-inflammatory effects, as was demonstrated in a number of short-term<br />

studies using GC-treatment [16, 17]; this was also shown in a study using methotrexate [18].<br />

In addition, confounding by indication should be kept in mind when evaluating the relation<br />

between GC use and glucose tolerance in RA patients in observational studies. This is the<br />

possibility that patients with higher cumulative inflammation (i.e. higher disease activity),<br />

resulting in a priori increased insulin resistance, were more likely to be given high-dose GCs<br />

than those with less inflammation (disease) activity. Thus, the impact of GC-treatment on<br />

glucose metabolism in RA patients requires further clarification.<br />

Previous studies that have addressed the effects of GCs on glucose tolerance, insulin sensitivity<br />

and beta-cell function in RA patients included a small number of patients [16], or relied solely<br />

on fasting parameters, i.e. HOMA-IR and HOMA-B [14]. As mentioned above, in as much as<br />

269<br />

Chapter 14<br />

14


Chapter 14 Glucocorticoids in chronic rheumatoid arthritis<br />

insulin sensitivity and insulin secretion are interrelated, the use of the HOMA formulas, both<br />

of which utilize the same fasting variables, i.e. fasting plasma insulin and glucose, may not be<br />

appropriate to discern changes in insulin sensitivity from those in insulin secretion [19]. For<br />

instance, if two subjects have the same fasting glucose level, but, one persons achieves this<br />

with higher fasting insulin levels, this person is more insulin resistant (thus has lower insulin<br />

sensitivity), which is expressed by a higher HOMA-IR score. Similarly, from fasting glucose and<br />

insulin levels, a HOMA-B score is calculated which gives an impression on beta-cell function.<br />

Although these model-derived indices are well-validated, they provide no information about<br />

the stimulated, postload state [19]. From dynamic tests, such as the frequently-sampled oral<br />

glucose tolerance test (OGTT), indices of postload insulin sensitivity and glucose-stimulated<br />

beta-cell function may be calculated, in order to provide more detailed information on glucose<br />

metabolism [20].<br />

Therefore, in the present study, we compared glucose tolerance and (fasting and dynamic)<br />

parameters of insulin sensitivity and beta-cell function from frequently-sampled OGTTs in a<br />

large group of chronic GC-using RA patients versus GC-naive RA patients. Furthermore, we<br />

included a control group of comparable age to create a perspective of our OGTT findings in RA<br />

patients, and to assess the association of RA per se on measures of insulin sensitivity and betacell<br />

function in subjects with normal glucose tolerance. Finally, we assessed the association<br />

between cumulative GC-dose and disease characteristics with these metabolic parameters.<br />

METHoDS<br />

Population: RA patients with established disease, i.e. defined as having a disease duration<br />

of more than 2 years, were recruited in 5 rheumatology clinics in the region of Utrecht,<br />

the Netherlands. Patients were either both current and chronic GC-users (RA+GC), which<br />

indicated GC treatment for at least 3 months, or they were GC usage naïve (RA-GC). Known<br />

T2DM (defined as receiving treatment) was an exclusion criterion. We included a control<br />

group (controls) with normal glucose tolerance and without first-degree relatives with T2DM,<br />

consisting of individuals who had undergone an OGTT for screening purposes for other studies<br />

at the Diabetes Center of the VU University Medical Center in Amsterdam. Accordingly, this<br />

group consisted of relatively overweight predominantly male individuals. We did not match<br />

the control population to the RA population, since our main focus was on studying the effects<br />

of GCs in RA patients (particularly compared to GC-naive RA patients). The healthy control<br />

group in our study served to give a perspective on the values obtained in the RA patients. An<br />

270


independent ethics committee approved the study and all subjects provided written informed<br />

consent before participation; the protocol was according to the ‘Declaration of Helsinki’.<br />

Protocol: Participants visited the clinic following an overnight fast of minimally 10 hours.<br />

A physical examination, including recording of length, weight and waist circumference<br />

was performed and fasting blood tests were acquired in all patients. In the RA patients,<br />

in addition, the disease activity score (DAS28 and DAS28-CRP) [21, 22] was calculated; in<br />

addition, DMARD-history taking, anti-citrullinated protein antibodies (ACPA) laboratory<br />

measurement, and X-rays of hands and feet (to detect erosive damage) were performed.<br />

Finally, all participants underwent a 2-hr 75 g OGTT. Blood samples for determination of<br />

glucose, insulin and C-peptide were collected at times 0, 10, 20, 30, 60, 90, and 120 minutes,<br />

starting immediately after the ingestion of the 75 g glucose solution. Since insulin clearance<br />

may vary considerably between subjects [23], plasma C-peptide levels may provide additional<br />

information on beta-cell function.<br />

Analytical determinations: Plasma glucose was measured using a chemical technique on<br />

a DXC-800 analyser (Beckman Coulter, Los Angeles, USA). Plasma insulin was measured<br />

using an immunometric technique on an Immulite 1000 Analyzer (Siemens Medical<br />

Solutions Diagnostics, Los Angeles, USA). Plasma C-peptide was measured using an<br />

electrochemiluminescence immunoassay on the Modular E170 (Roche Diagnostics GmbH,<br />

D-68298 Mannheim, Germany).<br />

Data analysis: Normal glucose tolerance was defined as fasting plasma glucose (FPG) < 6.1<br />

and a 2-hour glucose value < 7.8 mM; impaired glucose metabolism (IGM) as FPG between 6.1<br />

and 7.1 mM or a 2-hour glucose value between 7.8 mM and 11.1 mM; T2DM as FPG > 7.1 mM<br />

or a 2-hour glucose value > 11.1 mM. Area under the 2-hour glucose (AUC ), insulin (AUC ),<br />

gluc ins<br />

and C-peptide (AUC ) curves were determined using the trapezoidal rule. Insulin sensitivity<br />

c-pep<br />

in the fasted state was computed by HOMA-IR [24]. Estimated metabolic clearance rate<br />

(MCR /Stumpvoll Index) and oral glucose insulin sensitivity (OGIS) were used to estimate<br />

est<br />

postload insulin sensitivity [25]. Various measures of beta-cell function were calculated:<br />

HOMA-B was derived from fasting measures [24]. Dynamic measures of beta-cell function<br />

were derived from OGTT data and included: AUC /AUC ratio over the 2-hour period<br />

c-pep gluc<br />

and the insulinogenic index (IGI): insulin -insulin )/(gluc -gluc ), as measure for early<br />

t=30 t=0 t=30 t=0<br />

insulin secretion [26]. The oral disposition index (DI) was calculated by multiplying IGI and<br />

OGIS, to adjust insulin secretion for insulin sensitivity. Insulin clearance was calculated by<br />

dividing AUC and AUC [23].<br />

c-pep ins<br />

271<br />

Chapter 14<br />

14


Chapter 14 Glucocorticoids in chronic rheumatoid arthritis<br />

Statistical analysis:<br />

Comparison of parameters of glucose tolerance state, insulin sensitivity and beta-cell function<br />

of RA+-GC groups and controls:<br />

Data were presented as mean values ± SD or as median (interquartile range) in case of nonnormal<br />

distribution. Intergroup differences in continuous outcomes were tested by ANOVA,<br />

and with the Kruskal-Wallis tests in case of non-normal distribution. Differences between<br />

groups in dichotomous outcomes were tested with the chi-square test. Post-hoc Bonferroni<br />

correction was applied in case of multiple testing by multiplying the p-value times 2 (3 groups<br />

minus 1).<br />

Associations between patient and disease characteristics and parameters of glucose tolerance<br />

state, insulin sensitivity and beta-cell function:<br />

Associations between known determinants of insulin sensitivity and beta-cell function (age,<br />

waist circumference, BMI, and insulin clearance) and gender, and, within the RA populations,<br />

cumulative and daily GC-dose and disease activity (DAS28 and its components, DMARD-use,<br />

hand or feet erosions on X-ray, ACPA) and parameters of insulin sensitivity and beta-cell<br />

function were investigated with linear regression analysis; associations of the above factors<br />

with IGM and T2DM were investigated with logistic regression analyses. Non-normally<br />

distributed variables were log-transformed when used in multivariate linear regression<br />

analysis. In the multivariate analyses with OGTT-outcomes as dependent variable where the<br />

RA populations were compared with the controls, RA patients with previously unknown<br />

T2DM, IGM or with 1st degree relatives with T2DM were excluded (i.e. fitting the exclusion<br />

criteria of controls). SPSS for Mac version 16.0 (SPSS, Chicago, IL, USA) was used for all<br />

statistical analyses. A p-value


Glucose tolerance state: The prevalence of previously unknown T2DM was comparable<br />

between the two RA groups (Table 1). If those RA-patients who were excluded because of<br />

known T2DM (n=27) were included, then the prevalence of T2DM would be 19% (RA-GC 18%<br />

vs. RA+GC 21%, P=0.9). Within the RA groups, both cumulative and daily prednisolone dose<br />

was associated with incident T2DM in univariate analyses (odds ratio cumulative dose (g):<br />

1.04; P=0.002; daily dose (mg): 1.13; P=0.048). This association sustained after adjusting for<br />

disease activity and patient characteristics (DAS28, ACPA, erosions, DMARD history, disease<br />

duration, age, BMI, waist circumference, and gender; odds ratio cumulative dose (g): 1.04;<br />

P= 0.03; daily dose (g): 1.15; P=0.13), whereas it was decreased and less significant (a trend)<br />

after adjustment for current disease activity alone and patient characteristics (DAS28, age,<br />

BMI, waist circumference, and gender; odds ratio cumulative GC-dose (g): 1.02; P= 0.08; daily<br />

dose (g): 1.11; P=0.3).<br />

Metabolic responses during oGTT: Glucose levels during the OGTT were not different<br />

between the RA groups, whereas AUC gluc was higher in RA patients as compared to controls,<br />

which was mostly driven by higher glucose levels during the final 90 minutes of the test (Figure<br />

1A). Insulin levels were comparable between all groups (Figure 1B); however, C-peptide<br />

secretion was higher in the RA groups as compared to controls (Figure 1C), with no difference<br />

between the RA groups, indicating increased insulin clearance in RA patients as compared to<br />

controls (data not shown). Insulin clearance was decreased in RA+GC as compared to RA-GC<br />

(data not shown).<br />

Figure 1. Glucose and insulin levels during the oral glucose tolerance test (oGTT). Mean (±SD) for<br />

glucose and C-peptide levels during the OGTT for 1) control subjects, 2) glucocorticoid (GC)-naive RA<br />

patients and 3) GC-using RA patients are shown. *P


Chapter 14 Glucocorticoids in chronic rheumatoid arthritis<br />

Table 1. Baseline characteristics.<br />

vs. RA- vs.<br />

Controls RA-GCs RA+GCs GCs RA+GCs<br />

N 50 82 58 -<br />

Age (years) 56 ± 8 57 ± 12 59 ± 12 1.0 0.4<br />

274<br />

P-value Controls*<br />

Female (%) 38 71 71


Data represent means ± standard deviation or median (interquartile range) when data was not normally<br />

distributed. Intergroup differences in continuous outcomes were tested by ANOVA, and with both the<br />

Kruskal-Wallace and Mann-Whitney tests in case of non-normal distribution. Differences between groups<br />

in dichotomous outcomes were tested with the chi-square test. Post-hoc Bonferroni correction was<br />

applied in case of multiple testing (p-value times 2).<br />

$ p-Value controls is the intergroup difference tested by ANOVA, Mann-Whitney or chi-square test with the<br />

Bonferroni post-hoc test. p Values of the RA-GC RA+GC difference was not depicted; the only significant/<br />

trend differences were male HDL P=0.07; dyslipidaemia P=0.009.<br />

* Increased waist circumference was defined as > 102 cm in male and > 88 cm in female. Hypertension<br />

was defined as ≥ 140 mmHg systolic or 90 mmHg diastolic pressure.<br />

** Diabetes was defined as either fasting plasma glucose ≥ 7.1, or ≥ 11 at 120 min of OGTT; Impaired<br />

glucose metabolism was defined as either fasting plasma glucose (< 7.1 and > 6.1), or impaired glucose<br />

tolerance (7.8 at 120 min of OGTT).<br />

*** Dyslipidaemia was defined as Triglycerides > 1.7 mmol/l and/or HDL-cholesterol < 0.9 mmol/l (male)<br />

and < 1 mmol/l (female). Hypercholesterolemia was defined as Total cholesterol > 5 mmol/l and/or LDLcholesterol<br />

> 3 mmol/l.<br />

ANOVA: analysis of variance; BMI, body mass index; CCP: cyclic citrullinated peptide; DAS28, Disease<br />

activation score using 28 joints; DBP, diastolic blood pressure; DMARD: disease-modifying antirheumatic<br />

drug; ESR, erythrocyte sedimentation rate; FPG: fasting plasma glucose; GC: glucocorticoid; HDL:<br />

high-density lipoprotein; IGM, impaired glucose metabolism; LDL: low-density lipoprotein; OGTT:<br />

oral glucose tolerance test; RA: rheumatoid arthritis; RA+GCs, rheumatoid arthritis patients, currently<br />

using glucocorticoids; RA–GCs, rheumatoid arthritis patients, glucocorticoid naïve; SBP, systolic blood<br />

pressure;VAS, visual analogue scale.<br />

Parameters of insulin sensitivity: Parameters of both fasting (Figure 2A) and postload<br />

insulin sensitivity (Figure 2 B+C) were comparable between the RA groups. In multivariate<br />

linear regression analyses (correcting for age, gender, BMI, waist circumference, and disease<br />

activity) the presence of RA, waist circumference and cumulative GC dose were independent<br />

predictors of HOMA-IR (Table 2). MCRest was negatively associated with DAS28, ESR, age,<br />

BMI and waist circumference (Table 2); a similar pattern was observed for OGIS (data not<br />

shown). The healthy control group was more insulin sensitive in the fasted state, but had<br />

similar postload insulin sensitivity as compared to the RA groups.<br />

Figure 2. Parameters of insulin sensitivity. Mean (±SD) for insulin sensitivity indices homeostatic<br />

model assessment of insulin resistance (HOMA-IR; fasting index), metabolic clearance rate (MCR ) and<br />

est<br />

oral glucose insulin sensitivity (OGIS) are shown. **P


Chapter 14 Glucocorticoids in chronic rheumatoid arthritis<br />

Table 2. Association of risk factors and disease characteristics with HoMA-IR and MCRest<br />

Patient characteristics Beta (95% CI) Beta (95% CI) Beta (95% CI)<br />

(RA-patients (RA-patients only (RA-patients only<br />

(n=56), controls<br />

(n=50))<br />

(n=140))<br />

(n=140))<br />

Three multivariate regression analysis models with HOMA-IR as dependent variable<br />

RA-GC* 0.6 (0.08, 1.0)<br />

RA+GC* 1.0 (0.6, 1.5)<br />

Waist circumference 0.02 (-0.01, 0.04) 0.02 (-0.003, 0.05) 0.03 (0.0002, 0.06)<br />

Age 0.002 (-0.02, 0.02) 0.008 (-0.007, 0.02) 0.006 (-0.01, 0.02)<br />

BMI 0.03 (-0.05, 0.1) 0.05 (-0.2, 0.1) 0.03 (-0.04, 0.1)<br />

Female gender 0.2 (-0.2, 0.6) -0.1 (-0.5, 0.3) -0.06 (-0.5, 0.4)<br />

Cumulative GC-dose (g)** 0.01 (0.003, 0.02) 0.01 (0.003, 0.02)<br />

Daily GC-dose** (mg) 0.01 (-0.04, 0.05) 0.005 (-0.04, 0.05)<br />

DAS28*** 0.1 (-0.04, 0.3)<br />

Any erosions of the hands or feet -0.3 (-0.7, 0.2)<br />

Past DMARDs (n)**** -0.06 (-0.2, 0.05)<br />

ACPA 0.2 (-0.7, 0.2)<br />

Disease duration (years) 0.02 (-0.008, 0.04)<br />

Three multivariate regression analysis models with MCR as dependent variable<br />

est<br />

RA-GC* 0.4 (-0.5, 1.3)<br />

RA+GC* 0.4 (-0.5, 1.2)<br />

Waist -0.02 (-0.07, 0.03) -0.06 (-0.1, 0.01) -0.09 (-0.2, -0.02)<br />

Age -0.03 (-0.06, 0.005) -0.07 (-0.1, -0.03) -0.07 (-0.1, -0.03)<br />

BMI -0.3 (-0.4, -0.1) -0.2 (-0.4, -0.06) -0.1 (-0.3, -0.06)<br />

Female gender -0.8 (-1.6, 0.08) -0.08 (-1.3, 1.1) -0.5 (-1.7, 0.7)<br />

Cumulative GC-dose (g)** 0.002 (-0.01, 0.04) 0.01 (-0.01, 0.04)<br />

Daily GC-dose** (mg) -0.07 (-0.19, 0.05) -0.04 (-0.2, 0.08)<br />

DAS28*** -0.5 (-0.9, -0.1)<br />

Any erosions of the hands or feet 0.8 (-0.3, 2.0)<br />

Past DMARDs (n)**** 0.2 (-0.06, 0.5)<br />

ACPA -0.7 (-1.8, 0.4)<br />

Disease duration (years) 0.03 (-0.04, 0.1)<br />

Three multivariate models, correcting for age, BMI, and female gender were used to test<br />

associations with insulin sensitivity parameters HOMA-IR and MCRest: (1) tested RA populations<br />

(as compared to healthy controls), (2) tested cumulative and daily GC-dose, and (3) tested<br />

disease activity (DAS28 and its components, DMARD-use, hand or feet erosions on X-ray, ACPA).<br />

Significant associations are depicted in bold.<br />

* RA-GC and RA+GC populations are tested with the healthy control group used as the reference population;<br />

only RA patients with normal glucose tolerance during OGTT and without 1st degree relatives with type 2<br />

diabetes mellitus were included in the model for the comparison with healthy controls.<br />

** Cumulative GC-dose, daily GC-dose were separately tested in models of only the RA population.<br />

*** Disease parameters (DAS28, DAS28CRP, CRP, ESR) were separately tested in the model of only the RA<br />

population; only DAS28 is depicted, whereas ESR was also significantly associated with MCRest in this<br />

model.<br />

276


**** Number of DMARDs (both synthetic and biological; not glucocorticoids) that were used by a patient<br />

in the past.<br />

ACPA: anti-citrullinated protein antibodies; BMI, body mass index; CRP: C-reactive protein; DAS28,<br />

disease activation score measured by questioning general health physical examination of 28 joints and<br />

ESR or CRP; DMARD: disease-modifying antirheumatic drug; ESR, erythrocyte sedimentation rate; GC,<br />

glucocorticoid; HOMA-IR, homeostatic model assessment of insulin resistance; MCRest, insulin sensitivity<br />

index by Stumvoll; OGTT: oral glucose tolerance test; RA: rheumatoid arthritis.<br />

Parameters of beta-cell function: HOMA-B was higher in RA+GC as compared to RA-GC<br />

(Figure 3A), while all dynamic measures of beta-cell function were comparable between the<br />

RA groups (Figure 3B-3D). Positive associations between the presence of RA and cumulative<br />

GC-use with HOMA-B were found (corrected for age, gender, BMI, waist circumference, and<br />

disease activity; data not shown). Age and both RA-GC and RA+GC were negatively associated<br />

with IGI (corrected for age, gender, BMI, waist circumference; data not shown), whereas no<br />

patient characteristics correlated with the disposition index (table 3). As compared to healthy<br />

controls, RA patients had higher basal C-peptide secretion (higher HOMA-B), but impaired<br />

early insulin secretion, also when corrected for insulin sensitivity (Figure 3B+C). The total<br />

amount of C-peptide secreted during the entire OGTT relative to glucose levels, was higher in<br />

RA patients than healthy controls (Figure 3D).<br />

Figure 3. Parameters of beta-cell function. Mean (±SD) for beta cell indices homeostatic model<br />

assessment of beta-cell function (HOMA-B; fasting index), insulinogenic index (IGI), disposition index<br />

(DI), and area under the C-peptide curve divided by area under the glucose curve (AUC /AUC ratio)<br />

c-pep gluc<br />

are shown. . *P


Chapter 14 Glucocorticoids in chronic rheumatoid arthritis<br />

Table 3. Associations of risk factors and disease characteristics with the disposition index.<br />

Patient<br />

Beta (95% CI)<br />

Beta (95% CI)<br />

Beta (95% CI)<br />

Characteristics (RA-patients (n=56), (RA-patients only (RA-patients only<br />

controls (n=50)) (n=140))<br />

(n=140))<br />

Three multivariate regression analysis models with DI as dependent variable<br />

RA-GC* -42489 (-96745, 11767)<br />

RA+GC* -23040 (-75891, 29810)<br />

Waist circumference -1778 (-4895, 1338) 315 (-1831, 2462) 274 (-2009, 2557)<br />

Age -3295 (-5219, -1370) -1280 (-2502, -57) -1100 (-2385, 186)<br />

278<br />

BMI 6205 (-1930, 14340) -2771 (-8193, 2650) -3070 (-8893, 2752)<br />

Female gender -8287 (-52574, 36000) 25773 (-10358, 61904) 31014 (-6984, 69013)<br />

Cumulative GC-dose<br />

-482 (-1307, 343) -284 (-1150, 582)<br />

(g)**<br />

Daily GC-dose** -885 (-4669, 2899) 140 (-3731, 4010)<br />

DAS28*** -4759 (-16525, 7007)<br />

Any erosions of the<br />

-14480 (-50730, 21770)<br />

hands or feet<br />

Past DMARDs (n)<br />

662 (-7572, 8896)<br />

****<br />

ACPA 24336 (-9300, 57973)<br />

Disease duration<br />

-1392 (-3447, 664)<br />

(years)<br />

Three multivariate models, correcting for age, BMI, and female gender were used to test associations with<br />

the beta-cell parameter disposition index: (1) tested RA populations (as compared to healthy controls),<br />

(2) tested cumulative and daily GC-dose, and (3) tested disease activity (DAS28 and its components,<br />

DMARD-use, hand or feet erosions on X-ray, ACPA).<br />

Significant associations are depicted in bold.<br />

* RA-GC and RA+GC populations are tested against the control subject population; only RA patients with<br />

normal glucose tolerance during OGTT and without 1st degree relatives with type 2 diabetes mellitus<br />

were included for the comparison with control subjects. RA+GC was significantly associated with DI when<br />

these analyses were performed with log-transformed DI.<br />

** Cumulative GC-dose, daily GC-dose were separately tested in models of only the RA population.<br />

*** Disease parameters (DAS28, DAS28CRP, CRP, ESR) were separately tested in the model of only the RA<br />

population; only DAS28 and significant correlations are depicted.<br />

**** Number of DMARDs (both synthetic and biological; not glucocorticoids) that were used by a patient<br />

in the past.<br />

ACPA: anti-citrullinated protein antibodies; BMI, body mass index; CRP: C-reactive protein; DAS28,<br />

disease activation score measured by questioning general health physical examination of 28 joints and<br />

ESR or CRP; DI: disposition index; DMARD: disease-modifying antirheumatic drug; ESR, erythrocyte<br />

sedimentation rate; GC, glucocorticoid; RA: rheumatoid arthritis; RA+GCs, rheumatoid arthritis patients<br />

currently using glucocorticoids; RA–GCs, glucocorticoid naive rheumatoid arthritis patients.


Effect modification: As compared to RA patients, controls had a higher percentage of male<br />

gender, and had a higher body mass index (BMI) and waist circumference; in addition, RA+GC<br />

had higher disease activity as compared to RA-GC patients (Table 1). These factors were<br />

studied for effect modification using interaction-terms in the below-mentioned regression<br />

models, and were shown not to modify the effects of patient and disease characteristics on<br />

glucose metabolism outcomes in the multivariate models (data not shown).<br />

DISCUSSIoN<br />

In this study of RA patients with established disease, chronic GC-users and GC-naive patients<br />

had similar insulin sensitivity and beta-cell function parameters; however high cumulative<br />

or daily GC-dose was associated with T2DM. In addition, in all RA patients IGM and T2DM<br />

were frequently diagnosed, suggesting that glucose intolerance remains an underestimated<br />

problem in RA. As compared to a healthy control group, RA patients had impaired insulin<br />

sensitivity and beta-cell dysfunction, explaining their impaired metabolic state.<br />

To our knowledge, this is the first study that has examined glucose metabolism in a relatively<br />

large sample of RA patients with established disease in such detail. Few studies investigated<br />

glucose tolerance state in RA: One study showed an increased prevalence of T2DM when<br />

compared to age-matched controls [27]; another study reported 19% diabetes prevalence<br />

as part of a longitudinal medical record cohort on cardiovascular risk [8]. Both studies relied<br />

on self-reported T2DM and did not perform glucose measurements. Because of the OGTT<br />

measurements of glucose at 0 and 120 minutes we were now able to register 11% T2DM<br />

prevalence in RA patients with established disease without known T2DM, and in addition,<br />

identify high-risk patients, by detecting 35% prevalence of IGM. This shows that glucose<br />

intolerance is a considerable and underestimated problem in RA patients with established<br />

disease, and might (partially) explain their increased cardiovascular risk [1].<br />

The subject of insulin resistance in RA has been addressed in recent years, but was only<br />

evaluated by fasting measure HOMA-IR [2, 5, 6]. Unlike these studies, we were able to<br />

show a negative association of DAS28 with insulin sensitivity after correction for potential<br />

confounders and other risk factors, which could have been due to the fact that we also used<br />

stimulated measures of insulin sensitivity and because our sample size was larger.<br />

Another important finding of our study was impaired beta-cell function in RA patients as<br />

compared to controls, as was shown by decreased dynamic parameters IGI and DI, also after<br />

279<br />

Chapter 14<br />

14


Chapter 14 Glucocorticoids in chronic rheumatoid arthritis<br />

correction for age, BMI and waist circumference (in case of IGI). This indicates impaired insulin<br />

secretion during the early phase after glucose stimulation. So far, a limited number of other<br />

studies have reflected upon beta-cell function in RA, and only used the fasting state measure<br />

HOMA-B. In one retrospective study HOMA-B was decreased in RA patients with a higher level<br />

of inflammation when compared to RA patients with lower level of inflammation [6], which<br />

seems in line with our findings of impaired beta-cell function in RA patients when compared<br />

to the control population. In our current analysis, HOMA-B was higher in RA patients, which<br />

indicates increased basal C-peptide secretion. This seems contradictory next to the decrease<br />

in beta-cell function parameters obtained in the stimulated state. However, HOMA-B should<br />

always be interpreted in the context of prevailing insulin resistance [19]. In our study, RA+GC<br />

participants had higher HOMA-IR values, i.e. they were more insulin resistant. In order to<br />

maintain fasting glucose levels within the normal range, more insulin was secreted in the<br />

fasted state, which resulted in a higher HOMA-B score. However, this higher HOMA-B score<br />

does not imply improved beta-cell function, but merely indicates the potential to compensate<br />

for reduced insulin sensitivity in the fasted state.<br />

In addition, we addressed the specific role of (cumulative or daily) GC-dose and disease<br />

characteristics within RA patients and found strong indications that RA+GC patients were<br />

less glucose tolerant in a dose-dependent manner: Although no relation was shown between<br />

(cumulative or daily) GC dose and dynamic parameters of insulin sensitivity and beta-cell<br />

function, cumulative- and daily GC dose were associated with previously unknown T2DM and<br />

negatively affected fasting insulin sensitivity (HOMA-IR); independent of age, gender, BMI,<br />

waist circumference, and disease activity. In addition, RA+GC patients had a decreased insulin<br />

clearance, indicating hepatic insulin resistance, which is explained by the steeper insulin curve<br />

in the first part of the OGTT. Our results are in line with one other retrospective study of nondiabetic<br />

RA patients [14]; that study analysed a successive group of RA patients and showed<br />

that ever having taken oral prednisone and/or high doses of pulsed GCs was independently<br />

associated with decreased insulin sensitivity, independent of BMI.<br />

We acknowledge some limitations in our study design; one is the difference with regards<br />

to anthropometrics between controls (with normal glucose tolerance) and RA patients, i.e.<br />

controls were primarily recruited for other studies at the Diabetes Center of the VU University<br />

Medical Center and therefore consisted of more males and had a relatively high BMI, and<br />

lower insulin clearance. Compared to these control subjects RA patients were more insulin<br />

resistant and had impaired beta-cell function. Although the use of this control population,<br />

as compared to more lean, insulin sensitive controls, may be suboptimal, it is very likely<br />

that the impact of RA and the associated pro-inflammatory state on glucose metabolism as<br />

280


described here, may even be underestimated. Besides, in multivariate analyses we corrected<br />

for these anthropometrics, and furthermore the control subjects served mainly to create a<br />

perspective for the insulin resistance and beta-cell parameters of RA patients. Another point<br />

is confounding by indication that could have caused the effects of GCs on glucose metabolism,<br />

since cumulative GC-use could be a proxy for long-term disease activity, which itself influences<br />

glucose metabolism. This was shown also in our study by a decrease of the regression<br />

coefficient (beta) for the association between cumulative or daily GC-dose and T2DM when<br />

disease activity (DAS28) was added to the multivariate regression model.<br />

Concluding, because of (1) the stimulated state measurement of glucose metabolism<br />

parameters, (2) the size of our population, and (3) the contrast with control subjects, we were<br />

able to draw firm conclusions on the prevalence of glucose tolerance abnormalities in RA<br />

patients with established disease and to confirm the relation between RA (activity) and insulin<br />

resistance and beta-cell dysfunction. Chronic GC-use was associated with metabolic toxicity<br />

in a dose dependent way, but this association was difficult to assess due to confounding by<br />

indication.<br />

Until more clarity is given on the issue of glucose intolerance in GC-using RA patients, it remains<br />

important to keep the use of GCs of short duration and lowest possible dose, as is advised<br />

by the EULAR recommendations on RA treatment [28], and on systemic GC use [29]. The<br />

question how harmful long-term GCs are with regards to diabetogenic effects in established<br />

RA patients needs further assessment in longitudinal (randomized) trials. These trials should<br />

measure glucose metabolism with stimulated state measures, and study whether GCs exert<br />

direct metabolic toxicity or secondary to other GC-related phenomena, such as abdominal fat<br />

and adipocytokines, which are known mediators of metabolic toxicity in RA [30].<br />

Funding: This paper was written within the framework of project T1-106 of the Dutch Top<br />

Institute <strong>Pharma</strong>.<br />

281<br />

Chapter 14<br />

14


Chapter 14 Glucocorticoids in chronic rheumatoid arthritis<br />

REFERENCES<br />

1. Peters Mj, van Halm VP, Voskuyl AE, Smulders YM, Boers M, Lems WF, Visser M, Stehouwer CD,<br />

Dekker jM, Nijpels G, Heine R, Dijkmans BA, Nurmohamed MT. Does rheumatoid arthritis equal<br />

diabetes mellitus as an independent risk factor for cardiovascular disease? A prospective study.<br />

Arthritis Rheum. 2009;61(11):1571-9.<br />

2. Dessein PH, joffe BI, Stanwix A, Botha AS, Moomal Z. The acute phase response does not fully<br />

predict the presence of insulin resistance and dyslipidemia in inflammatory arthritis. J Rheumatol.<br />

2002;29(3):462-6.<br />

3. Giorgino F, Almahfouz A, Goodyear Lj, Smith Rj. Glucocorticoid regulation of insulin receptor<br />

and substrate IRS-1 tyrosine phosphorylation in rat skeletal muscle in vivo. j Clin Invest.<br />

1993;91(5):2020-30.<br />

4. Lambillotte C, Gilon P, Henquin jC. Direct glucocorticoid inhibition of insulin secretion. An in vitro<br />

study of dexamethasone effects in mouse islets. j Clin Invest. 1997;99(3):414-23.<br />

5. Chung CP, Oeser A, Solus jF, Gebretsadik T, Shintani A, Avalos I, Sokka T, Raggi P, Pincus T, Stein CM.<br />

Inflammation-associated insulin resistance: differential effects in rheumatoid arthritis and systemic<br />

lupus erythematosus define potential mechanisms. Arthritis Rheum. 2008;58(7):2105-12.<br />

6. Dessein PH, joffe BI. Insulin resistance and impaired beta cell function in rheumatoid arthritis.<br />

Arthritis Rheum. 2006;54(9):2765-75.<br />

7. Shahin D, Eltoraby E, Mesbah A, Houssen M. Insulin resistance in early untreated rheumatoid<br />

arthritis patients. Clin Biochem. 2010;43(7-8):661-5.<br />

8. Maradit-Kremers H, Nicola Pj, Crowson CS, Ballman KV, Gabriel SE. Cardiovascular death in<br />

rheumatoid arthritis: a population-based study. Arthritis Rheum. 2005;52(3):722-32.<br />

9. Simard jF, Mittleman MA. Prevalent rheumatoid arthritis and diabetes among NHANES III<br />

participants aged 60 and older. j Rheumatol. 2007;34(3):469-73.<br />

10. Wild S, Roglic G, Green A, Sicree R, King H. Global prevalence of diabetes: estimates for the year 2000<br />

and projections for 2030. Diabetes Care. 2004;27(5):1047-53.<br />

11. van Raalte DH, Ouwens DM, Diamant M. Novel insights into glucocorticoid-mediated diabetogenic<br />

effects: towards expansion of therapeutic options? Eur j Clin Invest. 2009;39(2):81-93.<br />

12. Gulliford MC, Charlton j, Latinovic R. Risk of diabetes associated with prescribed glucocorticoids in<br />

a large population. Diabetes Care. 2006;29(12):2728-9.<br />

13. Gurwitz jH, Bohn RL, Glynn Rj, Monane M, Mogun H, Avorn j. Glucocorticoids and the risk for<br />

initiation of hypoglycemic therapy. Arch Intern Med. 1994;154(1):97-101.<br />

14. Dessein PH, joffe BI, Stanwix AE, Christian BF, Veller M. Glucocorticoids and insulin sensitivity in<br />

rheumatoid arthritis. j Rheumatol. 2004;31(5):867-74.<br />

282


15. Wolfe F, Michaud K. Corticosteroids increase the risk of diabetes mellitus in rheumatoid arthritis<br />

and contribute to the risk of myocardial infarction and heart failure [abstract]. Ann Rheum Dis.<br />

2004;63:S1:495.<br />

16. Svenson KL, Lundqvist G, Wide L, Hallgren R. Impaired glucose handling in active rheumatoid<br />

arthritis: effects of corticosteroids and antirheumatic treatment. Metabolism. 1987;36(10):944-8.<br />

17. Hallgren R, Berne C. Glucose intolerance in patients with chronic inflammatory diseases is<br />

normalized by glucocorticoids. Acta Med Scand. 1983;213(5):351-5.<br />

18. Dessein PH, joffe BI, Stanwix AE. Effects of disease modifying agents and dietary intervention<br />

on insulin resistance and dyslipidemia in inflammatory arthritis: a pilot study. Arthritis Res.<br />

2002;4(6):R12.<br />

19. Wallace TM, Levy jC, Matthews DR. Use and abuse of HOMA modeling. Diabetes Care.<br />

2004;27(6):1487-95.<br />

20. Pacini G, Mari A. Methods for clinical assessment of insulin sensitivity and beta-cell function. Best<br />

Pract Res Clin Endocrinol Metab. 2003;17(3):305-22.<br />

21. Prevoo ML, van ‘t Hof MA, Kuper HH, van Leeuwen MA, van de Putte LB, van Riel PL. Modified disease<br />

activity scores that include twenty-eight-joint counts. Development and validation in a prospective<br />

longitudinal study of patients with rheumatoid arthritis. Arthritis Rheum. 1995;38(1):44-8.<br />

22. Wells G, Becker jC, Teng j, Dougados M, Schiff M, Smolen j, Aletaha D, van Riel PL. Validation of<br />

the 28-joint Disease Activity Score (DAS28) and European League Against Rheumatism response<br />

criteria based on C-reactive protein against disease progression in patients with rheumatoid<br />

arthritis, and comparison with the DAS28 based on erythrocyte sedimentation rate. Ann Rheum Dis.<br />

2009;68(6):954-60.<br />

23. Kotronen A, Vehkavaara S, Seppala-Lindroos A, Bergholm R, Yki-jarvinen H. Effect of liver fat on<br />

insulin clearance. Am j Physiol Endocrinol Metab. 2007;293(6):E1709-15.<br />

24. Levy jC, Matthews DR, Hermans MP. Correct homeostasis model assessment (HOMA) evaluation<br />

uses the computer program. Diabetes Care. 1998;21(12):2191-2.<br />

25. Mari A, Pacini G, Murphy E, Ludvik B, Nolan jj. A model-based method for assessing insulin sensitivity<br />

from the oral glucose tolerance test. Diabetes Care. 2001;24(3):539-48.<br />

26. Kosaka K, Hagura R, Kuzuya T, Kuzuya N. Insulin secretory response of diabetics during the period of<br />

improvement of glucose tolerance to normal range. Diabetologia. 1974;10(6):775-82.<br />

27. Han C, Robinson DW, jr., Hackett MV, Paramore LC, Fraeman KH, Bala MV. Cardiovascular disease<br />

and risk factors in patients with rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis.<br />

j Rheumatol. 2006;33(11):2167-72.<br />

283<br />

Chapter 14<br />

14


Chapter 14 Glucocorticoids in chronic rheumatoid arthritis<br />

28. Smolen jS, Landewe R, Breedveld FC, Dougados M, Emery P, Gaujoux-Viala C, Gorter S, Knevel R, Nam<br />

j, Schoels M, Aletaha D, Buch M, Gossec L, Huizinga T, Bijlsma jW, Burmester G, Combe B, Cutolo M,<br />

Gabay C, Gomez-Reino j, Kouloumas M, Kvien TK, Martin-Mola E, McInnes I, Pavelka K, van Riel P,<br />

Scholte M, Scott DL, Sokka T, Valesini G, van Vollenhoven R, Winthrop KL, Wong j, Zink A, van der<br />

Heijde D. EULAR recommendations for the management of rheumatoid arthritis with synthetic and<br />

biological disease-modifying antirheumatic drugs. Ann Rheum Dis. 2010;69(6):964-75.<br />

29. Hoes jN, jacobs jW, Boers M, Boumpas D, Buttgereit F, Caeyers N, Choy EH, Cutolo M, Da Silva jA,<br />

Esselens G, Guillevin L, Hafstrom I, Kirwan jR, Rovensky j, Russell A, Saag KG, Svensson B, Westhovens<br />

R, Zeidler H, Bijlsma jW. EULAR evidence-based recommendations on the management of systemic<br />

glucocorticoid therapy in rheumatic diseases. Ann Rheum Dis. 2007;66(12):1560-7.<br />

30. Rho YH, Chung CP, Solus jF, Raggi P, Oeser A, Gebretsadik T, Shintani A, Stein CM. Adipocytokines,<br />

insulin resistance, and coronary atherosclerosis in rheumatoid arthritis. Arthritis Rheum.<br />

2010;62(5):1259-64.<br />

284


PART<br />

Discussion<br />

V


Chapter 15<br />

General Discussion:<br />

“Diabetogenic Effects of Glucocorticoid<br />

Drugs: The Knowns and The Unknowns”<br />

D.H. van Raalte and M. Diamant<br />

Manuscript in Preparation


Chapter 15 General Discussion<br />

Glucocorticoids: efficacious anti-inflammatory agents limited by their side effects<br />

For decades, glucocorticoids (GCs) are the most commonly prescribed anti-inflammatory and<br />

immunosuppressive agents. GCs reduce the activity of the innate immune system by decreasing<br />

phagocytic activity and by attenuating the production of pro-inflammatory mediators [1].<br />

Additionally, GCs affect the acquired immune system by impairing a variety of T-cell functions<br />

[1]. Because of these combined immunomodulatory actions, GCs are the cornerstone in the<br />

treatment of numerous inflammatory diseases, including rheumatic diseases (rheumatoid<br />

arthritis (RA) [2] and polymyalgia rheumatica [3]), systemic lupus erythematosus [4], small<br />

and large vessel arteritis [5], inflammatory bowel disease [6], chronic obstructive pulmonary<br />

disease [7] and various other conditions of inflammatory origin. Already in 1949, the beneficial<br />

effects of systemic cortisone treatment, the first available GC, on some of these conditions<br />

were recognized and the published results were received with great enthusiasm. Pioneers in<br />

the discovery of adrenal gland hormones and their clinical effects, Tadeusz Reichstein, Philip<br />

Showalter Hench and Edward Calvin Kendall were awarded the Nobel Prize in 1950.<br />

In the following decade, however, the initial enthusiasm for GC treatment was tampered when<br />

its detrimental side effect profile became increasingly apparent. The earliest side effects<br />

reported, included salt and water retention, increased gastric acidity and psychosis [8]. Soon the<br />

full scope of side effects induced by GC treatment became evident and is now known to include<br />

weight gain, fluid retention, hypertension, impaired glucose metabolism, diabetes, skeletal<br />

muscle atrophy, osteoporosis, gastric ulcer disease, glaucoma, neuropsychiatric symptoms and<br />

increased risk to develop cardiovascular disease. And this summary of GC-related side effects<br />

may even not be complete [9, 10]. Despite the formulation of newer synthetic compounds,<br />

such as prednisolone and dexamethasone, the side-effect profile remained largely unchanged.<br />

This unfavorable side effect profile of systemic GC treatment has several important clinical<br />

consequences. First, physicians are often forced to lower GC dosages compromising<br />

efficacy of treatment of several disease entities. Secondly, resistance among physicians<br />

to prescribe and among patients to take GCs due to their infamous side effect profile<br />

is a well-characterized phenomenon, resulting in inadequate treatment of several<br />

disease entities [11]. Finally, in clinical practice, additional therapies are often initiated<br />

to mitigate the side effects induced by GC treatment, e.g. bisphosphonates to prevent<br />

osteoporosis [12] and proton pump inhibitors to prevent gastric ulcer disease [13].<br />

Glucocorticoids diabetogenic effects: focus of this thesis<br />

In this thesis, we have focused on the unfavorable effects of GCs on glucose metabolism, and to<br />

a lesser extent on lipid and protein metabolism. Additionally, we have performed pioneering<br />

studies to further detail the underlying mechanisms of the metabolic side effects. Specifically,<br />

290


the effects of GCs on insulin production (beta-cell function) and insulin sensitivity were<br />

assessed. We have concentrated on these glucometabolic side effects of GC treatment since<br />

GC-induced glucose intolerance and overt diabetes are frequently occurring adverse events<br />

of GC treatment [14-16]. However, these effects, in particular the mechanisms contributing<br />

to their development, are still only partly understood. Additionally, no guidelines currently<br />

exist how to treat or prevent this adverse effect, as has been the case for some of the forementioned<br />

side effects of systemic GC treatment.<br />

Glucocorticoids diabetogenic effects: importance of understanding the pathophysiology<br />

Understanding both the mechanisms of GC action and the mechanisms underlying GCs<br />

diabetogenic effects is relevant for several reasons. First, at a molecular level, increased<br />

insight to the (nuclear) actions of GCs has led to the current development of dissociated<br />

glucocorticoid receptor (GR) agonists or selective GR modulators (as extensively reviewed<br />

in chapter 2). These compounds are designed to specifically induce transrepression, but<br />

not transactivation, thus aiming to reduce metabolic side effects with unchanged antiinflammatory<br />

capacities, potentially leading to an enhanced therapeutic index [17-19]. At<br />

present, several pharmaceutical companies are developing such compounds, including Merck<br />

Sharp Dome (former NV Organon), our partner in this Top Institute <strong>Pharma</strong> consortium<br />

(see general introduction). In recent studies, they were able to show that their compound<br />

ORG214007-0 (C H N OS) was able to reduce inflammation both in vitro and in vivo in<br />

24 23 5<br />

collagen-induced arthritis mice to the same extent as prednisolone, while demonstrating<br />

reduced transactivation. As such, the expression of key enzymes in gluconeogenesis<br />

phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase (GPPase) was<br />

decreased, resulting in unchanged fasting glucose plasma levels in mice treated with<br />

ORG214007-0, while prednisolone induced hyperglycemia [20]. However, to enable further<br />

development of these compounds for use in humans, insight into the mechanisms of GCinduced<br />

side effects is necessary. More specifically, there is need for the identification of<br />

biomarkers that in a dose-dependent matter reflect or predict the occurrence of side effects of<br />

the classic GR agonists, in order to compare these adverse effects with novel GR modulators.<br />

Secondly, increased understanding of the mechanisms underlying GCs diabetogenic effects<br />

may improve strategies to treat or even prevent these glucometabolic abnormalities, since<br />

the novel dissociated GR activators are not likely to become available for clinical use within a<br />

short period of time. Thus, identification of the organs and pathways involved may provide an<br />

evidence base to develop therapeutic and preventive strategies in the context of chronic GC<br />

treatment and its associated diabetogenic side effects. Additionally, these data will be critical<br />

to expand our knowledge that can be harnessed for the development of targeted dissociated<br />

GR agonists.<br />

291<br />

Chapter 15<br />

15


Chapter 15 General Discussion<br />

Studies conducted in this thesis<br />

In order to study the mechanisms underlying the adverse metabolic effects of GCs, a number<br />

of studies were performed in this thesis. Most of the data in this thesis were obtained from the<br />

PANTHEON (I + II) studies: the PeripherAl effects of prednisolone oN glucose meTabolism,<br />

metabolic Hormones, insulin sEnsitivity and insulin secretioN in healthy young males: two<br />

randomized, placebo-controlled, double blind, dose-response intervention studies. In both<br />

PANTHEON-I and -II study, 32 healthy male volunteers were randomized to a two-week<br />

treatment with prednisolone 30 mg daily (so called induction dosage; n=12), prednisolone<br />

7.5 mg daily (so called maintenance dosage; n=12) or placebo (n=8) (chapters 5, 8, 9, 10,<br />

11 and 12). These dosages were chosen since they represent the commonly prescribed highand<br />

low dosage in clinical practice. In both studies, participants were treated for a period<br />

of two weeks in order to enable studying past the acute effects of prednisolone treatment.<br />

However, for ethical reasons, it was not feasible to administer healthy volunteers high-dose<br />

prednisolone for a more prolonged period of time.<br />

In the PANTHEON studies, the dose-dependent effects of GCs on glucose tolerance was<br />

assessed by standardized meal challenge tests (chapter 5). We studied the effects of GC<br />

treatment on insulin sensitivity of liver, skeletal muscle and adipose tissue (chapter 5 and<br />

8) by using hyperinsulinemic-euglycemic clamps with stable isotopes. To gain insight into<br />

mechanisms of GC-induced skeletal muscle insulin resistance, insulin signaling was studied<br />

in skeletal muscle biopsies before and during GC treatment (chapter 10). Additionally, the<br />

potential role for both accumulation of glycosphingolipids and the ganglioside GM-3, and<br />

mitochondrial dysfunction in GC-induced skeletal muscle insulin resistance was addressed in<br />

chapter 9. The contribution of microvascular dysfunction to the GC-induced attenuation of<br />

insulin-stimulated glucose uptake in skeletal muscle is reported in chapter 10.<br />

To characterize the effects of GC on adipose tissue function at the molecular level, subcutaneous<br />

adipose tissue biopsies were collected before and during prednisolone treatment and were<br />

analyzed for insulin signaling, lipolytic enzymes, markers of differentiation and secretion<br />

of adipocytokines (chapter 11). In addition, the role of the recently discovered protein<br />

angiopoietin-like protein (Angptl)-4 in GC-induced changes in lipid metabolism particularly<br />

in relation to insulin resistance was studied in chapter 12.<br />

In addition to the effects of GCs on insulin sensitivity, the effects of GCs on beta-cell function<br />

was assessed, both following acute (chapters 4 and 7) and more chronic exposure (chapter<br />

5). In chapter 3, we report that GC-induced beta-cell dysfunction is characterized by<br />

endoplasmic reticulum (ER) stress resulting in impaired insulin production and secretion and<br />

beta-cell apoptosis.<br />

292


In chapter 7, we have described a proof-of-concept study demonstrating the potential<br />

of glucagon-like peptide-1 receptor agonists (GLP-1 RA) to prevent GC-induced glucose<br />

intolerance and beta-cell dysfunction. In this study, called the PREDEX (PREdnisoloneinduced<br />

beta-cell Dysfunction prevented by EXenatide) study 8 healthy men were treated<br />

with a single gift of prednisolone 80 mg (or placebo) and received the GLP-1 RA exenatide (or<br />

saline) intravenously.<br />

Finally, we investigated the effects of GCs on glucose tolerance, insulin sensitivity and insulin<br />

secretion in patients with RA. In these studies, the metabolic effects of chronic inflammation<br />

and reduction hereof by prednisolone treatment could be added into the equation. To this<br />

extent, a single blind, randomized controlled trial in forty-one patients with early active RA<br />

was conducted with two high dosages of prednisolone (30 mg daily and 60 mg daily) dosed for<br />

a single week, as a sub study of the COBRA-light trial (Combinatie Behandeling Reumatoide<br />

Artritis) (chapter 13). Off note, the COBRA-light study was designed to compare two different<br />

treatment schedules for early RA (COBRA schedule according to BeSt compared to a modified<br />

COBRA schedule (‘light’) with lower prednisolone dosages).<br />

Moreover, a cross-sectional study in chronic RA patients was performed. In this study, glucose<br />

metabolism was compared among RA patients chronically treated with GCs and RA patients<br />

that had never used GCs, allowing studying the metabolic effects of GC treatment while<br />

correcting for the influence of the underlying disease (chapter 14).<br />

In the following section, the key organs involved in insulin action and insulin secretion that<br />

are affected by GC treatment are summarized in the section below, based both on the results<br />

obtained from our studies and on previous data or data recently obtained by other research<br />

groups.<br />

Glucocorticoids diabetogenic effects: involved organs and pathways<br />

1. Liver<br />

The liver is a key regulator of metabolism, within a complex regulatory network of hormonal,<br />

autonomic nervous and metabolic stimuli. Under fasting conditions, the liver maintains<br />

euglycemia by producing glucose through both gluconeogenesis and glycogenolysis. Insulin,<br />

which is secreted in response to a meal or carbohydrate load is the most important hormone<br />

that suppresses endogenous glucose production (EGP). On the other hands, the so-called<br />

contra-regulatory hormones cortisol and glucagon increase EGP under hypoglycemic<br />

conditions. The endogenous hormone cortisol as well as exogenously administered GCs act<br />

293<br />

Chapter 15<br />

15


Chapter 15 General Discussion<br />

Figure 1. Currently known mechanisms by which glucocorticoids (GCs) induce hyperglycemia in<br />

healthy humans. GCs impair beta-cell function by inducing endoplasmic reticulum (ER) stress and by<br />

reducing calcium signaling in beta cells, resulting in impaired insulin secretion. GCs furthermore impair<br />

the effects of a number of non-glucose insulin secretagogues, including arginine, acetylcholine and the gut<br />

hormones glucagon-like peptide (GLP)-1 and glucose-dependent insulinotropic peptide (GIP). In addition,<br />

GC impair alpha-cell function, since glucagon levels are inappropriately elevated both under fasting<br />

conditions and in the postprandial state. Increased glucagon levels may stimulate endogenous glucose<br />

production (EGP) by the liver. In addition, GCs induce liver insulin resistance, thus further contributing<br />

to GC-induced increment of EGP, especially in the hyperinsulinemic state. GCs also induce adipose tissue<br />

insulin resistance, resulting in impaired glucose uptake and increased lipolysis in the postprandial<br />

period. Moreover, plasma levels of adipokines are altered towards a more diabetogenic profile. Finally,<br />

GCs impair glucose uptake in skeletal muscle by impairing insulin signaling. This reduction in insulinstimulated<br />

glucose uptake may in part be explained by impaired insulin-stimulated capillary recruitment,<br />

since GCs also induce vascular insulin resistance. By these combined actions, GCs induce hyperglycemia<br />

in healthy men. (See page 367 for full color figure)<br />

partly under influence of the autonomic nervous system (ANS) that innervates the adrenal<br />

gland. As such, in a number of studies [21-23], but not in all [24-27], short-term GC treatment<br />

was shown to augment fasting EGP in healthy volunteers. Indeed, we confirmed and expanded<br />

these findings by showing a relatively small increase in fasting EGP following a two-week<br />

treatment with prednisolone 30 mg daily as measured by hyperinsulinemic-euglycemic<br />

clamp with labeled glucose as described in chapter 8 [28]. Low-dose prednisolone did not<br />

294


affect EGP in the fasted state. As reviewed in chapter 2, GCs may increase EGP and particular<br />

gluconeogenesis by several mechanisms. First, GCs increase the expression and activity of<br />

rate limiting enzymes of gluconeogenesis including PEPCK and G6Pase [29-31]. Indeed, the<br />

PEPCK gene contains a glucocorticoid response element (GRE) in its promoter region and is<br />

considered a key player in GC-induced hyperglycemia [32], but several other genes in liver<br />

glucose metabolism were identified as GC target genes in gene expression profiling studies<br />

[33]. Interestingly, it was recently demonstrated that other nuclear receptors, including<br />

peroxisome-proliferator activated receptor (PPAR)-alpha and liver X receptors (LXRs),<br />

important regulators of lipid and cholesterol metabolism respectively, are potentiating<br />

factors in GC-induction of PEPCK and G6Pase, since PPAR and LXR knock-out mice models<br />

were shown to be in part resistant to the metabolic effects of GC treatment by reducing the<br />

activation of these enzymes [34, 35]. In addition, GC treatment promotes breakdown of<br />

protein and fat stores [36], thus providing increased supply of substrates for gluconeogenesis,<br />

such as alanine and glycerol, to the liver [37, 38]. As such, we could demonstrate in chapter<br />

8 that during hyperinsulinemic conditions, high-dose prednisolone treatment impaired<br />

the insulin-mediated suppression of lipolysis and proteolysis, whereas low-dose treatment<br />

only increased lipolysis [28]. Finally, GCs may potentiate the effect of other glucoregulatory<br />

hormones, such as glucagon and epinephrine, thus further enhancing fasting EGP [35, 39].<br />

The strongest effects of GCs on liver glucose metabolism in our studies, however, were<br />

observed under hyperinsulinemic conditions. In healthy volunteers, insulin infusion at 200<br />

mU/m2 .min decreased EGP by approximately 80%. This insulin-effect was markedly and<br />

dose-dependently reduced by prednisolone treatment, by 17±6 and 46±7 % respectively<br />

(chapter 8) [28]. The observation that glucocorticoids blunt the suppressive effects of insulin<br />

on EGP was shown previously during a 24-hr high-dose continuous cortisol infusion [21, 22].<br />

We could demonstrate for the first time that this effect is already present at prednisolone<br />

dosages just above physiological levels (7.5 mg daily). The mechanisms underlying GCinduced<br />

liver insulin resistance are at present not clarified. In rats, dexamethasone was<br />

shown to impair insulin-induced phosphorylation of insulin receptor substrate (IRS)-1 and<br />

phosphatidylinositol 3-kinase (PI-3K) in liver cells indicating impaired insulin signaling [40].<br />

We conclude that the liver is a key player in the diabetogenic effects induced by GC treatment<br />

through increased EGP, and more particular, by inappropriate suppression of glucose<br />

production in the hyperinsulinemic state (Figure 1). Thus, several experimental approaches<br />

have been undertaken to further characterize the involved mechanisms and to assess strategies<br />

to reduce these hepatic metabolic effects of excess GC levels. First, treatment of rats with a<br />

liver-selective GR antagonist resulted in reduced fasting plasma glucose levels and decreased<br />

EGP during a hyperinsulinemic-euglycemic clamp [41]. In addition, also in rats, activation of<br />

295<br />

Chapter 15<br />

15


Chapter 15 General Discussion<br />

AMP-activated protein kinase (AMPK) was shown to suppress GC-induced hyperglycemia by<br />

reducing PEPCK and G6Pase expression [42]. Interestingly, the most commonly prescribed<br />

blood-glucose lowering agent metformin has been shown to exert its effects by activating<br />

AMPK, which potentially renders this drug capable to prevent or counteract GC-induced<br />

diabetogenic effects [43].<br />

2. Skeletal Muscle<br />

Skeletal muscle tissue is the most important site for insulin-stimulated glucose uptake in the<br />

postprandial period and plays a crucial role in glucose metabolism [44]. Already in studies<br />

performed several decades ago, GCs were shown to reduce insulin-stimulated glucose disposal<br />

in a dose-dependent matter [21, 45, 46], which was confirmed in our studies (chapters 5, 9,10<br />

and 11). In agreement with other studies [47, 48] we could demonstrate that particularly<br />

nonoxidative glucose disposal (reflecting glycogen synthesis) is impaired by GC treatment,<br />

whereas glucose oxidation rates remained intact (chapter 9) [28]. Despite the fact that GCinduced<br />

skeletal muscle insulin resistance is a well-known observation, the pathways in<br />

skeletal muscle tissue responsible for this impairment presently remain incompletely detailed.<br />

From rodent studies, it has become evident that insulin signaling is impaired by GC treatment.<br />

As such, the phosphorylation of several proteins in the insulin-signaling cascade including<br />

IRS-1, PI-3K, Akt and glycogen synthase kinase (GSK) was shown to be reduced [40, 49-52].<br />

We could for the first time confirm this finding in humans, where high-dose prednisolone<br />

treatment impaired insulin-induced phosphorylation of its downstream targets in skeletal<br />

muscle (chapter 10) [53]. However, to date, it is unclear which molecular mechanisms are<br />

targeted by GC treatment that could interfere with insulin signaling.<br />

It has been postulated that GC-induced changes in protein metabolism may be involved. Indeed,<br />

GC treatment is frequently associated with skeletal muscle wasting due to proteolysis [54-59].<br />

GCs not only reduce skeletal muscle mass, but may also increase plasma levels of amino acids<br />

which were shown to impair insulin signaling in skeletal muscle [60, 61]. As shown in chapter<br />

8, and in line with previous reports [57], we observed that high-dose prednisolone treatment<br />

impaired the suppressive effects of insulin on whole-body valine turnover, but that it did not<br />

affect whole-body valine turnover under fasting conditions. Given the small effect size that<br />

was observed and the absence of a correlation with measures of glucose metabolism, we feel<br />

that altered amino acid metabolism is unlikely to be responsible for the dramatic reduction in<br />

insulin-stimulated glucose uptake following short-term GC treatment.<br />

Additionally, GC-induced changes in lipid metabolism could play a role in skeletal muscle<br />

insulin resistance. Glucorticoids were shown to impair the suppressive effects of insulin<br />

296


on whole-body lipolysis, resulting in increased levels of nonesterified fatty acids (NEFA)<br />

(chapter 8) [28]. NEFA spillover may enhance the accumulation of intramyocellular lipids<br />

(IMCL) [62], including triacylglycerol, but also more toxic intermediates such as fatty acyl<br />

CoA, diacylglycerol, sphingolipids such as ceramide and gangliosides such as GM-3, which<br />

have been shown to impair insulin signal transduction and have been associated with reduced<br />

skeletal muscle insulin sensitivity [63-66]. We were particularly interested in the role of<br />

sphingolipids, since attenuation of ceramide synthesis either by pharmacological treatment<br />

with myriocin or through genetic ablation of dihydroceramide desaturase (des1) was shown<br />

to prevent GC-induced insulin resistance in rodents [67]. However, in the presence of marked<br />

insulin resistance, skeletal muscle concentrations of ceramide, glucosylceramide and GM-3<br />

were not affected by prednisolone treatment (chapter 9) [28]. An explanation for this<br />

discrepant finding as compared to the rodent study may possibly be explained by the different<br />

tissue examined. Although GC treatment was shown to attenuate insulin-mediated glucose<br />

uptake in skeletal muscle, increased ceramide levels were particularly demonstrated in liver,<br />

and not in skeletal muscle. In addition, several other factors, including different treatment<br />

duration and differences in lipid metabolism between rodents and humans, may contribute<br />

to the observed difference.<br />

Recently, impaired mitochondrial function has received much interest given its association with<br />

reduced peripheral insulin sensitivity in subjects with T2DM [68]. It was hypothesized that<br />

impaired mitochondrial function results in accumulation of toxic lipid intermediates, which<br />

affect insulin signaling and glucose metabolism. Since GCs induce myopathy, we hypothesized<br />

that mitochondrial dysfunction could be involved in GC-induced insulin resistance. Thus, we<br />

measured ex vivo mitochondrial respirometry capacity in human skeletal muscle, adjusted<br />

for mitochondrial copy number, before and after 14-days prednisolone treatment. However,<br />

despite marked insulin resistance, mitochondrial function was not altered by prednisolone<br />

treatment (chapter 9). It should be noted that several measures of mitochondrial function<br />

exist in literature, where the gold standard measure remains to be established [68]. Therefore,<br />

we cannot completely rule out a potential role of mitochondrial dysfunction in GC-induced<br />

insulin resistance [69, 70].<br />

Other factors may contribute to GC-induced impairment in insulin signaling and insulinstimulated<br />

glucose disposal. First, GCs were shown, despite its general anti-inflammatory<br />

properties, to induce the acute phase response in skeletal muscle tissue, which could reduce<br />

insulin sensitivity [71]. Moreover, the endoplasmic reticulum (ER) has been identified as a<br />

major regulator of metabolic homeostasis and inflammatory responses and ER stress was<br />

shown to have unbeneficial effects on insulin sensitivity [72]. Whereas GCs were recently<br />

297<br />

Chapter 15<br />

15


Chapter 15 General Discussion<br />

shown to induce ER stress in beta cells [73], it is uncertain whether GCs may also alter ER<br />

function in skeletal muscle tissue. Finally, GCs may impair insulin sensitivity by impairing the<br />

delivery of insulin and glucose to skeletal muscle by reducing capillary recruitment through<br />

the induction of vascular insulin resistance as detailed below [53].<br />

We conclude that skeletal muscle tissue is a key player in GC-mediated insulin resistance<br />

resulting in a dose-dependent reduction in insulin-stimulated glucose uptake (Figure 1).<br />

However, a single causative pathway could not be identified in our studies.<br />

3. Adipose tissue<br />

Although the adverse metabolic effects of elevated GC levels on glucose [74] and protein<br />

metabolism [75] are reasonably well defined, the effects on lipid metabolism are presently<br />

less clear [76]. In addition, the involvement of a number of organs in the development of GCs<br />

diabetogenic effects, including pancreatic islet cells, skeletal muscle and liver are understood<br />

to a greater extent than the role of adipose tissue [9]. However, several observations indicate<br />

that GCs exert unfavorable effects on adipose tissue and lipid metabolism.<br />

As such, chronic GC excess in Cushing’s syndrome or during GC treatment increases fat<br />

deposition in the visceral compartment [77] and promotes liver fat accumulation [78], at the<br />

cost of subcutaneous fat deposition. This increment in visceral adipose tissue (VAT) by GCs<br />

may be induced by several factors, including increased intake of high-caloric “comfort food”<br />

[79], increased lipoprotein lipase (LPL) activity particularly in VAT [80, 81], increased VAT<br />

adipogenesis through stimulation of differentiation of pre-adipocytes into mature adipocytes<br />

[82] and possibly enhanced de novo lipogenesis [83]. Although we observed no significant<br />

changes in body weight, body composition, body fat distribution and liver fat content following<br />

two-week treatment with low- or high-dose prednisolone (chapter 8) [28], in subcutaneous<br />

adipose tissue (SAT) biopsies decreased protein expression of adipogenesis markers<br />

(peroxisome-proliferator activated receptor (PPAR)-γ and Kruppel-like factor (KLF)-4) was<br />

observed (chapter 11). Although our study did not allow to examine VAT, on extrapolation,<br />

these findings are in line with and may ultimately lead to the classical body fat distribution<br />

observed following chronic GC treatment as indicated above. The GC-induced reduction in<br />

SAT with concomitant increase in VAT is clinically relevant since VAT is well known to be<br />

associated with an untoward metabolic profile as opposed to SAT and is associated with<br />

increased cardiovascular risk [84].<br />

In addition to altering adipose tissue distribution, GCs may also affect adipose tissue function.<br />

We were able to show that GC treatment impaired insulin signaling in SAT in vivo in healthy<br />

298


humans. The insulin-stimulated phosphorylation of well-known insulin target proteins<br />

Akt/PKB and the downstream target PRAS-40 was impaired by high-dose prednisolone<br />

treatment (chapter 11). These data confirm and expand in vitro and in vivo rodent studies,<br />

where GC treatment impaired insulin signaling [85]. Impaired insulin action in adipose tissue<br />

may adversely affect both glucose and lipid metabolism. Indeed, an intact insulin-signaling<br />

pathway is of pivotal importance for insulin-stimulated glucose uptake through recruitment<br />

of the glucose transporter (GLUT)-4 to the plasma membrane enabling glucose transport<br />

into the cell [86]. Although the overall contribution of glucose uptake by adipose tissue in<br />

the postprandial state is thought to be relatively modest [44], it does contribute to glucose<br />

tolerance as was elegantly demonstrated in adipose tissue-specific glut4-/- mice [87]. These<br />

mice developed hyperglycemia and were characterized by liver and skeletal muscle insulin<br />

resistance, most likely due to altered secretion of adipokines [87]. In our study, prednisolone<br />

30 mg altered the circulating levels of various adipokines towards a more diabetogenic<br />

profile, as was previously oberved in in vitro studies [88]. Thus, plasma concentrations of<br />

adiponectin, which are generally positively associated with insulin sensitivity, were reduced<br />

following high-dose prednisolone treatment, whereas adiponectin protein expression in SAT<br />

was decreased by both prednisolone dosages. The adipokines resistin and leptin, which are<br />

usually negatively associated with insulin sensitivity, were both increased by prednisolone<br />

30 mg (chapter 11). Although increased leptin levels have been shown to inhibit food intake<br />

in the central nervous system (CNS), leptin also acts as pro-inflammatory factor in adipose<br />

tissue by promoting the release of pro-inflammatory adipokines. Similarly, resistin has also<br />

been demonstrated to promote circulating pro-inflammatory adipokine levels [89].<br />

In addition to promoting glucose uptake in adipose tissue, insulin is a key hormone in the<br />

regulation of lipid metabolism, amongst others by inhibiting adipose tissue lipolysis and thus<br />

decreasing plasma NEFA levels [90]. In contrast, GCs were shown, despite their lipogenic<br />

actions in VAT, to acutely increase fasting lipolysis rates in a number of in vitro studies [76].<br />

GC-induced induction of lipolysis could be explained by increased activity of key lipolytic<br />

enzymes adipose triglyceride lipase (ATGL) [91] and hormone sensitive lipase (HSL)<br />

[92] and possibly by augmented beta-adrenergic signaling following GC treatment [76]. It<br />

should be noted that differences were observed between various adipose tissue depots<br />

where GC-induced augmentation in lipolysis rates was particularly observed in VAT and less<br />

pronounced in SAT [93]. In addition, antilipolytic effects of GC treatment have also observed<br />

in vitro [76]. In line with most of these in vitro observations, acute GC administration was<br />

shown to increase fasting whole-body lipolysis in healthy humans [83, 94, 95]. During more<br />

prolonged GC exposure, however, this lipolytic effect was no longer observed most likely<br />

due to compensatory hyperinsulinemia [25, 96, 97]. In our study, high-dose prednisolone<br />

treatment even lowered fasting lipolysis (chapter 8). In line with these findings, we observed<br />

299<br />

Chapter 15<br />

15


Chapter 15 General Discussion<br />

decreased fasting ATGL expression, the rate-limiting enzyme in adipose tissue lipolysis,<br />

during both low- and high-dose prednisolone treatments, while fasting HSL expression was<br />

unchanged (chapter 11).<br />

In the same study, however, we observed for the first time that prednisolone treatment dosedependently<br />

impaired insulin-stimulated suppression of lipolysis and increased plasma NEFA<br />

levels during hyperinsulinemia, indicating adipose tissue insulin resistance. Since both ATGL<br />

expression and phosphorylation of HSL in SAT during insulin infusion were unaltered by<br />

prednisolone treatment, it is unclear through which mechanisms this effect occurs. Possibly,<br />

the inhibiting effects of insulin on protein kinase A and perilipin expression, a lipid dropletassociating<br />

protein, are reduced by prednisolone treatment, thereby promoting triglyceride<br />

lipolysis. An alternative hypothesis is that the increment in whole-body lipolysis is mostly<br />

derived from VAT, from which no biopsies could be obtained in our study design. Increased<br />

lipolysis and the ensuing consequently enhanced plasma NEFA levels have been associated<br />

with impaired glucose metabolism via their inhibiting effects on muscle and liver insulin<br />

signaling [98].<br />

In conclusion, based on our findings in healthy volunteers, GCs diabetogenic effects also<br />

involve actions on adipose tissue, i.e. induction of adipose tissue insulin resistance, resulting<br />

increased lipolysis, as well as induction of untoward plasma adipokine levels, and therefore it<br />

is likely that adipose tissue is involved in GCs diabetogenic effects.<br />

4. Pancreatic beta cells<br />

The pancreatic beta-cell plays a crucial role in glucose metabolism and in the past decades,<br />

beta-cell dysfunction has been acknowledged as the key defect underlying the development<br />

of T2DM [99]. Under physiological conditions, insulin secretion is directly related to insulin<br />

sensitivity through a hyperbolic relation [100]. The product of these parameters, known as<br />

the disposition index, remains constant. Thus, when the workload on the beta cell increases<br />

(by factors such as obesity, insulin resistance or low-grade inflammation), healthy beta cells<br />

can adapt by augmenting insulin secretion to meet this increased demand, thus maintaining<br />

euglycemia [101]. Before addressing the various aspects of beta-cell function that are altered<br />

by GC treatment, it is important to state that beta-cell function cannot be quantified by one<br />

single measure. Fasting measures include homeostatic model assessment of beta-cell function<br />

(HOMA-B) [102] and pro-insulin/insulin ratio (PI/I ratio), the latter giving an impression of<br />

insulin processing in the beta cell [103]. Dynamic parameters, including the insulinogenic<br />

index (IGI) and the total C-peptide response in relation to prevalent glucose levels (AUC / CPEP<br />

AUC ) may be obtained from oral and intravenous glucose challenge tests. In addition,<br />

GLUC<br />

300


mathematical models have been developed to calculate various parameters of beta-cell<br />

function from dynamic tests. In the model developed by Mari, a dose-response curve relating<br />

insulin secretion and glucose levels is calculated, from which glucose sensitivity of the beta<br />

cell is derived [104]. This dose-response curve is modulated by two additional factors. First,<br />

the insulin response to the rate of change of glucose concentrations is calculated as rate<br />

sensitivity. Secondly, the effects of non-glucose insulin secretagogues, including non-glucose<br />

substrates, gastrointestinal hormones and neurotransmitters are expressed as a potentiation<br />

factor [104]. The gold standard of beta-cell function measurements is the disposition index,<br />

where insulin secretion is adjusted for insulin sensitivity, both determined by clamp method.<br />

Thus, insulin secretion is assessed by the hyperglycemic clamp, whereas insulin sensitivity is<br />

obtained from the hyperinsulinemic-euglycemic clamp [105]. Arginine may additionally be<br />

added during the hyperglycemic clamp to obtain maximal secretory capacity at this of level of<br />

hyperglycemia [106].<br />

Direct effects of glucocorticoids on beta-cell function: in vitro studies<br />

As reviewed extensively in chapter 2, GCs were shown to directly impair insulin secretion in<br />

vitro in insulinoma cell lines and in rodent-derived islets [9]. GCs reduced glucose-stimulated<br />

insulin secretion (GSIS) by reducing glucose uptake and oxidation, resulting in reduced<br />

ATP synthesis and calcium influx. However, GCs also impaired more distal pathways in the<br />

insulin secretory process. As such, glucorticoid treatment reduced the activity of protein<br />

kinase A (PKA) and protein kinase C (PKC), two key enzymes involved in insulin exocytosis.<br />

These distal sites of interference by GCs in the insulin secretory process may explain the<br />

wide range of non-glucose insulin secretagogues that are also inhibited by GC treatment,<br />

including the PKA ligand ketoisocaproate, the amino acid arginine, the sulfonylurea drugs<br />

tolbutamide and gliclazide and the phorbolester PMA, which activates PKC [9]. Interestingly,<br />

GCs may also impair insulin secretion by disturbing autonomous nervous system (ANS)<br />

function. As such, GCs inhibited the phospholipase C (PLC)-PKC pathway, which is used by<br />

acetylcholine to induce insulin secretion [107]. In addition, GCs increased the expression of<br />

α2 adrenergic receptors, thus enhancing sympathetic activity, leading to reduced PKA activity<br />

and subsequent decreased insulin release [108, 109].<br />

In addition to reducing insulin release, GCs may display other detrimental effects on beta-cell<br />

function by several different mechanisms. First, GC treatment was shown to reduce insulin<br />

biosynthesis by reducing ATP/ADP ratio and pancreatic and duodenal homeobox (PDX)-1<br />

expression [73, 110, 111]. Second, in vitro studies have indicated that GCs induce beta-cell<br />

apoptosis, which may ultimately result in reduced beta-cell mass [112].<br />

301<br />

Chapter 15<br />

15


Chapter 15 General Discussion<br />

Interestingly, endoplasmic reticulum (ER) stress may be an important factor in these GCinduced<br />

beta-cell effects [73]. The ER is a highly dynamic cell organelle, which is responsible for<br />

the synthesis of all secreted proteins. In the ER, proteins are translated, folded and assessed for<br />

quality before they are released. In the beta cell, (pro)-insulin is the most abundant produced<br />

protein, and in the glycemia-stimulated state, it may account for up to 50% of total protein<br />

production. Sustained increased demand for insulin due to e.g. hyperglycemia, however,<br />

may impose burden or ‘stress’ on the ER. ER stress is characterized by an accumulation of<br />

misfolded proteins inside the organelle. In response, the so-called unfolded protein response<br />

(UPR) is initiated, which aims to restore ER homeostasis by decreasing ER protein load and by<br />

increasing folding capacity. When the UPR fails to alleviate ER stress, however, the UPR triggers<br />

apoptosis [113]. We observed that prednisolone induced ER stress and initiated the UPR as<br />

demonstrated by increased expression of activating transcription factor (ATF) 6 and inositolrequiring<br />

enzyme (IRE)-1/X-box binding protein (XBP)-1 pathways. These modulations of<br />

ER stress pathways were accompanied by upregulation of calpain 10 and increased cleaved<br />

caspase 3, confirming that exposure to prednisolone may promote apoptosis (chapter 3)<br />

[73].<br />

Acute effects of glucocorticoids on beta-cell function in humans<br />

Nearly four decades ago, the acute effects of GCs on glucose metabolism were studied for the<br />

first time. Cortisol infusion induced fasting hyperglycemia in the abscence of an appropriate<br />

insulin response [114], suggesting impaired beta-cell function. Moreover, 24-hr treatment<br />

with prednisolone 60 mg was shown to impair insulin secretion during low-dose glucose<br />

infusion [115]. We could confirm and extend these acute effects of prednisolone treatment<br />

on glucose metabolism and insulin secretion. Prednisolone 80 mg acutely impaired firstphase<br />

GSIS, arginine-induced insulin secretion and beta-cell disposition index as measured<br />

by hyperglycemic clamps in healthy males (chapter 7). In addition, treatment with a similar<br />

prednisolone dosage acutely increased postprandial glucose levels by impairing various betacell<br />

function parameters. These included empirical measures of beta-cell function such as<br />

IGI and AUC /AUC , but also model-derived parameters including glucose sensivity and<br />

CPEP gluc<br />

the potentiation factor (chapter 4). Thus, high-dose GC treatment acutely impairs beta-cell<br />

function resulting in postprandial glucose excursions.<br />

Subacute glucocorticoid treatment in healthy humans<br />

A number of studies have addressed the subacute effects of GCs on beta-cell function, typically<br />

with treatment duration of 3 days. We have extended these studies by prolonging the treatment<br />

duration to a two-week period as this more closely mimics clinical treatment schedules and<br />

by studying dose-dependency (chapter 5 and 6). The difficulty of studying the subacute<br />

302


effects of GCs on insulin secretion, however, is the fact that GCs also induce insulin resistance,<br />

which influences the response of the beta cell. As such, short-term treatment with high doses<br />

of dexamethasone or prednisolone in healthy subjects resulted in increased fasting insulin<br />

levels [23, 45, 46, 116-122] and increased insulin secretion during oral glucose tolerance tests<br />

(OGTT) [23, 118, 119], standardized meal tolerance tests [121, 122], intravenous glucose<br />

tolerance tests (IVGTT) [45, 120] and hyperglycemic clamp studies [23, 46, 116, 117, 121].<br />

This increased beta-cell response does not indicate improved beta-cell function, but likely<br />

compensation for GC-induced insulin resistance. As such, we observed that the increase<br />

in first- and second-phase GSIS was no longer significant when we adjusted for insulin<br />

sensitivity (M-value obtained by hyperinsulinemic-euglycemic clamp), i.e. that the disposition<br />

index remained unchanged following prednisolone treatment. Other studies calculating<br />

different variants of the disposition index, similarly reported adequate compensation in<br />

healthy volunteers. However, in susceptible populations, including normoglycemic insulin<br />

resistant individuals [46] or low GSIS [23, 117] prior to treatment with GCs, healthy, firstdegree<br />

relatives of patients with T2DM [45] and obese women [123], GSIS was not enhanced<br />

to such extent to compensate for GC-induced impairment of insulin sensitivity, and thus, the<br />

disposition index decreased following GC treatment.<br />

In addition to first- and second-phase GSIS, we additionally studied other parameters of<br />

beta-cell function during the two-week treatment. On top of the hyperglycemic clamp, 5 g of<br />

arginine was administered to assess maximal insulin secretion capacity at the level of 10 mM<br />

glucose. Interestingly, arginine-stimulated insulin secretion was dose-dependently reduced<br />

by prednisolone treatment. In addition, various aspects of beta-cell function were measured<br />

during standardized meal tests using the model developed by Mari as described. We observed<br />

that prednisolone reduced fasting insulin secretion rates when adjusted for prevailing glucose<br />

levels. In addition, the potentiation factor, which comprises insulin secretion induced by nonglucose<br />

stimuli, was reduced by both low- and high-dose prednisolone treatment (Figure 1).<br />

From both the combined hyperglycemic clamp and the meal test data we conclude that<br />

GC treatment induces beta-cell dysfunction in healthy individuals. This effect is detectable<br />

on specific beta-cell function parameters that seem to involve potentiation phenomena<br />

[104, 106, 124]. On the other hand, the enhancement of first- and second phase GSIS as<br />

well as postprandial insulin release is mainly mediated by an increase in glucose levels<br />

and a compensatory response to GC-induced insulin resistance. Although it appeared that<br />

prednisolone already at a low dose impaired several aspects of beta-cell function, our study<br />

was limited in its power to detect these changes.<br />

303<br />

Chapter 15<br />

15


Chapter 15 General Discussion<br />

Mechanisms of glucocorticoid-induced beta-cell function<br />

The mechanisms underlying GC-induced beta-cell dysfunction following short-term<br />

treatment presently remain unclear. It is unlikely that chronic processes such as glucotoxicity,<br />

lipotoxicity, deposition of amyloid or loss of beta-cell volume due to extensive apoptosis<br />

are involved. Given GC-induced insulin resistance, ER stress, on the other hand, could be a<br />

mechanism limiting the production of a bioactive insulin molecule. In addition we found<br />

evidence that impaired sympathovagal balance may contribute to beta-cell dysfunction. As<br />

detailed previously, whereas the parasympathetic nervous system (PNS; neurotransmitter:<br />

acetylcholine) promotes insulin secretion, this is inhibited by the sympathetic nervous<br />

system (SNS; neurotransmitter: adrenaline). In vitro, dexamethasone was to reduce insulin<br />

secretion by increasing α2 adrenergic receptors [108, 109] and by impairing acetylcholineinduced<br />

insulin secretion [125]. In our healthy participants, we observed a tendency towards<br />

increased SNS over PNS activity as assessed by cardiovascular ANS measurements derived from<br />

continuous beat-to-beat finger blood pressure recordings, which was associated with fasting<br />

glucose levels and arginine-stimulated insulin secretion (chapter 5). Further experimental<br />

data are needed to clarify the role of ANS in prednisolone-induced beta-cell effects.<br />

Chronic glucorticoid treatment in healthy humans<br />

At present there are no data available from studies addressing the chronic effects of GCs<br />

on beta-cell function in healthy individuals, due to the obvious ethical reasons. However,<br />

the chronic effects of GCs on beta-cell function have been addressed in patients with RA, as<br />

detailed below. However, it is difficult to compare these data to the results obtained in healthy<br />

volunteers, since chronic inflammation also compromises both beta-cell function[126] and<br />

insulin sensitivity [127].<br />

Glucocorticoid receptor polymorphisms and beta-cell function<br />

GCs exert many effects by binding to its cytosolic GR, following which the ligand-activated<br />

GR translocates to the nucleus where it regulates target gene transcriptional activity [128-<br />

130]. Functional single nucleotide polymorphisms (SNPs) in the GR gene (NR3C1), which<br />

demonstrated altered GC sensitivity in vitro, were linked to various metabolic parameters<br />

in vivo [128-130]. Since pancreatic GR overexpression in mice resulted in the development<br />

of beta-cell dysfunction and diabetes [131], we hypothesized that alterations in the GR gene<br />

could be related to measures of beta-cell function. We addressed this hypothesis in 449<br />

subjects with normal- or impaired glucose tolerance, who underwent a hyperglycemic clamp.<br />

We observed that the N363S and ER22/23EK polymorphisms were negatively associated<br />

with first-phase GSIS and disposition index in women, but not in men [132]. The N363S<br />

304


SNP displays increased GC sensitivity in vitro [133] and is associated with features of a<br />

Cushingoid phenotype including increased BMI, waist-to-hip ratio, dyslipidemia and fasting<br />

hyperinsulinemia [130, 134]. The ER22/23EK SNP on the other hand, demonstrated reduced<br />

GR activation in vitro, and relative GC resistance in vivo. As such, in men, the ER22/23EK<br />

was associated with a beneficial metabolic phenotype, including increased muscle mass<br />

and strength, lower LDL cholesterol and insulin levels [135, 136]. On the other hand, female<br />

carriers of the ER22/23EK SNP were at increased risk to develop cardiovascular disease<br />

(CVD) [137]. In another cohort, carriers of the ER22/23EK had higher HbA1c levels as<br />

compared to noncarriers, thus raising doubt on the hypothesis that this SNP may induce a<br />

more favourable metabolic profile, especially in women [138]. Although we only observed<br />

the negative association between these NR3C1 SNPs and beta-cell function parameters in<br />

women, the outcome of this study strengthens the evidence for a key role of pancreatic betacell<br />

dysfunction in the development of GCs diabetogenic effects.<br />

5. Pancreatic Alpha Cells<br />

By secreting glucagon, the pancreatic alpha cell has an important role in glucose metabolism<br />

[139]. As previously mentioned, glucagon stimulates hepatic glucose production by<br />

promoting glycogenolysis and gluconeogenesis [140]. In many patients with T2DM, glucagon<br />

levels are increased in the fasted state and are incompletely suppressed in the postprandial<br />

state. Thus, elevated glucagon levels were shown to contribute importantly to both fasting<br />

and postprandial hyperglycemia [141].<br />

Already in 1971, GCs were shown to augment glucagon levels. This was demonstrated both<br />

in healthy persons treated with dexamethasone 2 mg daily for 3 days and in patients with<br />

Cushing’s syndrome. In both groups, fasting glucagon levels were increased and glucagon<br />

concentrations were incompletely suppressed following ingestion of a protein meal or<br />

following alanine infusion [142]. After this single study, the effects of GCs on glucagon levels<br />

were not studied for another 4 decades. Here, we studied the dose-dependent effects of GCs<br />

on glucagon levels and observed that a two-week treatment with low-dose prednisolone<br />

treatment (7.5 mg daily) did not alter fasting or postprandial glucagon levels, however,<br />

that high-dose prednisolone treatment (30 mg daily) administered for this time period<br />

increased fasting glucagon levels and impaired the suppression of glucagon levels following<br />

a standardized meal test and during insulin infusion (chapter 5 and 8) [28, 121]. These<br />

effects of GC treatment were already observed in the acute setting, since a single gift of 80 mg<br />

prednisolone increased postprandial glucagon levels, as described in chapter 7 [143]. Similar<br />

results were obtained by Hansen and colleagues, who observed increased glucagon levels<br />

following a 12-day combined treatment with prednisolone 37.5 mg daily, physical inactivity<br />

305<br />

Chapter 15<br />

15


Chapter 15 General Discussion<br />

and a hypercaloric diet. Glucagon levels were increased in the fasted state and incompletely<br />

suppressed during an OGTT and isoglycemic intravenous glucose infusion [118, 119]. We<br />

conclude that both fasting and postprandial hyperglucagonemia are present in GC-induced<br />

hyperglucagonemia (Figure 1).<br />

6. The Gut<br />

The incretin hormones, glucagon-like peptide-1 (GLP-1) and glucose-dependent<br />

insulinotrophic polypeptide (GIP) are hormones secreted by the gut that are released<br />

following nutrient ingestion [144]. GLP-1 lowers (postprandial) blood glucose through<br />

several mechanisms, including stimulation of meal-related insulin response and suppression<br />

of glucagon secretion in a glucose-dependent manner. In addition, GLP-1 slows down gastric<br />

emptying, promotes satiety, decreases appetite and reduces body weight. In human beta<br />

cells and in vivo in animals and in humans, exogenous GLP-1 administration, in the presence<br />

of elevated blood glucose, acutely induces insulin-secretion while more prolonged GLP-1<br />

exposure may result in increased insulin production. Interestingly, in rodents, chronic GLP-<br />

1 treatment was shown to increase beta-cell mass by promoting beta-cell regeneration and<br />

proliferation, and inhibiting apoptosis [145, 146]. In addition, GLP-1 treatment may protect<br />

beta cells against several toxic processes, including ER stress [147].<br />

When a similar level of glycemia is induced by glucose ingested by the oral versus the<br />

intravenous route in healthy individuals, the oral glucose yields an insulin response that is<br />

approximately 50-70% higher than the intravenous administered glucose. In patients with<br />

T2DM, this so-called incretin effect is substantially diminished [148]. Although post-load GLP-<br />

1 secretion was found to be reduced in some studies, but not in others, the reduced incretin<br />

effect in T2DM seems rather attributed to a decreased sensitivity of beta cells to endogenous<br />

GLP-1 and GIP [149]. In T2DM patients, a vast body of research [144] indicates that an<br />

impaired incretin effect contributes substantially to (postprandial) hyperglycemia.<br />

Up to recently, the effects of GCs on both GLP-1 and GIP secretion and their insulinotropic<br />

actions have been little addressed. At the onset of our studies, we speculated that GC<br />

treatment would reduce secretion of GLP-1 and GIP based on data obtained in rodents, where<br />

GC treatment resulted in reduced mRNA stability of the preproglucagon gene, a precursor<br />

of GLP-1 [150]. However, during our standardized meal tests, GLP-1 and GIP levels were<br />

not reduced by low- or high-dose prednisolone treatment (chapter 5) [23]. In contrast, a<br />

trend towards increased levels of GIP was observed in the prednisolone 30 mg daily group.<br />

A similar observation was made in two recent studies by another research group, who found<br />

that high-dose prednisolone in combination with physical inactivity and high calorie diet did<br />

306


not affect GLP-1 secretion but increased GIP levels during an OGTT and liquid meal test [118,<br />

119]. Interestingly, the insulinotropic effects of GLP-1 and/or GIP were shown to be reduced,<br />

since the incretin effect was significantly decreased [118]. Indeed, the potentiation of GSIS<br />

by incretin hormones was shown to be reduced in a recent study [151]. It is unclear whether<br />

this impaired incretin effect is a specific defect induced by GC treatment or whether it may<br />

be secondary to general GC-induced beta-cell dysfunction, as detailed previously. Thus, an<br />

impaired gut-islet axis characterized by impaired insulinotropic effects of GLP-1 and GIP is<br />

present in GC-induced hyperglycemia (Figure 1).<br />

7. Microcirculation<br />

As discussed to extent previously, GCs dose-dependently induce insulin resistance at the<br />

level of liver, adipose tissue and skeletal muscle, resulting in impaired suppression of EGP<br />

and reduced insulin-stimulated glucose uptake [28]. However, insulin action is not restricted<br />

to these metabolically active tissues. It is now evident that vascular tissue, and particularly<br />

endothelial cells, represents an important physiological target for insulin. Insulin exerts<br />

a vasodilatory action by promoting nitric oxide (NO) release from the endothelial cells.<br />

This involves the phosphorylation of endothelial NO synthase (eNOS), which on its turn<br />

is mediated by the PI3K/Akt pathway [152]. By increasing blood flow and by recruiting<br />

capillaries to expand the endothelial transporting surface available for nutrient exchange,<br />

the vascular actions of insulin significantly contribute to overall insulin-stimulated glucose<br />

uptake [153-155]. In addition, the local actions of insulin on (micro)vascular tissue may be<br />

important in the regulation of blood pressure and the pathogenesis of hypertension [7, 156,<br />

157]. Prior to our experiments (chapter 10), the effects of GCs on microvascular function, in<br />

particular in the presence of insulin, in vivo in humans had not been investigated. In addition,<br />

it was unclear whether impairments in microvascular function could contribute to GCinduced<br />

hyperglycemia. An indication that GCs may impair microvascular function came from<br />

observations in resistance arterioles isolated from C57BL/6j mice. Treatment of these mice<br />

with dexamethasone suppressed endothelial eNOS protein levels and impaired endotheliumdependent<br />

vasodilation [158].<br />

We assessed the effects of a two-week treatment with low- and high-dose prednisolone<br />

on microvascular function, as measured by capillary microscopy, in healthy humans both<br />

in the fasted state and during insulin infusion. Prednisolone dose-dependently impaired<br />

capillary recruitment during insulin infusion, but not in the fasted state, when prednisolone<br />

induced fasting hyperglycemia. Moreover, prednisolone-induced impairment in insulinstimulated<br />

capillary recruitment was related to insulin-stimulated glucose disposal<br />

during the hyperinsulinemic-euglycemic clamp, as well as to postprandial glucose levels<br />

307<br />

Chapter 15<br />

15


Chapter 15 General Discussion<br />

and postprandial insulin sensitivity, suggesting that capillary recruitment may also be an<br />

important factor contributing to postprandial glucose metabolism. In addition, the decrease<br />

in insulin-stimulated capillary recruitment was associated with the prednisolone-induced<br />

increase in systolic blood pressure. In isolated cremaster resistance arteries of prednisolonetreated<br />

male Wistar rats, the vasodilatory actions of insulin were similarly attenuated.<br />

At the molecular level, prednisolone impaired the vascular effects of insulin by reducing<br />

phosphorylation of the Akt pathway, thus providing a mechanism for the vascular insulin<br />

resistance observed in vivo [53]. In summary, prednisolone impairs the vasodilatory actions<br />

of insulin at the level of the microcirculation by attenuating insulin signaling through the Akt<br />

pathway, resulting in impaired glucose disposal, impaired glucose tolerance and increments<br />

in systolic blood pressure. Thus, impaired microvascular function may be regarded among<br />

the mechanisms by which GC act to exert their diabetogenic effects. Figure 1 summarizes the<br />

adverse effects of GCs on glucose metabolism.<br />

8. other potential glucocorticoid-target organs<br />

A number of organs that were not addressed in the sections above may also be involved in<br />

GC-associated adverse metabolic effects. The brain most likely represents a very important<br />

contributor to GC-induced hyperglycemia, but this area was not studied in our conducted trials,<br />

which focused on the peripheral effects of GC treatment. Indeed, GRs are expressed in various<br />

part of the brains, including the hypothalamus [159]. In rodents, intracerebroventricular<br />

infusion of dexamethasone increased food intake [160] and decreased peripheral glucose<br />

uptake [161]. In addition, it was recently shown that dexamethasone administration in the<br />

arcuate nucleus of the hypothalamus resulted in hepatic insulin resistance through activation<br />

of sympathetic pathways to the liver [162].<br />

In addition, the kidney has been shown to be play a role in glucose homeostasis. Glucose is<br />

freely filtered by the glomerulus and is reabsorbed in the proximal tubule mainly by the sodium<br />

glucose transport protein (SGTL)2. Increased SGTL2 activity, particularly in the presence of<br />

hyperglycemia, stimulates tubular glucose absorption and may sustain hyperglycemia [163].<br />

Interestingly, in a mouse sepsis model, GCs were shown to decrease glucose excretion by<br />

upregulating SGLT2 [163]. The role of the kidney in GC-induced hyperglycemia in humans,<br />

however, remains to be established.<br />

Finally, GCs were shown to alter cardiac glucose and lipid metabolism, possibly by altering<br />

cardiac insulin sensitivity. Dexamethasone treatment was shown to reduce glucose oxidation<br />

in myocytes in rodents. In addition, GC treatment increased NEFA uptake and oxidation, but<br />

was also shown to promote myocardial NEFA overload [164]. Importantly, the latter has been<br />

308


linked to the development of cardiomyopathy [164]. Although the effects of GCs on cardiac<br />

substrate metabolism could in part explain the GC-associated increased risk to develop heart<br />

failure or cardiovascular disease [165], it is presently unclear whether changes in cardiac<br />

metabolism may contribute to the systemic diabetogenic effects of GC treatment. Although<br />

the brain, kidneys and heart seem important GC target organs, they were beyond the scope<br />

of our studies.<br />

Glucocorticoids diabetogenic effects and interaction with inflammation:<br />

studies in rheumatoid arthritis patients<br />

Addressing glucocorticoids metabolic effects in RA patients is interesting for several reasons.<br />

First, in RA patients GCs are used on large scale [2], thus rendering these patients into a<br />

very clinically relevant population. In addition, in RA patients, it is possible to examine the<br />

interaction of GC treatment and systemic inflammation, of which the latter also affects both<br />

beta-cell function [126] and insulin sensitivity [127]. Moreover, some RA patients are treated<br />

chronically with GCs, thus providing data that extend the short-term studies conducted in<br />

healthy individuals.<br />

Irrespective of treatment given, RA patients are at increased risk to develop impaired<br />

glucose metabolism and eventually T2DM likely due to chronic inflammation [166]. As such,<br />

correlations were found between insulin resistance, glucose tolerance and various markers of<br />

systemic inflammation in several studies [167-171]. The metabolic effects GC treatment in RA<br />

population, however, are a topic of extensive discussion.<br />

Short-term intervention studies<br />

Reduction of inflammation by GC treatment was shown to improve glucose tolerance in<br />

two short-term studies [172, 173]. We similarly studied the short-term effects of high-dose<br />

(60 mg or 30 mg once daily administered for 1 week) prednisolone treatment in recentlydiagnosed,<br />

drug-naive RA patients with active disease (chapter 13) [170]. Glucose tolerance,<br />

beta-cell function and insulin sensitivity were measured during frequently-sampled OGTTs.<br />

Following treatment, we observed no deterioration of glucose tolerance in the entire group,<br />

however, there was considerable between-subjects variability. The incidence of T2DM<br />

increased from 3% to 10%, but a significant number of patients also improved their glucose<br />

tolerance state. The duration of disease was the most important predictor to determine<br />

whether patients progressed to T2DM or improved their glucometabolic state. Fasting insulin<br />

sensitivity (HOMA-IR) was reduced by treatment, but this was compensated for by enhanced<br />

beta-cell response during the OGTT. To conclude, it remains unclear whether the reduction of<br />

inflammation may completely off set the adverse metabolic effects induced by GCs, however,<br />

309<br />

Chapter 15<br />

15


Chapter 15 General Discussion<br />

short-term treatment in RA patients does not seem to be complicated by marked glucose<br />

intolerance as is characterized in healthy subjects, although inter-individual variability is<br />

considerable (chapter 13) (Figure 2).<br />

Figure 2. Hypothetic representation of the balance between systemic inflammation severity and<br />

(high)-dose glucocorticoid (GC) treatment in relation to glucose tolerance. In healthy volunteers, the<br />

diabetogenic effects of GCs are well described. However, in patients with inflammation, GCs may improve<br />

glucose tolerance by reducing systemic inflammation. In patients treated with GCs for conditions that<br />

are usually characterized by low-grade systemic inflammation, such as chronic obstructive pulmonary<br />

disease (COPD), the diabetogenic effects of GC treatment may dominate its anti-inflammatory actions<br />

with respect to glucose metabolism. However, in patients with high-grade inflammation, such as recently<br />

diagnosed rheumatoid arthritis (RA) patients, GCs anti-inflammatory actions may outweigh their<br />

diabetogenic effects. (See page 367 for full color figure)<br />

Cross-sectional studies in chronic rheumatoid arthritis patients<br />

In a number of small cross-sectional studies, chronic GC exposure was related to impaired<br />

fasting insulin sensitivity as measured by HOMA-IR [174] and tended to predict T2DM,<br />

although in these studies the presence of a chronic disease (i.e. RA) affecting these<br />

parameteres was poorly addressed [175, 176]. In order to further characterize the effects of<br />

GCs in chronic RA patients, we recruited a large cohort of chronic RA patients treated with<br />

GCs for over 3 months (RA+GC; n=58) and assessed glucose tolerance, beta-cell function<br />

and insulin sensitivity by frequently-sampled OGTTs. This group was compared to chronic<br />

310


RA patients who were GC-naïve (RA-GC; n=82) (chapter 14). We observed no differences in<br />

glucose tolerance and static or dynamic measures of beta-cell function and insulin sensitivity<br />

between groups, however, cumulative GC dosage was independently associated with HOMA-<br />

IR and T2DM prevalence, also after adjusting for disease activity and patient characteristics.<br />

However, in this cross-sectional study, confounding by indication should be kept in mind. This<br />

indicates that cumulative GC use might be a proxy for long-term disease activity, which itself<br />

influences glucose metabolism, since GCs are mostly given to the patients with refractory<br />

disease activity [171]. We concluded that chronic GC use may be related to T2DM as compared<br />

to other RA medications, however, since these findings were not substantiated in randomized<br />

controlled trials, no definitive conclusions can be drawn.<br />

From the studies in RA patients it is evident that the metabolic effects of GCs are much less<br />

pronounced than in healthy controls, as GC may simultaneously alleviate the diabetogenic<br />

effects of the inflammatory disease. The amount of systemic inflammation when GC therapy<br />

is initiated may be an important determinant of the net effects of GC treatment on glucose<br />

tolerance (Figure 2).<br />

Glucocorticoids diabetogenic effects: implications for the development of dissociated<br />

glucocorticoid receptor agonists<br />

A number of lessons may be appreciated from the above-summarized studies for the current<br />

development of dissociated GR agonists. First, it is clear that various organ systems and<br />

different pathways contribute to the diabetogenic effects of GCs. This should be kept in mind<br />

when assessing the metabolic effects of novel GR modulators. In addition, we have shown that<br />

several unfavorable effects (i.e. increased postprandial glucose levels, impaired suppression<br />

of EGP and lipolysis by insulin) may already develop during treatment with dosages as low<br />

as 7.5 mg prednisolone daily (chapters 5 and 8) [28, 121] (Table 1), which were previously<br />

considered to be ‘safe’. Thus, ideally, the novel GR agonists should display anti-inflammatory<br />

properties similar to high-dose prednisolone treatment, but should have a side-effect profile<br />

of an equivalent prednisolone dose lower than 7.5 mg daily. Finally, since many metabolic<br />

adverse effects become apparent in the postprandial state or during hyperinsulinemia<br />

(detailed below), the metabolic effects of classic GR agonists and dissociated GR agonists,<br />

when compared in future head-to-head studies in humans, should be assessed using dynamic<br />

tests such as OGTTs and clamp tests, since fasting measures may underestimate the GCinduced<br />

effects.<br />

311<br />

Chapter 15<br />

15


Chapter 15 General Discussion<br />

Table 1. Adverse metabolic effects induced by low-dose glucocorticoid treatment in healthy men<br />

as observed in our studies.<br />

Glucose levels<br />

312<br />

Elevated fasting plasma glucose levels*<br />

Postprandial hyperglycemia<br />

Potentially involved mechanisms<br />

Liver Impaired suppression of EGP by insulin<br />

Skeletal muscle Reduced insulin-stimulated glucose uptake*<br />

Adipose tissue Impaired suppression of lipolysis by insulin<br />

Increased NEFA levels during hyperinsulinemia<br />

Decreased markers of adipogenesis in SAT<br />

Decreased adiponectin expression in SAT<br />

Beta cells Reduced glucose-adjusted basal insulin secretion*<br />

Impaired potentiation of insulin secretion by non-glucose stimuli*<br />

Postprandial midday hyperinsulinemia<br />

* Indicates trend. EGP: endogenous glucose production; NEFA: non-esterified fatty acids; SAT:<br />

subcutaneous adipose tissue.<br />

Glucocorticoids diabetogenic effects: from mechanism to treatment?<br />

Despite the vast number of documented observations clearly showing that glucocorticoids<br />

induce glucose intolerance and diabetes in the clinic [14-16], remarkably, there are no studies<br />

that have investigated how GC-induced diabetes may best be treated, or preferentially, could be<br />

prevented. It has become increasingly clear that the most pronounced effects of GC treatment<br />

are in the postprandial period. We could demonstrate increased postprandial glucose<br />

levels, increased EGP, lipolysis and proteolysis, and impaired capillary recruitment during<br />

hyperinsulinemic conditions, whereas all these parameters were not affected in the fasted<br />

state when prednisolone treatment induced fasting hyperinsulinemia (chapters 4,5,7,8<br />

and 10) [28, 53, 121, 122, 143]. In line with these data, patients with Cushing’s syndrome<br />

have predominantly increased postprandial glucose levels as opposed to near normal fasting<br />

glucose levels [177]. And more recently, similar observations have also been observed in<br />

clinical studies in patients treated with medium-to-high dose GCs [16, 178, 179]. Patients<br />

with chronic obstructive pulmonary disease (COPD) that were treated with GCs at 08.00 in<br />

the morning displayed significant glucose elevation resulting in afternoon and evening, but<br />

not overnight, hyperglycemia [179]. In another study, this phenomenon was explained by an<br />

early inhibition of insulin secretion followed by decreased insulin action later during the day.<br />

All metabolic parameters were completely restored by the next morning [178].


These observations have important implications for treatment of GC-induced hyperglycemia.<br />

Glucose-lowering therapy should be predominantly directed at the time period between<br />

midday and midnight, and caution should be exercised with the use of long-acting basal<br />

insulin, because it may precipitate nocturnal hypoglycemia when the effect of prednisolone<br />

wanes. Given this specific period of time, short-acting insulin analogues seem a reasonable<br />

choice to treat the typical pattern of hyperglycemia.<br />

In this thesis, we have also explored the potential of GLP-1 based treatment to treat GCinduced<br />

hyperglycemia [143]. Since GLP-1 treatment stimulates insulin secretion [180, 181],<br />

reduces glucagon secretion [182] and improves postprandial insulin sensitivity [183, 184], it<br />

addresses at least three important pathophysiological features of GC-induced hyperglycemia.<br />

Indeed, in a proof-of-concept study in healthy volunteers, infusion of the GLP-1 receptor<br />

agonist (GLP-1 RA) exenatide prevented glucose intolerance and islet-cell dysfunction<br />

induced by acute prednisolone treatment (chapter 7) [143]. In addition, in the long term,<br />

GLP-1 RA reduce appetite [185], stimulate weight loss [186], decrease visceral fat tissue<br />

[187] and hepatic steatosis [188], improve postprandial lipid profiles [189] and favorably<br />

change adipose tissue biology [187], effects that could counteract the features associated with<br />

chronic GC treatment. Currently, studies are ongoing in which the effects of more prolonged<br />

treatment with the dipeptidyl-peptidase (DPP)-4 inhibitor sitagliptin on glucose metabolism<br />

are assessed in men with the metabolic syndrome concomitantly treated with high-dose<br />

prednisolone [190]. It will be interesting to see whether also in this population, incretinbased<br />

therapies may be useful in the treatment of GC-induced hyperglycemia.<br />

Thus, expanding knowledge of the pathophysiology of the diabetogenic effects of GC treatment<br />

should in due time result in a more tailored therapy to treat the associated hyperglycemia.<br />

313<br />

Chapter 15<br />

15


Chapter 15 General Discussion<br />

REFERENCES<br />

1. Zen M, Canova M, Campana C, Bettio S, Nalotto L, Rampudda M, Ramonda R, Iaccarino L, Doria A.<br />

The kaleidoscope of glucorticoid effects on immune system. Autoimmun Rev. 2011;10(6):305-10.<br />

2. Bijlsma jW, van der Goes MC, Hoes jN, jacobs jW, Buttgereit F, Kirwan j. Low-dose glucocorticoid<br />

therapy in rheumatoid arthritis: an obligatory therapy. Ann N Y Acad Sci. 2010;1193:123-6.<br />

3. Hernandez-Rodriguez j, Cid MC, Lopez-Soto A, Espigol-Frigole G, Bosch X. Treatment of polymyalgia<br />

rheumatica: a systematic review. Arch Intern Med. 2009;169(20):1839-50.<br />

4. Parker Bj, Bruce IN. High dose methylprednisolone therapy for the treatment of severe systemic<br />

lupus erythematosus. Lupus. 2007;16(6):387-93.<br />

5. Salvarani C, Cantini F, Hunder GG. Polymyalgia rheumatica and giant-cell arteritis. Lancet.<br />

2008;372(9634):234-45.<br />

6. Lichtenstein GR, Abreu MT, Cohen R, Tremaine W, American Gastroenterological A. American<br />

Gastroenterological Association Institute medical position statement on corticosteroids,<br />

immunomodulators,<br />

2006;130(3):935-9.<br />

and infliximab in inflammatory bowel disease. Gastroenterology.<br />

7. Rabe KF, Wedzicha jA. Controversies in treatment of chronic obstructive pulmonary disease. Lancet.<br />

2011;378(9795):1038-47.<br />

8. Hillier SG. Diamonds are forever: the cortisone legacy. j Endocrinol. 2007;195(1):1-6.<br />

9. van Raalte DH, Ouwens DM, Diamant M. Novel insights into glucocorticoid-mediated diabetogenic<br />

effects: towards expansion of therapeutic options? Eur j Clin Invest. 2009;39(2):81-93.<br />

10. Schacke H, Docke WD, Asadullah K. Mechanisms involved in the side effects of glucocorticoids.<br />

<strong>Pharma</strong>col Ther. 2002;96(1):23-43.<br />

11. van Tuyl LH, Plass AM, Lems WF, Voskuyl AE, Dijkmans BA, Boers M. Why are Dutch rheumatologists<br />

reluctant to use the COBRA treatment strategy in early rheumatoid arthritis? Ann Rheum Dis.<br />

2007;66(7):974-6.<br />

12. Compston j. Management of glucocorticoid-induced osteoporosis. Nat Rev Rheumatol. 2010;6(2):82-<br />

8.<br />

13. Trikudanathan S, McMahon GT. Optimum management of glucocorticoid-treated patients. Nat Clin<br />

Pract Endocrinol Metab. 2008;4(5):262-71.<br />

14. Gulliford MC, Charlton j, Latinovic R. Risk of diabetes associated with prescribed glucocorticoids in<br />

a large population. Diabetes Care. 2006;29(12):2728-9.<br />

15. Gurwitz jH, Bohn RL, Glynn Rj, Monane M, Mogun H, Avorn j. Glucocorticoids and the risk for<br />

initiation of hypoglycemic therapy. Arch Intern Med. 1994;154(1):97-101.<br />

16. Clore jN, Thurby-Hay L. Glucocorticoid-induced hyperglycemia. Endocr Pract. 2009;15(5):469-74.<br />

314


17. Rosen j, Miner jN. The search for safer glucocorticoid receptor ligands. Endocr Rev. 2005;26(3):452-<br />

64.<br />

18. Schacke H, Rehwinkel H, Asadullah K, Cato AC. Insight into the molecular mechanisms of<br />

glucocorticoid receptor action promotes identification of novel ligands with an improved therapeutic<br />

index. Exp Dermatol. 2006;15(8):565-73.<br />

19. Schacke H, Berger M, Rehwinkel H, Asadullah K. Selective glucocorticoid receptor agonists (SEGRAs):<br />

novel ligands with an improved therapeutic index. Mol Cell Endocrinol. 2007;275(1-2):109-17.<br />

20. van Lierop Mj, Alkema W, Laskewitz Aj, Dijkema R, van der Maaden HM, Smit Mj, Conti PGM, jams<br />

CGjM, Timmers M, van Boeckel SCAA, Lusher Sj, McGuire R, van Schaik RC, de Vlieg j, Smeets RL,<br />

Hofstra CL, Boots AMH, van Duin M, Ingelse BA, Schoonen WGEJ, Grefhorst A, van Dijk T, Kuipers<br />

F, Dokter WHA. A novel non-steroidal selective glucocorticoid receptor modulator with full antiinflammatory<br />

properties and improved therapeutic index. Submitted. 2011.<br />

21. Rizza RA, Mandarino Lj, Gerich jE. Cortisol-induced insulin resistance in man: impaired suppression<br />

of glucose production and stimulation of glucose utilization due to a postreceptor detect of insulin<br />

action. j Clin Endocrinol Metab. 1982;54(1):131-8.<br />

22. Rooney DP, Neely RD, Cullen C, Ennis CN, Sheridan B, Atkinson AB, Trimble ER, Bell PM. The effect of<br />

cortisol on glucose/glucose-6-phosphate cycle activity and insulin action. j Clin Endocrinol Metab.<br />

1993;77(5):1180-3.<br />

23. Wajngot A, Giacca A, Grill V, Vranic M, Efendic S. The diabetogenic effects of glucocorticoids are more<br />

pronounced in low- than in high-insulin responders. Proc Natl Acad Sci U S A. 1992;89(13):6035-9.<br />

24. Nielsen MF, Caumo A, Chandramouli V, Schumann WC, Cobelli C, Landau BR, Vilstrup H, Rizza<br />

RA, Schmitz O. Impaired basal glucose effectiveness but unaltered fasting glucose release and<br />

gluconeogenesis during short-term hypercortisolemia in healthy subjects. Am j Physiol Endocrinol<br />

Metab. 2004;286(1):E102-10.<br />

25. Miyoshi H, Shulman GI, Peters Ej, Wolfe MH, Elahi D, Wolfe RR. Hormonal control of substrate cycling<br />

in humans. j Clin Invest. 1988;81(5):1545-55.<br />

26. [26] Schneiter P, Tappy L. Kinetics of dexamethasone-induced alterations of glucose metabolism in<br />

healthy humans. Am j Physiol. 1998;275(5 Pt 1):E806-13.<br />

27. Wajngot A, Khan A, Giacca A, Vranic M, Efendic S. Dexamethasone increases glucose cycling, but not<br />

glucose production, in healthy subjects. Am j Physiol. 1990;259(5 Pt 1):E626-32.<br />

28. van Raalte DH, Brands M, van der Zijl Nj, Muskiet MH, Pouwels Pj, Ackermans MT, Sauerwein HP,<br />

Serlie Mj, Diamant M. Low-dose glucocorticoid treatment affects multiple aspects of intermediary<br />

metabolism in healthy humans: a randomised controlled trial. Diabetologia. 2011;54(8):2103-12.<br />

29. jin jY, DuBois DC, Almon RR, jusko Wj. Receptor/gene-mediated pharmacodynamic effects of<br />

methylprednisolone on phosphoenolpyruvate carboxykinase regulation in rat liver. j <strong>Pharma</strong>col Exp<br />

Ther. 2004;309(1):328-39.<br />

315<br />

Chapter 15<br />

15


Chapter 15 General Discussion<br />

30. Sasaki K, Cripe TP, Koch SR, Andreone TL, Petersen DD, Beale EG, Granner DK. Multihormonal<br />

regulation of phosphoenolpyruvate carboxykinase gene transcription. The dominant role of insulin.<br />

j Biol Chem. 1984;259(24):15242-51.<br />

31. Vander Kooi BT, Onuma H, Oeser JK, Svitek CA, Allen SR, Vander Kooi CW, Chazin WJ, O’Brien RM.<br />

The glucose-6-phosphatase catalytic subunit gene promoter contains both positive and negative<br />

glucocorticoid response elements. Mol Endocrinol. 2005;19(12):3001-22.<br />

32. Vegiopoulos A, Herzig S. Glucocorticoids, metabolism and metabolic diseases. Mol Cell Endocrinol.<br />

2007;275(1-2):43-61.<br />

33. Frijters R, Fleuren W, Toonen Ej, Tuckermann jP, Reichardt HM, van der Maaden H, van Elsas A, van<br />

Lierop Mj, Dokter W, de Vlieg j, Alkema W. Prednisolone-induced differential gene expression in<br />

mouse liver carrying wild type or a dimerization-defective glucocorticoid receptor. BMC Genomics.<br />

2010;11:359.<br />

34. Patel R, Patel M, Tsai R, Lin V, Bookout AL, Zhang Y, Magomedova L, Li T, Chan jF, Budd C, Mangelsdorf<br />

Dj, Cummins CL. LXRbeta is required for glucocorticoid-induced hyperglycemia and hepatosteatosis<br />

in mice. j Clin Invest. 2011;121(1):431-41.<br />

35. Bernal-Mizrachi C, Weng S, Feng C, Finck BN, Knutsen RH, Leone TC, Coleman T, Mecham RP, Kelly DP,<br />

Semenkovich CF. Dexamethasone induction of hypertension and diabetes is PPAR-alpha dependent<br />

in LDL receptor-null mice. Nat Med 2003;9(8):1069-75.<br />

36. Zimmerman T, Horber F, Rodriguez N, Schwenk WF, Haymond MW. Contribution of insulin resistance<br />

to catabolic effect of prednisone on leucine metabolism in humans. Diabetes. 1989;38(10):1238-44.<br />

37. Kraus-Friedmann N. Hormonal regulation of hepatic gluconeogenesis. Physiol Rev. 1984;64(1):170-<br />

259.<br />

38. Whorwood CB, Donovan Sj, Flanagan D, Phillips DI, Byrne CD. Increased glucocorticoid receptor<br />

expression in human skeletal muscle cells may contribute to the pathogenesis of the metabolic<br />

syndrome. Diabetes. 2002;51(4):1066-75.<br />

39. Dirlewanger M, Schneiter PH, Paquot N, jequier E, Rey V, Tappy L. Effects of glucocorticoids on<br />

hepatic sensitivity to insulin and glucagon in man. Clin Nutr. 2000;19(1):29-34.<br />

40. Saad Mj, Folli F, Kahn jA, Kahn CR. Modulation of insulin receptor, insulin receptor substrate-1,<br />

and phosphatidylinositol 3-kinase in liver and muscle of dexamethasone-treated rats. j Clin Invest.<br />

1993;92(4):2065-72.<br />

41. Zinker B, Mika A, Nguyen P, Wilcox D, Ohman L, von Geldern TW, Opgenorth T, jacobson P. Liverselective<br />

glucocorticoid receptor antagonism decreases glucose production and increases glucose<br />

disposal, ameliorating insulin resistance. Metabolism. 2007;56(3):380-7.<br />

42. Nader N, Ng SS, Lambrou GI, Pervanidou P, Wang Y, Chrousos GP, Kino T. AMPK regulates metabolic<br />

actions of glucocorticoids by phosphorylating the glucocorticoid receptor through p38 MAPK. Mol<br />

Endocrinol. 2010;24(9):1748-64.<br />

43. Towler MC, Hardie DG. AMP-activated protein kinase in metabolic control and insulin signaling. Circ<br />

Res. 2007;100(3):328-41.<br />

316


44. DeFronzo RA, jacot E, jequier E, Maeder E, Wahren j, Felber jP. The effect of insulin on the disposal<br />

of intravenous glucose. Results from indirect calorimetry and hepatic and femoral venous<br />

catheterization. Diabetes. 1981;30(12):1000-7.<br />

45. Henriksen jE, Alford F, Ward GM, Beck-Nielsen H. Risk and mechanism of dexamethasone-induced<br />

deterioration of glucose tolerance in non-diabetic first-degree relatives of NIDDM patients.<br />

Diabetologia. 1997;40(12):1439-48.<br />

46. Larsson H, Ahren B. Insulin resistant subjects lack islet adaptation to short-term dexamethasoneinduced<br />

reduction in insulin sensitivity. Diabetologia. 1999;42(8):936-43.<br />

47. Ekstrand A, Schalin-jantti C, Lofman M, Parkkonen M, Widen E, Franssila-Kallunki A, Saloranta<br />

C, Koivisto V, Groop L. The effect of (steroid) immunosuppression on skeletal muscle glycogen<br />

metabolism in patients after kidney transplantation. Transplantation. 1996;61(6):889-93.<br />

48. Henriksen jE, Alford F, Vaag A, Handberg A, Beck-Nielsen H. Intracellular skeletal muscle glucose<br />

metabolism is differentially altered by dexamethasone treatment of normoglycemic relatives of type<br />

2 diabetic patients. Metabolism. 1999;48(9):1128-35.<br />

49. Ewart HS, Somwar R, Klip A. Dexamethasone stimulates the expression of GLUT1 and GLUT4<br />

proteins via different signalling pathways in L6 skeletal muscle cells. FEBS Lett. 1998;421(2):120-4.<br />

50. Long W, Barrett Ej, Wei L, Liu Z. Adrenalectomy enhances the insulin sensitivity of muscle protein<br />

synthesis. Am j Physiol Endocrinol Metab. 2003;284(1):E102-9.<br />

51. Ruzzin j, Wagman AS, jensen j. Glucocorticoid-induced insulin resistance in skeletal muscles: defects<br />

in insulin signalling and the effects of a selective glycogen synthase kinase-3 inhibitor. Diabetologia.<br />

2005;48(10):2119-30.<br />

52. Weinstein SP, Wilson CM, Pritsker A, Cushman SW. Dexamethasone inhibits insulin-stimulated<br />

recruitment of GLUT4 to the cell surface in rat skeletal muscle. Metabolism. 1998;47(1):3-6.<br />

53. van Raalte DH, Diamant M, Ouwens DM, Ijzerman RG, Linssen MML, Guigas B, Eringa EC, Serne EH.<br />

Glucocorticoid treatment impairs microvascular function in healthy males, in association with its<br />

adverse effects on blood pressure and glucose metabolism. Submitted. 2011.<br />

54. Biedasek K, Andres j, Mai K, Adams S, Spuler S, Fielitz j, Spranger j. Skeletal muscle 11beta-HSD1<br />

controls glucocorticoid-induced proteolysis and expression of E3 ubiquitin ligases atrogin-1 and<br />

MuRF-1. PLoS One. 2011;6(1):e16674.<br />

55. Schakman O, Gilson H, Kalista S, Thissen jP. Mechanisms of muscle atrophy induced by glucocorticoids.<br />

Horm Res. 2009;72 Suppl 1:36-41.<br />

56. Hu Z, Wang H, Lee IH, Du j, Mitch WE. Endogenous glucocorticoids and impaired insulin signaling<br />

are both required to stimulate muscle wasting under pathophysiological conditions in mice. j Clin<br />

Invest. 2009;119(10):3059-69.<br />

57. Short KR, Bigelow ML, Nair KS. Short-term prednisone use antagonizes insulin’s anabolic effect on<br />

muscle protein and glucose metabolism in young healthy people. Am j Physiol Endocrinol Metab.<br />

2009;297(6):E1260-8.<br />

317<br />

Chapter 15<br />

15


Chapter 15 General Discussion<br />

58. Lofberg E, Gutierrez A, Wernerman J, Anderstam B, Mitch WE, Price SR, Bergstrom J, Alvestrand<br />

A. Effects of high doses of glucocorticoids on free amino acids, ribosomes and protein turnover in<br />

human muscle. Eur j Clin Invest. 2002;32(5):345-53.<br />

59. Khaleeli AA, Edwards RH, Gohil K, McPhail G, Rennie Mj, Round j, Ross Ej. Corticosteroid myopathy:<br />

a clinical and pathological study. Clin Endocrinol (Oxf). 1983;18(2):155-66.<br />

60. Patti ME, Brambilla E, Luzi L, Landaker Ej, Kahn CR. Bidirectional modulation of insulin action by<br />

amino acids. j Clin Invest. 1998;101(7):1519-29.<br />

61. Krebs M, Krssak M, Bernroider E, Anderwald C, Brehm A, Meyerspeer M, Nowotny P, Roth E,<br />

Waldhausl W, Roden M. Mechanism of amino acid-induced skeletal muscle insulin resistance in<br />

humans. Diabetes. 2002;51(3):599-605.<br />

62. Schrauwen-Hinderling VB, Hesselink MK, Schrauwen P, Kooi ME. Intramyocellular lipid content in<br />

human skeletal muscle. Obesity (Silver Spring). 2006;14(3):357-67.<br />

63. Holland WL, Summers SA. Sphingolipids, insulin resistance, and metabolic disease: new insights<br />

from in vivo manipulation of sphingolipid metabolism. Endocr Rev. 2008;29(4):381-402.<br />

64. Bikman BT, Summers SA. Ceramides as modulators of cellular and whole-body metabolism. j Clin<br />

Invest. 2011;121(11):4222-30.<br />

65. Eckardt K, Taube A, Eckel j. Obesity-associated insulin resistance in skeletal muscle: role of lipid<br />

accumulation and physical inactivity. Rev Endocr Metab Disord. 2011;12(3):163-72.<br />

66. Holland WL, Knotts TA, Chavez jA, Wang LP, Hoehn KL, Summers SA. Lipid mediators of insulin<br />

resistance. Nutr Rev. 2007;65(6 Pt 2):S39-46.<br />

67. Holland WL, Brozinick jT, Wang LP, Hawkins ED, Sargent KM, Liu Y, Narra K, Hoehn KL, Knotts<br />

TA, Siesky A, Nelson DH, Karathanasis SK, Fontenot GK, Birnbaum Mj, Summers SA. Inhibition<br />

of ceramide synthesis ameliorates glucocorticoid-, saturated-fat-, and obesity-induced insulin<br />

resistance. Cell Metab. 2007;5(3):167-79.<br />

68. Schrauwen-Hinderling VB, Roden M, Kooi ME, Hesselink MK, Schrauwen P. Muscular mitochondrial<br />

dysfunction and type 2 diabetes mellitus. Curr Opin Clin Nutr Metab Care. 2007;10(6):698-703.<br />

69. Brands M, Hoeks j, Sauerwein HP, Ackermans MT, Ouwens M, Lammers NM, van der Plas MN,<br />

Schrauwen P, Groen AK, Serlie Mj. Short-term increase of plasma free fatty acids does not interfere<br />

with intrinsic mitochondrial function in healthy young men. Metabolism. 2011;60(10):1398-405.<br />

70. Parish R, Petersen KF. Mitochondrial dysfunction and type 2 diabetes. Curr Diab Rep. 2005;5(3):177-<br />

83.<br />

71. Viguerie N, Picard F, Hul G, Roussel B, Barbe P, Iacovoni jS, Valle C, Langin D, Saris WH. Multiple<br />

effects of a short-term dexamethasone treatment in human skeletal muscle and adipose tissue.<br />

Physiol Genomics. 2012;44(2):141-51.<br />

72. Hotamisligil GS. Endoplasmic reticulum stress and the inflammatory basis of metabolic disease. Cell.<br />

2010;140(6):900-17.<br />

318


73. Linssen MM, van Raalte DH, Toonen Ej, Alkema W, van der Zon GC, Dokter WH, Diamant M, Guigas B,<br />

Ouwens DM. Prednisolone-induced beta cell dysfunction is associated with impaired endoplasmic<br />

reticulum homeostasis in INS-1E cells. Cell Signal. 2011;23(11):1708-15.<br />

74. McMahon M, Gerich j, Rizza R. Effects of glucocorticoids on carbohydrate metabolism. Diabetes<br />

Metab Rev. 1988;4(1):17-30.<br />

75. Hamel FG, Fawcett j, Bennett RG, Duckworth WC. Control of proteolysis: hormones, nutrients, and<br />

the changing role of the proteasome. Curr Opin Clin Nutr Metab Care. 2004;7(3):255-8.<br />

76. Peckett Aj, Wright DC, Riddell MC. The effects of glucocorticoids on adipose tissue lipid metabolism.<br />

Metabolism. 2011;60(11):1500-10.<br />

77. Rockall AG, Sohaib SA, Evans D, Kaltsas G, Isidori AM, Monson jP, Besser GM, Grossman AB, Reznek<br />

RH. Computed tomography assessment of fat distribution in male and female patients with Cushing’s<br />

syndrome. Eur j Endocrinol. 2003;149(6):561-7.<br />

78. Rockall AG, Sohaib SA, Evans D, Kaltsas G, Isidori AM, Monson jP, Besser GM, Grossman AB, Reznek<br />

RH. Hepatic steatosis in Cushing’s syndrome: a radiological assessment using computed tomography.<br />

Eur j Endocrinol. 2003;149(6):543-8.<br />

79. Dallman MF, la Fleur SE, Pecoraro NC, Gomez F, Houshyar H, Akana SF. Minireview: glucocorticoids-<br />

-food intake, abdominal obesity, and wealthy nations in 2004. Endocrinology. 2004;145(6):2633-8.<br />

80. Fried SK, Russell CD, Grauso NL, Brolin RE. Lipoprotein lipase regulation by insulin and<br />

glucocorticoid in subcutaneous and omental adipose tissues of obese women and men. j Clin Invest.<br />

1993;92(5):2191-8.<br />

81. Ottosson M, Vikman-Adolfsson K, Enerback S, Olivecrona G, Bjorntorp P. The effects of cortisol<br />

on the regulation of lipoprotein lipase activity in human adipose tissue. j Clin Endocrinol Metab.<br />

1994;79(3):820-5.<br />

82. Hauner H, Entenmann G, Wabitsch M, Gaillard D, Ailhaud G, Negrel R, Pfeiffer EF. Promoting effect of<br />

glucocorticoids on the differentiation of human adipocyte precursor cells cultured in a chemically<br />

defined medium. J Clin Invest. 1989;84(5):1663-70.<br />

83. Macfarlane DP, Forbes S, Walker BR. Glucocorticoids and fatty acid metabolism in humans: fuelling<br />

fat redistribution in the metabolic syndrome. j Endocrinol. 2008;197(2):189-204.<br />

84. Ibrahim MM. Subcutaneous and visceral adipose tissue: structural and functional differences. Obes<br />

Rev. 2010;11(1):11-8.<br />

85. Turnbow MA, Keller SR, Rice KM, Garner CW. Dexamethasone down-regulation of insulin receptor<br />

substrate-1 in 3T3-L1 adipocytes. j Biol Chem. 1994;269(4):2516-20.<br />

86. Kanzaki M. Insulin receptor signals regulating GLUT4 translocation and actin dynamics. Endocr j.<br />

2006;53(3):267-93.<br />

87. Abel ED, Peroni O, Kim jK, Kim YB, Boss O, Hadro E, Minnemann T, Shulman GI, Kahn BB.<br />

Adipose-selective targeting of the GLUT4 gene impairs insulin action in muscle and liver. Nature.<br />

2001;409(6821):729-33.<br />

319<br />

Chapter 15<br />

15


Chapter 15 General Discussion<br />

88. Fasshauer M, Paschke R. Regulation of adipocytokines and insulin resistance. Diabetologia.<br />

2003;46(12):1594-603.<br />

89. Ouchi N, Parker JL, Lugus JJ, Walsh K. Adipokines in inflammation and metabolic disease. Nat Rev<br />

Immunol. 2011;11(2):85-97.<br />

90. Dimitriadis G, Mitrou P, Lambadiari V, Maratou E, Raptis SA. Insulin effects in muscle and adipose<br />

tissue. Diabetes Res Clin Pract. 2011;93 Suppl 1:S52-9.<br />

91. Xu C, He j, jiang H, Zu L, Zhai W, Pu S, Xu G. Direct effect of glucocorticoids on lipolysis in adipocytes.<br />

Mol Endocrinol. 2009;23(8):1161-70.<br />

92. Campbell JE, Peckett AJ, D’Souza A M, Hawke TJ, Riddell MC. Adipogenic and lipolytic effects of<br />

chronic glucocorticoid exposure. Am j Physiol Cell Physiol. 2011;300(1):C198-209.<br />

93. Lundgren M, Buren J, Lindgren P, Myrnas T, Ruge T, Eriksson JW. Sex- and depot-specific lipolysis<br />

regulation in human adipocytes: interplay between adrenergic stimulation and glucocorticoids.<br />

Horm Metab Res. 2008;40(12):854-60.<br />

94. Divertie GD, jensen MD, Miles jM. Stimulation of lipolysis in humans by physiological<br />

hypercortisolemia. Diabetes. 1991;40(10):1228-32.<br />

95. Samra jS, Clark ML, Humphreys SM, Macdonald IA, Matthews DR, Frayn KN. Effects of morning rise<br />

in cortisol concentration on regulation of lipolysis in subcutaneous adipose tissue. Am j Physiol.<br />

1996;271(6 Pt 1):E996-1002.<br />

96. Birkenhager JC, Timmermans HA, Lamberts SW. Depressed plasma FFA turnover rate in Cushing’s<br />

syndrome. j Clin Endocrinol Metab. 1976;42(1):28-32.<br />

97. Saunders J, Hall SE, Sonksen PH. Glucose and free fatty acid turnover in Cushing’s syndrome. J<br />

Endocrinol Invest. 1980;3(3):309-11.<br />

98. Boden G. Obesity, insulin resistance and free fatty acids. Curr Opin Endocrinol Diabetes Obes.<br />

2011;18(2):139-43.<br />

99. Kahn SE. The relative contributions of insulin resistance and beta-cell dysfunction to the<br />

pathophysiology of Type 2 diabetes. Diabetologia. 2003;46(1):3-19.<br />

100. Weyer C, Bogardus C, Mott DM, Pratley RE. The natural history of insulin secretory dysfunction and<br />

insulin resistance in the pathogenesis of type 2 diabetes mellitus. j Clin Invest. 1999;104(6):787-94.<br />

101. van Raalte DH, Diamant M. Glucolipotoxicity and beta cells in type 2 diabetes mellitus: Target for<br />

durable therapy? Diabetes Res Clin Pract. 2011;93 Suppl 1:S37-46.<br />

102. Matthews DR, Hosker jP, Rudenski AS, Naylor BA, Treacher DF, Turner RC. Homeostasis model<br />

assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin<br />

concentrations in man. Diabetologia. 1985;28(7):412-9.<br />

103. Ward WK, LaCava EC, Paquette TL, Beard jC, Wallum Bj, Porte D, jr. Disproportionate elevation of<br />

immunoreactive proinsulin in type 2 (non-insulin-dependent) diabetes mellitus and in experimental<br />

insulin resistance. Diabetologia. 1987;30(9):698-702.<br />

320


104. Mari A, Tura A, Gastaldelli A, Ferrannini E. Assessing insulin secretion by modeling in multiple-meal<br />

tests: role of potentiation. Diabetes. 2002;51 Suppl 1:S221-6.<br />

105. DeFronzo RA, Triplitt C, Qu Y, Lewis MS, Maggs D, Glass LC. Effects of exenatide plus rosiglitazone<br />

on beta-cell function and insulin sensitivity in subjects with type 2 diabetes on metformin. Diabetes<br />

Care. 2010;33(5):951-7.<br />

106. Taborsky Gj, jr., Smith PH, Halter jB, Porte D, jr. Glucose infusion potentiates the acute insulin response<br />

to nonglucose stimuli during the infusion of somatostatin. Endocrinology. 1979;105(5):1215-20.<br />

107. Zawalich WS, Tesz Gj, Yamazaki H, Zawalich KC, Philbrick W. Dexamethasone suppresses<br />

phospholipase C activation and insulin secretion from isolated rat islets. Metabolism. 2006;55(1):35-<br />

42.<br />

108. Hamamdzic D, Duzic E, Sherlock jD, Lanier SM. Regulation of alpha 2-adrenergic receptor expression<br />

and signaling in pancreatic beta-cells. Am j Physiol. 1995;269(1 Pt 1):E162-71.<br />

109. Angel I, Niddam R, Langer SZ. Involvement of alpha-2 adrenergic receptor subtypes in hyperglycemia.<br />

j <strong>Pharma</strong>col Exp Ther. 1990;254(3):877-82.<br />

110. jeong IK, Oh SH, Kim Bj, Chung jH, Min YK, Lee MS, Lee MK, Kim KW. The effects of dexamethasone<br />

on insulin release and biosynthesis are dependent on the dose and duration of treatment. Diabetes<br />

Res Clin Pract. 2001;51(3):163-71.<br />

111. Philippe j, Missotten M. Dexamethasone inhibits insulin biosynthesis by destabilizing insulin<br />

messenger ribonucleic acid in hamster insulinoma cells. Endocrinology. 1990;127(4):1640-5.<br />

112. Ranta F, Avram D, Berchtold S, Dufer M, Drews G, Lang F, Ullrich S. Dexamethasone induces cell death<br />

in insulin-secreting cells, an effect reversed by exendin-4. Diabetes. 2006;55(5):1380-90.<br />

113. Eizirik DL, Cardozo AK, Cnop M. The role for endoplasmic reticulum stress in diabetes mellitus.<br />

Endocr Rev. 2008;29(1):42-61.<br />

114. Shamoon H, Soman V, Sherwin RS. The influence of acute physiological increments of cortisol on<br />

fuel metabolism and insulin binding to monocytes in normal humans. j Clin Endocrinol Metab.<br />

1980;50(3):495-501.<br />

115. Kalhan SC, Adam PA. Inhibitory effect of prednisone on insulin secretion in man: model for<br />

duplication of blood glucose concentration. j Clin Endocrinol Metab. 1975;41(3):600-10.<br />

116. Beard jC, Halter jB, Best jD, Pfeifer MA, Porte D, jr. Dexamethasone-induced insulin resistance<br />

enhances B cell responsiveness to glucose level in normal men. Am j Physiol. 1984;247(5 Pt<br />

1):E592-6.<br />

117. Grill V, Pigon j, Hartling SG, Binder C, Efendic S. Effects of dexamethasone on glucose-induced insulin<br />

and proinsulin release in low and high insulin responders. Metabolism. 1990;39(3):251-8.<br />

118. Hansen KB, Vilsboll T, Bagger jI, Holst jj, Knop FK. Reduced glucose tolerance and insulin resistance<br />

induced by steroid treatment, relative physical inactivity, and high-calorie diet impairs the incretin<br />

effect in healthy subjects. j Clin Endocrinol Metab. 2010;95(7):3309-17.<br />

321<br />

Chapter 15<br />

15


Chapter 15 General Discussion<br />

119. Hansen KB, Vilsboll T, Bagger jI, Holst jj, Knop FK. Increased postprandial GIP and glucagon<br />

responses, but unaltered GLP-1 response after intervention with steroid hormone, relative physical<br />

inactivity, and high-calorie diet in healthy subjects. j Clin Endocrinol Metab. 2011;96(2):447-53.<br />

120. Matsumoto K, Yamasaki H, Akazawa S, Sakamaki H, Ishibashi M, Abiru N, Uotani S, Matsuo H,<br />

Yamaguchi Y, Tokuyama K, Nagataki S. High-dose but not low-dose dexamethasone impairs glucose<br />

tolerance by inducing compensatory failure of pancreatic beta-cells in normal men. j Clin Endocrinol<br />

Metab. 1996;81(7):2621-6.<br />

121. van Raalte DH, Kwa KAA, van Genugten RE, Tushuizen ME, Holst jj, Deacon CF, Karemaker jM, Heine<br />

Rj, Mari A, Diamant M. Islet-cel dysfunction by glucorticoid treatment: potential role for altered<br />

sympathovagal balance? Submitted. 2011.<br />

122. van Raalte DH, Nofrate V, Bunck MC, van Iersel T, Elassaiss Schaap j, Nassander UK, Heine Rj, Mari<br />

A, Dokter WH, Diamant M. Acute and 2-week exposure to prednisolone impair different aspects of<br />

beta-cell function in healthy men. Eur j Endocrinol. 2010;162(4):729-35.<br />

123. Besse C, Nicod N, Tappy L. Changes in insulin secretion and glucose metabolism induced by<br />

dexamethasone in lean and obese females. Obes Res. 2005;13(2):306-11.<br />

124. Bonuccelli S, Muscelli E, Gastaldelli A, Barsotti E, Astiarraga BD, Holst jj, Mari A, Ferrannini E.<br />

Improved tolerance to sequential glucose loading (Staub-Traugott effect): size and mechanisms. Am<br />

j Physiol Endocrinol Metab. 2009;297(2):E532-7.<br />

125. Lambillotte C, Gilon P, Henquin jC. Direct glucocorticoid inhibition of insulin secretion. An in vitro<br />

study of dexamethasone effects in mouse islets. j Clin Invest. 1997;99(3):414-23.<br />

126. Donath MY, Boni-Schnetzler M, Ellingsgaard H, Ehses JA. Islet inflammation impairs the pancreatic<br />

beta-cell in type 2 diabetes. Physiology (Bethesda). 2009;24:325-31.<br />

127. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444(7121):860-7.<br />

128. Derijk RH. Single nucleotide polymorphisms related to HPA axis reactivity. Neuroimmunomodulation.<br />

2009;16(5):340-52.<br />

129. van Rossum EF, Lamberts SW. Polymorphisms in the glucocorticoid receptor gene and their<br />

associations with metabolic parameters and body composition. Recent Prog Horm Res. 2004;59:333-<br />

57.<br />

130. Manenschijn L, van den Akker EL, Lamberts SW, van Rossum EF. Clinical features associated with<br />

glucocorticoid receptor polymorphisms. An overview. Ann N Y Acad Sci. 2009;1179:179-98.<br />

131. Davani B, Portwood N, Bryzgalova G, Reimer MK, Heiden T, Ostenson CG, Okret S, Ahren B, Efendic<br />

S, Khan A. Aged transgenic mice with increased glucocorticoid sensitivity in pancreatic beta-cells<br />

develop diabetes. Diabetes. 2004;53 Suppl 1:S51-9.<br />

132. van Raalte DH, van Leeuwen N, Simonis-Bik AMC, Nijpels G, van Haeften TW, Schafer SA, Boomsma<br />

DI, Kramer MHH, Heine Rj, Maassen jA, Staiger H, Machicao F, Haring HU, Slagboom PE, Willemsen G,<br />

de Geus Ej, Dekker jM, Fritsche A, Eekhoff EM, Diamant M, ‘t Hart LM. Glucocorticoid Receptor Gene<br />

Polymorphisms are Associated with Reduced First-phase Glucose-Stimulated Insulin Secretion and<br />

Disposition Index in Women, but not in Men. Submitted. 2011.<br />

322


133. Russcher H, Smit P, van den Akker EL, van Rossum EF, Brinkmann AO, de jong FH, Lamberts SW,<br />

Koper jW. Two polymorphisms in the glucocorticoid receptor gene directly affect glucocorticoidregulated<br />

gene expression. j Clin Endocrinol Metab. 2005;90(10):5804-10.<br />

134. Roussel R, Reis AF, Dubois-Laforgue D, Bellanne-Chantelot C, Timsit j, Velho G. The N363S<br />

polymorphism in the glucocorticoid receptor gene is associated with overweight in subjects with<br />

type 2 diabetes mellitus. Clin Endocrinol (Oxf). 2003;59(2):237-41.<br />

135. van Rossum EF, Koper jW, Huizenga NA, Uitterlinden AG, janssen jA, Brinkmann AO, Grobbee DE, de<br />

jong FH, van Duyn CM, Pols HA, Lamberts SW. A polymorphism in the glucocorticoid receptor gene,<br />

which decreases sensitivity to glucocorticoids in vivo, is associated with low insulin and cholesterol<br />

levels. Diabetes. 2002;51(10):3128-34.<br />

136. van Rossum EF, Voorhoeve PG, te Velde Sj, Koper jW, Delemarre-van de Waal HA, Kemper HC,<br />

Lamberts SW. The ER22/23EK polymorphism in the glucocorticoid receptor gene is associated<br />

with a beneficial body composition and muscle strength in young adults. J Clin Endocrinol Metab.<br />

2004;89(8):4004-9.<br />

137. Koeijvoets KC, van Rossum EF, Dallinga-Thie GM, Steyerberg EW, Defesche jC, Kastelein jj,<br />

Lamberts SW, Sijbrands Ej. A functional polymorphism in the glucocorticoid receptor gene and its<br />

relation to cardiovascular disease risk in familial hypercholesterolemia. j Clin Endocrinol Metab.<br />

2006;91(10):4131-6.<br />

138. Kuningas M, Mooijaart SP, Slagboom PE, Westendorp RG, van Heemst D. Genetic variants in the<br />

glucocorticoid receptor gene (NR3C1) and cardiovascular disease risk. The Leiden 85-plus Study.<br />

Biogerontology. 2006;7(4):231-8.<br />

139. Gromada j, Franklin I, Wollheim CB. Alpha-cells of the endocrine pancreas: 35 years of research but<br />

the enigma remains. Endocr Rev. 2007;28(1):84-116.<br />

140. Dunning BE, Gerich jE. The role of alpha-cell dysregulation in fasting and postprandial hyperglycemia<br />

in type 2 diabetes and therapeutic implications. Endocr Rev. 2007;28(3):253-83.<br />

141. D’Alessio D. The role of dysregulated glucagon secretion in type 2 diabetes. Diabetes Obes Metab.<br />

2011;13 Suppl 1:126-32.<br />

142. Wise JK, Hendler R, Felig P. Influence of glucocorticoids on glucagon secretion and plasma amino<br />

acid concentrations in man. j Clin Invest. 1973;52(11):2774-82.<br />

143. van Raalte DH, van Genugten RE, Linssen MM, Ouwens DM, Diamant M. Glucagon-like peptide-1<br />

receptor agonist treatment prevents glucocorticoid-induced glucose intolerance and islet-cell<br />

dysfunction in humans. Diabetes Care. 2011;34(2):412-7.<br />

144. Drucker Dj. Biological actions and therapeutic potential of the glucagon-like peptides.<br />

Gastroenterology. 2002;122(2):531-44.<br />

145. Salehi F, Turner MC, Phillips KP, Wigle DT, Krewski D, Aronson Kj. Review of the etiology of breast<br />

cancer with special attention to organochlorines as potential endocrine disruptors. j Toxicol Environ<br />

Health B Crit Rev. 2008;11(3-4):276-300.<br />

323<br />

Chapter 15<br />

15


Chapter 15 General Discussion<br />

146. Vilsboll T. The effects of glucagon-like peptide-1 on the beta cell. Diabetes Obes Metab. 2009;11<br />

Suppl 3:11-8.<br />

147. Cunha DA, Ladriere L, Ortis F, Igoillo-Esteve M, Gurzov EN, Lupi R, Marchetti P, Eizirik DL, Cnop M.<br />

Glucagon-like peptide-1 agonists protect pancreatic beta-cells from lipotoxic endoplasmic reticulum<br />

stress through upregulation of BiP and junB. Diabetes. 2009;58(12):2851-62.<br />

148. Nauck M, Stockmann F, Ebert R, Creutzfeldt W. Reduced incretin effect in type 2 (non-insulindependent)<br />

diabetes. Diabetologia. 1986;29(1):46-52.<br />

149. Meier jj, Nauck MA. Is the diminished incretin effect in type 2 diabetes just an epi-phenomenon of<br />

impaired beta-cell function? Diabetes. 2010;59(5):1117-25.<br />

150. Zhang R, Packard BA, Tauchi M, D’Alessio DA, Herman JP. Glucocorticoid regulation of preproglucagon<br />

transcription and RNA stability during stress. Proc Natl Acad Sci U S A. 2009;106(14):5913-8.<br />

151. Hansen KB, Vilsboll T, Bagger JI, Holst JJ, Knop FK. Impaired Incretin-Induced Amplification of<br />

Insulin Secretion after Glucose Homeostatic Dysregulation in Healthy Subjects. j Clin Endocrinol<br />

Metab. 2012.<br />

152. Muniyappa R, Sorkin jD, Veldhuis jD, Harman SM, Munzer T, Bhasin S, Blackman MR. Long-term<br />

testosterone supplementation augments overnight growth hormone secretion in healthy older men.<br />

Am j Physiol Endocrinol Metab. 2007;293(3):E769-75.<br />

153. Barrett Ej, Eggleston EM, Inyard AC, Wang H, Li G, Chai W, Liu Z. The vascular actions of insulin<br />

control its delivery to muscle and regulate the rate-limiting step in skeletal muscle insulin action.<br />

Diabetologia. 2009;52(5):752-64.<br />

154. Kubota T, Kubota N, Kumagai H, Yamaguchi S, Kozono H, Takahashi T, Inoue M, Itoh S, Takamoto<br />

I, Sasako T, Kumagai K, Kawai T, Hashimoto S, Kobayashi T, Sato M, Tokuyama K, Nishimura S,<br />

Tsunoda M, Ide T, Murakami K, Yamazaki T, Ezaki O, Kawamura K, Masuda H, Moroi M, Sugi K, Oike<br />

Y, Shimokawa H, Yanagihara N, Tsutsui M, Terauchi Y, Tobe K, Nagai R, Kamata K, Inoue K, Kodama T,<br />

Ueki K, Kadowaki T. Impaired insulin signaling in endothelial cells reduces insulin-induced glucose<br />

uptake by skeletal muscle. Cell Metab. 2011;13(3):294-307.<br />

155. Serne EH, de jongh RT, Eringa EC, RG Ij, Stehouwer CD. Microvascular dysfunction: a potential<br />

pathophysiological role in the metabolic syndrome. Hypertension. 2007;50(1):204-11.<br />

156. Kim jA, Montagnani M, Koh KK, Quon Mj. Reciprocal relationships between insulin resistance<br />

and endothelial dysfunction: molecular and pathophysiological mechanisms. Circulation.<br />

2006;113(15):1888-904.<br />

157. Li R, Zhang H, Wang W, Wang X, Huang Y, Huang C, Gao F. Vascular insulin resistance in prehypertensive<br />

rats: role of PI3-kinase/Akt/eNOS signaling. Eur j <strong>Pharma</strong>col. 2010;628(1-3):140-7.<br />

158. Schafer SC, Wallerath T, Closs EI, Schmidt C, Schwarz PM, Forstermann U, Lehr HA. Dexamethasone<br />

suppresses eNOS and CAT-1 and induces oxidative stress in mouse resistance arterioles. Am j<br />

Physiol Heart Circ Physiol. 2005;288(1):H436-44.<br />

159. Aronsson M, Fuxe K, Dong Y, Agnati LF, Okret S, Gustafsson jA. Localization of glucocorticoid receptor<br />

mRNA in the male rat brain by in situ hybridization. Proc Natl Acad Sci U S A. 1988;85(23):9331-5.<br />

324


160. Zakrzewska KE, Cusin I, Stricker-Krongrad A, Boss O, Ricquier D, jeanrenaud B, Rohner-jeanrenaud<br />

F. Induction of obesity and hyperleptinemia by central glucocorticoid infusion in the rat. Diabetes.<br />

1999;48(2):365-70.<br />

161. Cusin I, Rouru j, Rohner-jeanrenaud F. Intracerebroventricular glucocorticoid infusion in normal rats:<br />

induction of parasympathetic-mediated obesity and insulin resistance. Obes Res. 2001;9(7):401-6.<br />

162. Yi CX, Foppen E, Abplanalp W, Gao Y, Alkemade A, la Fleur SE, Serlie Mj, Fliers E, Buijs RM, Tschop MH,<br />

Kalsbeek A. Glucocorticoid signaling in the arcuate nucleus modulates hepatic insulin sensitivity.<br />

Diabetes. 2012;61(2):339-45.<br />

163. Schmidt C, Hocherl K, Bucher M. Regulation of renal glucose transporters during severe inflammation.<br />

Am j Physiol Renal Physiol. 2007;292(2):F804-11.<br />

164. Qi D, Rodrigues B. Glucocorticoids produce whole body insulin resistance with changes in cardiac<br />

metabolism. Am j Physiol Endocrinol Metab. 2007;292(3):E654-67.<br />

165. Walker BR. Glucocorticoids and cardiovascular disease. Eur j Endocrinol. 2007;157(5):545-59.<br />

166. Wasko MC, Kay j, Hsia EC, Rahman MU. Diabetes and insulin resistance in patients with rheumatoid<br />

arthritis: Risk reduction in a chronic inflammatory disease. Arthritis Care Res (Hoboken). 2010.<br />

167. Chung CP, Oeser A, Solus jF, Gebretsadik T, Shintani A, Avalos I, Sokka T, Raggi P, Pincus T, Stein CM.<br />

Inflammation-associated insulin resistance: differential effects in rheumatoid arthritis and systemic<br />

lupus erythematosus define potential mechanisms. Arthritis Rheum. 2008;58(7):2105-12.<br />

168. Shahin D, Eltoraby E, Mesbah A, Houssen M. Insulin resistance in early untreated rheumatoid<br />

arthritis patients. Clin Biochem. 2010;43(7-8):661-5.<br />

169. Dessein PH, joffe BI. Insulin resistance and impaired beta cell function in rheumatoid arthritis.<br />

Arthritis Rheum. 2006;54(9):2765-75.<br />

170. den Uyl D, van Raalte DH, Nurmohamed MT, Lems WF, Bijlsma jW, Hoes jN, Dijkmans BA, Diamant M.<br />

Metabolic effects of high-dose prednisolone treatment in early rheumatoid arthritis: Diabetogenic<br />

effects and inflammation reduction on the balance. Arthritis Rheum. 2011.<br />

171. Hoes jN, van der Goes MC, van Raalte DH, van der Zijl Nj, den Uyl D, Lems WF, Lafeber FP, jacobs jW,<br />

Welsing PM, Diamant M, Bijlsma jW. Glucose tolerance, insulin sensitivity and beta-cell function in<br />

patients with rheumatoid arthritis treated with or without low-to-medium dose glucocorticoids.<br />

Ann Rheum Dis. 2011;70(11):1887-94.<br />

172. Svenson KL, Lundqvist G, Wide L, Hallgren R. Impaired glucose handling in active rheumatoid<br />

arthritis: effects of corticosteroids and antirheumatic treatment. Metabolism. 1987;36(10):944-8.<br />

173. Hallgren R, Berne C. Glucose intolerance in patients with chronic inflammatory diseases is<br />

normalized by glucocorticoids. Acta Med Scand. 1983;213(5):351-5.<br />

174. Dessein PH, joffe BI, Stanwix AE, Christian BF, Veller M. Glucocorticoids and insulin sensitivity in<br />

rheumatoid arthritis. j Rheumatol. 2004;31(5):867-74.<br />

325<br />

Chapter 15<br />

15


Chapter 15 General Discussion<br />

175. van Tuyl LH, Boers M, Lems WF, Landewe RB, Han H, van der Linden S, van de Laar M, Westhovens<br />

R, van Denderen jC, Westedt ML, Peeters Aj, jacobs P, Huizinga TW, van de Brink H, Dijkmans BA,<br />

Voskuyl AE. Survival, comorbidities and joint damage 11 years after the COBRA combination therapy<br />

trial in early rheumatoid arthritis. Ann Rheum Dis. 2010;69(5):807-12.<br />

176. Wolfe F, Michaud K. Corticosteroids increase the risk of diabetes mellitus in rheumatoid arthritis<br />

and contribute to the risk of myocardial infarction and heart failure (abstract). Ann Rheum Dis.<br />

2004;63(Suppl1):495.<br />

177. Arnaldi G, Angeli A, Atkinson AB, Bertagna X, Cavagnini F, Chrousos GP, Fava GA, Findling jW, Gaillard<br />

RC, Grossman AB, Kola B, Lacroix A, Mancini T, Mantero F, Newell-Price j, Nieman LK, Sonino N,<br />

Vance ML, Giustina A, Boscaro M. Diagnosis and complications of Cushing’s syndrome: a consensus<br />

statement. j Clin Endocrinol Metab. 2003;88(12):5593-602.<br />

178. Yuen KC, McDaniel PA, Riddle MC. Twenty-four hour profiles of plasma glucose, insulin, C-peptide<br />

and free fatty acid in subjects with varying degrees of glucose tolerance following short-term<br />

medium-dose prednisone (20 mg/day) treatment: evidence for differing effects on insulin secretion<br />

and action. Clin Endocrinol (Oxf). 2011.<br />

179. Burt MG, Roberts GW, Aguilar-Loza NR, Frith P, Stranks SN. Continuous monitoring of circadian<br />

glycemic patterns in patients receiving prednisolone for COPD. j Clin Endocrinol Metab.<br />

2011;96(6):1789-96.<br />

180. Bunck MC, Diamant M, Corner A, Eliasson B, Malloy jL, Shaginian RM, Deng W, Kendall DM,<br />

Taskinen MR, Smith U, Yki-jarvinen H, Heine Rj. One-year treatment with exenatide improves<br />

beta-cell function, compared with insulin glargine, in metformin-treated type 2 diabetic patients: a<br />

randomized, controlled trial. Diabetes Care. 2009;32(5):762-8.<br />

181. van Genugten RE, van Raalte DH, Diamant M. Does glucagon-like peptide-1 receptor agonist therapy<br />

add value in the treatment of type 2 diabetes? Focus on exenatide. Diabetes Res Clin Pract. 2009;86<br />

Suppl 1:S26-34.<br />

182. Garber AJ. Long-acting glucagon-like peptide 1 receptor agonists: a review of their efficacy and<br />

tolerability. Diabetes Care. 2011;34 Suppl 2:S279-84.<br />

183. Cervera A, Wajcberg E, Sriwijitkamol A, Fernandez M, Zuo P, Triplitt C, Musi N, DeFronzo RA,<br />

Cersosimo E. Mechanism of action of exenatide to reduce postprandial hyperglycemia in type 2<br />

diabetes. Am j Physiol Endocrinol Metab. 2008;294(5):E846-52.<br />

184. Zheng D, Ionut V, Mooradian V, Stefanovski D, Bergman RN. Exenatide sensitizes insulin-mediated<br />

whole-body glucose disposal and promotes uptake of exogenous glucose by the liver. Diabetes.<br />

2009;58(2):352-9.<br />

185. Baggio LL, Drucker Dj. Biology of incretins: GLP-1 and GIP. Gastroenterology. 2007;132(6):2131-57.<br />

186. van Genugten RE, van Raalte DH, Diamant M. Dipeptidyl peptidase-4 inhibitors and preservation<br />

of pancreatic islet-cell function: a critical appraisal of the evidence. Diabetes Obes Metab.<br />

2012;14(2):101-11.<br />

326


187. Bunck MC, Diamant M, Eliasson B, Corner A, Shaginian RM, Heine Rj, Taskinen MR, Yki-jarvinen H,<br />

Smith U. Exenatide affects circulating cardiovascular risk biomarkers independently of changes in<br />

body composition. Diabetes Care. 2010;33(8):1734-7.<br />

188. Tushuizen ME, Bunck MC, Pouwels Pj, van Waesberghe jH, Diamant M, Heine Rj. Incretin mimetics<br />

as a novel therapeutic option for hepatic steatosis. Liver Int. 2006;26(8):1015-7.<br />

189. Bunck MC, Corner A, Eliasson B, Heine Rj, Shaginian RM, Wu Y, Yan P, Smith U, Yki-jarvinen H, Diamant<br />

M, Taskinen MR. One-year treatment with exenatide vs. insulin glargine: effects on postprandial<br />

glycemia, lipid profiles, and oxidative stress. Atherosclerosis. 2010;212(1):223-9.<br />

190. http://clinicaltrials.gov/ct2/show/NCT00721552?term=SPHINX&rank=1.<br />

327<br />

Chapter 15<br />

15


Nederlandse<br />

Samenvatting<br />

Diabetogene effecten van glucocorticoïde<br />

medicatie: bekende en onbekende aspecten<br />

Dutch summary


Nederlandse samenvatting<br />

ontdekking van het bijnierschorshormoon cortisol<br />

Cortisol is een hormoon dat in ons lichaam wordt geproduceerd in de schors (ook wel cortex<br />

genoemd) van de bijnieren. De bijnieren zijn organen die, zoals de naam al aangeeft, in de<br />

buurt van de nieren liggen. Cortisol wordt vanwege zijn structuur een steroïd hormoon<br />

genoemd (er zijn nog vele andere steroïde hormonen in ons lichaam), en omdat snel duidelijk<br />

werd dat cortisol een belangrijke rol speelt in de glucose stofwisseling werd het geclassifeerd<br />

als glucocorticosteroïd hormoon. Dit werd later in de naamgeving verkort tot glucocorticoïd<br />

hormoon.<br />

Al begin 20ste eeuw werd duidelijk dat dieren en mensen die geen of onvoldoende actieve<br />

bijnieren hebben verschillende problemen krijgen, zoals te lage bloedsuikers, te lage<br />

bloeddruk en klachten van algehele zwakte, tezamen ook wel de ziekte van Addison genoemd.<br />

Uit dierproeven uitgevoerd rond 1930 werd tevens duidelijk dat stoffen uit de bijnieren een<br />

belangrijke rol spelen bij stressreacties van het lichaam. Het was toen echter nog onduidelijk<br />

welke stof hiervoor precies verantwoordelijk was. Hierin kwam verandering toen de inactieve<br />

vorm van cortisol (cortison) in 1936 voor het eerst werd geïsoleerd uit de bijnieren van<br />

runderen door Poolse chemicus Tadeusz Reichstein en door de Amerikaan wetenschapper<br />

Edward Calvin Kendall. Hierdoor werd het mogelijk de effecten van cortison nauwkeurig te<br />

bestuderen (het menselijk lichaam zet cortison om in cortisol, waardoor het actief wordt).<br />

Cortisol zelf werd een tiental jaren later ook geïsoleerd.<br />

ontwikkeling van synthetische bijnierschorshormonen tot medicijnen<br />

Hoewel de isolatie van cortison een grote stap vooruit was om de werking van het hormoon<br />

precies te kunnen onderzoeken, werd meteen een grote beperking duidelijk: de hoeveelheid<br />

die geïsoleerd kon worden uit runderen was zeer beperkt. Daarom werd gezocht naar een<br />

methode om cortison synthetisch te produceren. Dit proces werd versneld door Amerikaanse<br />

deelname aan de tweede wereldoorlog. Er gingen hardnekkige, maar nooit bewezen geruchten<br />

dat Duitse Luftwaffe piloten bijnierextracten gebruikten om zo beter bestand te zijn tegen<br />

zuurstof tekort ten gevolge van vliegen op grote hoogte. Ondanks enorme inspanningen werd<br />

het eerste synthetische cortison vlak na afloop van de oorlog geproduceerd.<br />

In 1948 konden met het nieuw geproduceerd cortison de eerste studies in mensen uitgevoerd<br />

worden. Reumatoloog Philip Showalter Hench (een collega van Kendall) toonde aan dat<br />

cortison inderdaad het bijnierschors hormoon was dat de ziekte van Addison kon verhelpen<br />

en daarmee werd de kritieke rol van cortisol in de regulatie van bloeddruk en glucose<br />

stofwisseling bevestigd. Maar cortison behandeling bleek meer gunstige effecten te hebben:<br />

het gaf een dramatische afname van gewrichtsklachten in patiënten met reumatoïde artritis en<br />

330


het bleek ook heel effectief bij de behandeling van andere auto-immuunziekten (aandoeningen<br />

waarbij het lichaam eigen weefsels aanvalt en doet ontsteken). Het wondermiddel cortison<br />

werd in 1950 officieel gelanceerd als medicijn en in hetzelfde jaar kregen Reichstein, Kendall<br />

en Hench de Nobelprijs voor de Geneeskunde voor hun onderzoek naar de structuur en<br />

werking van bijnierschorshormonen.<br />

Glucocorticoïden: een zorgwekkend bijwerking profiel<br />

Na een eerste periode van enorm enthousiasme werd helaas snel duidelijk dat cortison<br />

therapie ging met forse bijwerkingen. Hiertoe behoorden onder andere de ontwikkeling<br />

van overgewicht, suikerziekte, hoge bloeddruk, versnelde bot- en spierafbraak, verhoogde<br />

kans op infecties, maagzweren, depressie of psychose, huidafwijkingen, verhoogde<br />

kans op hart- en vaatziekten en staar. En deze lijst is zeker nog niet compleet. Met als<br />

doel het bijwerkingenprofiel te verbeteren werden nieuwe synthetische glucocorticoïd<br />

hormonen gemaakt, zoals prednisolon (=prednison) (1965) en dexamethason (1966).<br />

Hoewel deze middelen minder bijwerkingen vertoonden ten opzichte van cortison, bleven<br />

teleurstellenderwijs de meesten hiervan bestaan, zeker in hoge dosering.<br />

Dit heeft hedendaags nog steeds consequenties voor het gebruik van glucocorticoïden bij<br />

patiënten. je bent als arts door de bijwerkingen gedwongen snel de dosis te verlagen wat<br />

kan leiden tot onderbehandeling van de onderliggende ziekte. Verder hebben patiënten vaak<br />

weerstand prednisolon te gebruiken vanwege zijn beruchte bijwerkingen (het wordt vaak<br />

‘een paardenmiddel’ genoemd). Tenslotte moeten artsen vaak extra medicatie starten om<br />

bijwerkingen te voorkomen of te bestrijden, zoals middelen die botontkalking remmen en het<br />

maagslijmvlies beschermen.<br />

Nadelige effecten van glucocorticoïden op de glucose stofwisseling<br />

Al snel na de introductie werd bekend dat behandeling met glucocorticoïden belangrijke<br />

nadelige effecten heeft op de glucose stofwisseling. Schattingen zijn dat bij 30% van de<br />

patiënten die langdurig glucocorticoïden gebruiken suikerziekte optreedt. Ondanks dat deze<br />

bijwerkingen al relatief lang bekend zijn, zijn de mechanismen hierachter minder duidelijk.<br />

In de jaren ‘60-‘70 werd bekend dat glucocorticoïden de werking van het hormoon insuline<br />

verminderen (‘insuline resistentie’). Ook zijn er op dit moment geen duidelijke richtlijnen hoe<br />

suikerziekte veroorzaakt door glucocorticoïden het best kan worden behandeld of kan worden<br />

voorkomen. Na de jaren ‘70 raakte het onderzoek naar de bijwerkingen van glucocorticoïden<br />

op de stofwisseling wat uit beeld.<br />

331<br />

Nederlandse samenvatting


Nederlandse samenvatting<br />

Hierin kwam de laatste 10 jaar verandering door de ontwikkeling van nieuwe glucocorticoïd<br />

medicijnen, die wel krachtig ontsteking kunnen remmen, maar waarschijnlijk minder<br />

bijwerkingen hebben op de stofwisseling. Deze medicijnen in ontwikkeling worden<br />

zogenaamde “gedissocieerde glucocorticoïd receptor agonisten” genoemd. Om de<br />

ontwikkeling van deze nieuwe medicatie verder mogelijk te maken is echter meer kennis<br />

nodig van de bijwerkingen van de huidige glucocorticoïden in mensen, zodat in een latere<br />

fase de middelen met elkaar vergeleken kunnen worden in studies. Daarom hebben we in<br />

mijn proefschrift de metabole bijwerkingen van glucocorticoïden bestudeerd en biomarkers<br />

geprobeerd te vinden die het bijwerkingenprofiel zo goed mogelijk weergeven. We hebben<br />

de effecten van het glucocorticoïd prednisolon bestudeerd, omdat dit wereldwijd het meest<br />

wordt voorgeschreven en gekozen voor een hoge (inductie) dosis (30 mg per dag) en een<br />

zogenaamde onderhoudsdosering (7,5 mg per dag).<br />

In het eerste deel van mijn proefschrift beschrijven wij de introductie van glucocorticoïden<br />

als medicatie in de jaren ’50 (hoofdstuk 1) en de stand van zaken omtrent de kennis van<br />

metabole bijwerkingen van glucocorticoïden toen ons onderzoek startte (hoofdstuk 2).<br />

Hierin wordt duidelijk dat er in de vroegere artikelen veel nadruk werd gelegd op het ontstaan<br />

van insuline resistentie in spier, lever en vetweefsel door glucocorticoïd behandeling.<br />

In deel 2 van mijn proefschrift laten we zien dat verstoring van de alvleesklierfunctie een<br />

belangrijke rol speelt bij het ontstaan van de glucocorticoïd-geïnduceerde bijwerkingen op<br />

de stofwisseling.<br />

In hoofdstuk 3 tonen we dat de bèta cellen in de eilandjes van Langerhans onvoldoende<br />

insuline kunnen produceren doordat de functie van het endoplasmatisch reticulum, de<br />

eiwitfabriek van de cellen, door glucocorticoïd behandeling wordt verstoord. Ook vonden<br />

we dat behandeling van bèta cellen met prednisolon leidt tot celdood, wat de functie van de<br />

eilandjes van Langerhans verder belemmert.<br />

In hoofdstuk 4 worden de effecten van prednisolon op bèta-cel functie in gezonde mannen<br />

beschreven. We vonden voor het eerst dat bèta-cel functie specifiek na het eten van een<br />

maaltijd verstoord is, zowel na acute als meer langdurige prednisolon therapie. Hiervoor<br />

maakten we gebruik van mathematische modeling analyse van glucose en C-peptide curves<br />

na de maaltijd voor en na prednisolon behandeling.<br />

In hoofdstuk 5 hebben we dit concept verder onderzocht. We vonden opnieuw dat bèta-<br />

cel functie na de maaltijd verstoord wordt, maar ook dat de maximale secretoire capaciteit<br />

van de bèta cel, gemeten tijdens intraveneuze glucose en arginine toediening, verlaagd wordt<br />

332


door prednisolon behandeling. In dit hoofdstuk beschrijven we tevens dat de functie van<br />

de alpha cellen, die eveneens gelegen zijn binnen de eilandjes van Langerhans, ook door<br />

prednisolon behandeling wordt verstoord. Alpha cellen scheiden namelijk te veel van het<br />

hormoon glucagon uit, met name tijdens de maaltijden, hetgeen leidt tot overmatige glucose<br />

productie door de lever. Zowel de negatieve veranderingen in alpha- als bèta-cel functie<br />

leken gerelateerd aan ongunstige wijzigingen in de balans tussen het parasympathische en<br />

sympathische gedeelte van het autonome zenuwstelsel, die insuline en glucagon secretie in<br />

de eilandjes reguleert.<br />

In hoofdstuk 6 beschrijven we dat mensen die genetische veranderingen in het DNA hebben<br />

dat de glucocorticoïd receptor codeert, verminderde hoeveelheid insuline uitscheiden bij<br />

een glucose stimulus (dus verminderde functie van de bèta cel hebben). Hoewel dit een<br />

associatieve studie betreft in tegenstelling tot de eerdere interventie studies, benadrukt<br />

dit hoofdstuk onze mening dat verstoorde alvleesklierfunctie, gekenmerkt door relatief te<br />

weinig insuline productie, een cruciale rol speelt bij het ontstaan van hoge bloedsuikers door<br />

prednisolon behandeling.<br />

Door het toenemende inzicht in de verstoring van de alvleesklierfunctie door prednisolon<br />

hebben we een additionele studie geïniteerd om te kijken of we de negatieve effecten van<br />

prednisolon konden voorkomen door gelijktijdige behandeling met de glucagon-achtige<br />

peptide-1 (GLP-1) receptor agonist exenatide. Dit middel is onlangs geregistreerd voor de<br />

behandeling van suikerziekte omdat het de productie van insuline stimuleert en de aanmaak<br />

van glucagon remt, met name in de periode na de maaltijden. Behandeling met exenatide<br />

voorkwam inderdaad de effecten van prednisolon op de suikerstofwisseling door glucagon<br />

productie te remmen en insuline productie te stimuleren; dit hebben wij beschreven in het<br />

laatste hoofdstuk van deel 2, te weten hoofdstuk 7. Omdat thans geen goede richtlijnen<br />

bestaan over welke middelen het beste gebruikt kunnen worden om suikerziekte veroorzaakt<br />

door glucocorticoïd therapie te behandelen, postuleren wij dat GLP-1 receptor agonisten zoals<br />

exenatide een geschikte kandidaat zouden kunnen zijn. Momenteel worden de beschermende<br />

effecten van zogenaamde DPP-4 remmers, medicatie die het GLP-1 in het lichaam verhoogt,<br />

op de suikerstofwisseling onderzocht tijdens prednisolon gebruik.<br />

Insuline resistentie op het niveau van de lever, skeletspier en vetweefsel is een belangrijke<br />

oorzaak voor de nadelige effecten van glucocorticoïden op de stofwisseling, zoals reeds<br />

decennia bekend is. Echter, de wijze waarop dit gebeurt is nog steeds voor een groot deel<br />

onduidelijk. In deel 3 hebben we mechanistische studies ondernomen om nader te kijken<br />

naar onderliggende oorzaken.<br />

333<br />

Nederlandse samenvatting


Nederlandse samenvatting<br />

In hoofdstuk 8, bevestigen we dat hoge dosering insuline resistentie geeft in de skeletspier,<br />

lever en vetweefsel, echter wij tonen aan dat, met name gedurende hogere insuline<br />

concentraties in het bloed, een lage dosering prednisolon, tot voor kort veilig geacht, ook de<br />

insuline gevoeligheid vermindert van het vetweefsel en de lever, wat op langere termijn zou<br />

kunnen leiden tot verstoring van bloed glucose waarden en vetten in het bloed.<br />

In hoofdstuk 9 zoeken we naar de onderliggende mechanismen van prednisolon<br />

geïnduceerde insuline resistentie in de skeletspier. We hypothetiseerden dat prednisolon<br />

schadelijke vetten in spier zou doen ophopen (zoals ceramides), echter dit bleek niet het<br />

geval. Ook veronderstelden we dat mitochondriën, de energiecentrale van de cel, minder<br />

goed functioneerden met als gevolg minder glucose opname in de skeletspier, maar de<br />

mitochondriële functie bleef intact.<br />

Een “orgaan” waarvan wij voor het eerst vonden dat het een rol speelt bij de metabole<br />

bijwerkingen van prednisolon is de microcirculatie, de kleinste vaatjes in ons lichaam<br />

waar energie uitwisseling plaats vindt. Tijdens hoge insuline concentraties in het bloed,<br />

het belangrijkste moment van glucose opname door het lichaam, verstoort prednisolon de<br />

functie van de kleinste bloedvaten leidend tot minder glucose opname (hoofdstuk 10). Dus<br />

concludeerden wij dat prednisolon behandeling de kleinste bloedvaatjes resistent maakt<br />

voor de effecten van insuline, leidend tot minder glucose uptake, met name in de skeletspier.<br />

Ook de stijging in bloeddruk die we vonden na prednisolon behandeling was gecorreleerd<br />

met afwijkende werking van de kleinste bloedvaatjes waarvan bekend is dat ze de bloeddruk<br />

reguleren door de vaatweerstand te bepalen.<br />

In hoofdstuk 11 hebben we naar de effecten van prednisolon op het subcutane vetweefsel<br />

gekeken. We vonden voor het eerst in mensen dat er een afgenomen activatie is tijdens<br />

insuline toediening van belangrijke eiwitten betrokken bij glucose opname in het vetweefsel.<br />

Ook scheidt het vetweefsel ongunstige hormonen uit die elders in het lichaam de stofwisseling<br />

kunnen ontregelen.<br />

In het laatste hoofdstuk van deel 3, hoofdstuk 12, hebben we specifiek gegeken naar het eiwit<br />

Angiopoietine-like protein 4, kortweg Angptl4. Angptl4 is op belangrijke wijze betrokken in<br />

onze vetstofwisseling. Interessant genoeg werd onlangs aangetoond dat muizen die geen<br />

Angptl4 hebben, minder bijwerkingen ondervonden van prednisolon behandeling. Daarom<br />

besloten we dit eiwit in onze deelnemers te meten voor en na behandeling. Hoewel de<br />

vetstofwisseling bij iedere deelnemer ongunstig werd veranderd door prednisolon, het eiwit<br />

Angptl4 leek hier geen rol van belang bij te spelen. Wel verstoort prednisolon de regulatie van<br />

Angptl4 door insuline.<br />

334


In deel 4 van mijn proefschrift beschrijven we de effecten van prednisolon bij patiënten met<br />

reumatoide artritis (RA). Dit hebben wij gedaan omdat RA patienten vaak glucocorticoïden<br />

krijgen voor hun ziekte en daarmee vormen ze dus een zeer relevante onderzoekspopulatie.<br />

Verder biedt het de mogelijkheid om de effecten van glucocorticoïden op de stofwisseling<br />

in mensen met systemische ontsteking te meten. Systemische ontsteking zelf namelijk is<br />

ook ongunstig voor de stofwisseling omdat het insuline resistentie geeft en ook de werking<br />

van de bèta cel vermindert, daarom zouden glucocorticoïden zelfs een gunstig effect op de<br />

stofwisseling kunnen hebben door inflammatie te verminderen. Bij patiënten die onlangs<br />

met RA zijn gediagnosticeerd en hoge ontstekingswaarden in het bloed hebben, bleken<br />

de negatieve effecten van glucocorticoïden op de suikerstofwisseling inderdaad minder<br />

uitgesproken, waarschijnlijk door vermindering van systemische ontsteking. Dit laten we<br />

zien in hoofdstuk 13. In mensen met langer bestaande RA is glucocorticoïd therapie echter<br />

geassocieerd met het voorkomen van suikerziekte, zoals beschreven in hoofdstuk 14. Het<br />

moeilijke echter is dat de meest zieke mensen vaker glucocorticoïden krijgen, derhalve is het<br />

lastig om in te schatten of de ziekte zelf, of juist de therapie, heeft geleid tot suikerziekte.<br />

Tenslotte worden in het vijfde deel alle resultaten van het proefschrift samengevat in een<br />

overzichtsartikel (hoofdstuk 15) dat een volledige update tracht te geven over de wijze<br />

waarop glucocorticoïden de stofwisseling verstoren en de kans op suikerziekte sterk<br />

verhogen.<br />

De belangrijke conclusies in mijn proefschrift zijn dat: 1. tot voor kort veilig geachte lage<br />

doseringen prednisolon (7,5 mg equivalent) niet geheel veilig zijn uit een metabool perspectief,<br />

2. meest uitgesproken bijwerkingen van prednisolon niet in de nuchtere toestand optreden<br />

maar na de maaltijd typisch in de middag en avond, wanneer er hoge insuline concentraties<br />

zijn en 3. dysfunctie van de alpha en bèta cellen in de eilandjes van Langerhans op belangrijke<br />

wijze bijdragen aan de metabole bijwerkingen van glucocorticoïd behandeling.<br />

Deze conclusies hebben enkele belangrijke consequenties voor de ontwikkeling van de<br />

zogenaamde gedissocieerde glucocorticoïd receptor agonisten en de behandeling van<br />

suikerziekte veroorzaakt door prednisolon. Bij de ontwikkeling van de nieuwe gedissocieerde<br />

glucocorticoïd receptor agonisten moet er goed gekeken worden naar de postprandiale fase,<br />

omdat in de nuchtere toestand belangrijke negatieve effecten van prednisolon therapie worden<br />

gemist en derhalve het bijwerkingenprofiel kan worden onderschat. Ook is het noodzaak<br />

een compound te ontwikkelen met een bijwerkingen profiel minder dan prednisolon 7,5 mg<br />

per dag equivalent. Verder kunnen deze nieuwe inzichten leiden tot betere behandeling van<br />

suikerziekte ontstaan door glucocorticoïd therapie in de kliniek. Men zou met name de hoge<br />

335<br />

Nederlandse samenvatting


Nederlandse samenvatting<br />

bloed glucose spiegels in de middag en avond moeten behandelen met kortwerkende insuline<br />

preparaten. Thans worden veel patienten nog met langwerkende preparaten behandeld<br />

waarbij er in de ochtend te lage en potentieel levensbedreigende glucose concentraties<br />

kunnen ontstaan. Tevens is het vertrouwen op nuchtere bloed glucose waarden niet mogelijk<br />

vanwege dit specifieke patroon van te hoge bloedsuiker concentraties later op de dag en<br />

avond.Verdere studies zijn echter nodig die prospectief onderzoeken hoe de bijwerkingen<br />

van prednisolon op de suikerstofwisseling het beste in de kliniek kunnen worden behandeld<br />

of liever nog kunnen worden voorkomen.<br />

336


Dankwoord<br />

Acknowledgements


Dankwoord<br />

Een van de weinige absolute waarheden in ieder proefschrift is de zinsnede “promoveren<br />

doe je niet alleen”, en aangezien dit proefschrift niet anders is wil ik graag een aantal mensen<br />

bedanken.<br />

Allereerst alle deelnemers van de Pantheon-I, -II, en PREDEX studies. Zonder jullie was dit<br />

proefschrift niet tot stand gekomen. Ook dank voor alle gezelligheid tijdens de ongeveer 2<br />

duizend uur die de clamps en maaltijdtesten in totaal in beslag namen. Ik kan me niet meer<br />

herinneren hoeveel GB’s aan muziek en films we hebben gedraaid en uitgewisseld!<br />

Dan natuurlijk mijn promotor, Prof. Diamant. Beste Michaela, er zijn weinig personen die zo<br />

moeilijk in een kort stukje zijn te karakteriseren als jij. Enorm gedreven, perfectionistisch tot<br />

op het bot, veelzijdige ideeëngenerator, warm, betrokken en humorvol. En niet te vergeten<br />

behoorlijk mesjogge. je hebt altijd goed aangevoeld op welke manier we met ons onderzoek<br />

op zinnige wijze aan het veld konden bijdragen. Ik heb altijd het vertrouwen gehad dat<br />

wanneer ik jou ergens van kon overtuigen, het daarna overal stand zou kunnen houden. Voor<br />

een promotie per se ben je bij jou eigenlijk aan het verkeerde adres; in 4 tot 5 jaar word je de<br />

mogelijkheid geboden jezelf te ontwikkelen tot een onafhankelijk onderzoeker. Dank voor al<br />

het vertrouwen en de geboden ruimte tot groei en ontwikkeling afgelopen jaren.<br />

Graag wil ik de leden van de leescommissie bedanken.<br />

Geachte Prof. Heine, beste Rob. Ik heb door je vertrek naar Eli Lilly in de VS maar een jaar<br />

intensief met je samengewerkt; - ik heb die periode als zeer plezierig ervaren. Daarna bleef je,<br />

ondanks een paar duizend km afstand, altijd zeer geïnteresseerd en betrokken waar nodig, en<br />

was je op een of andere manier in staat mijn mailtjes nog sneller te beantwoorden dan toen je<br />

nog in het VUmc werkte. Niet onbelangrijk, al bij mijn eerste sollicitatiegesprek stelde je me<br />

gerust hoe te functioneren in een omgeving met zo veel dominante vrouwen... Dank!<br />

Geachte Prof. Sauerwein, beste Hans. Op een NVDO congres in de buurt van Arnhem<br />

bespraken we om 02.30 in de bar voor het eerst de mogelijkheid om d.m.v. stabiele isotopen<br />

metabole fluxen te meten voor en na prednisolon behandeling. Een aantal weken later was het<br />

Pantheon-II protocol een feit. Ik heb enorm veel geleerd van uw precieze methodologische<br />

benadering, heldere formulering van probleemstellingen en oneindige lijkende kennis van de<br />

stofwisseling en biochemie. Het was zeer leerzaam om een tijd bij de metabole groep op F5<br />

mee te mogen kijken! In 1 adem wil ik hierbij graag ook Dr. Serlie bedanken. Beste Mireille,<br />

hartelijk dank voor je hulp en begeleiding vanaf het allereerste begin bij het opzetten van het<br />

protocol tot aan het corrigeren van de rebuttals van onze papers!<br />

340


Geachte Dr. Ouwens, beste Margriet. We hebben een turbulente periode binnen <strong>TI</strong>P meegemaakt<br />

na jouw vertrek naar Dusseldorf. jouw oneindig enthousiasme werkt aanstekelijk en ik wil je<br />

bedanken voor alle samenwerking binnen de bioptstudies, waar ik je altijd kon benaderen<br />

voor allerhande ideeën en plannen.<br />

Geachte dr. Rustemeijer, beste Cees, veel dank voor het beoordelen van mijn proefschrift.<br />

Tevens wil ik je bedanken voor de gastvrijheid met betrekking tot de onderzoekskamer in het<br />

Amstelland ziekenhuis, hoewel mijn studies daar uiteindelijk niet zijn uitgevoerd.<br />

Geachte Prof. Dr. Kramer, beste Mark, dank voor het kritisch beoordelen van mijn proefschrift.<br />

Ik ben blij onder jouw begeleiding de opleiding tot internist te volgen!<br />

Geachte Prof. Groen en Boers, veel dank voor het kritisch lezen van mijn proefschrift.<br />

Verder gaat veel dank uit naar mijn <strong>TI</strong> <strong>Pharma</strong> collega’s met wie we het translationele project<br />

tot uitvoer hebben gebracht.<br />

LUMC. Margot, dank voor je hulp bij alle bioptstudies. Ik hoop dat je op een gegeven moment<br />

in staat bent om je onderzoeken alsnog te voltooien. Dear Bruno, thanks for stepping in into<br />

<strong>TI</strong>P and for your help with the analysis of the skeletal muscle biopsies. Geachte prof. Maassen,<br />

beste Ton, hartelijk dank voor je input gedurende het project!<br />

Ook uit Leiden, maar niet van <strong>TI</strong>P: Nienke en Leen. Samen hebben we de glucocorticoid<br />

receptor data geanalyseerd en opgeschreven. Veel dank voor het begeleiden van een dokter<br />

bij het uitvoeren en beschrijven van genetische analyses!<br />

UMCG. Beste Anke, dank voor je voor de gezelligheid tijdens alle <strong>TI</strong>P meetings! Ik bewonder<br />

dat je je proefschrift hebt weten te af te ronden na zo’n moeilijke periode. Veel succes met je<br />

opleiding bij de klinische chemie in Leeuwarden! Theo, veel dank voor je suggesties tijdens<br />

onze besprekingen.<br />

Organon/Schering-Plough/Merck. Beste Wim, als PI van het overkoepelende project had jij de<br />

niet eenvoudige taak iedereen in het gareel te houden. Ik vond het leerzaam binnen de keuken<br />

van een farmaceutische gigant mee te mogen kijken en ik waardeer enorm dat je ondanks<br />

uiterst roerige tijden je altijd 100% hebt ingezet om wetenschappelijke belangen te laten<br />

prevaleren. Veel dank ook naar Ulla en jeroen bij het opschrijven van de Organon maaltijd<br />

studies en Erik Toonen voor de input tijdens de <strong>TI</strong>P vergaderingen.<br />

341<br />

Dankwoord


Dankwoord<br />

Tot slot wil ik mijn reumatologie collega’s binnen <strong>TI</strong>P bedanken voor de prettige samenwerking.<br />

VUmc. Debby, het was fijn met je samenwerken met mijn kamer midden tussen de<br />

reumatologen in! Het eindresultaat mag er zijn! Veel succes met de afronding van jouw boekje<br />

en de start van je opleiding! Ook Marieke, veel succes met de afronding van jouw COBRA-Light<br />

data. Verder gaat mijn dank uit naar Prof. Lems, dr. Nurmohamed en Prof. Dijkmans voor het<br />

mogelijk maken van de OGTTs binnen het toch al uitgebreide COBRA-Light protocol.<br />

UMCU. Beste jos, met de MacBookjes gewired hebben we ons door de ingewikkelde statistische<br />

analyses van het Utrecht cohort gewerkt, was mooi! Succes met de laatste loodjes van je<br />

vooropleiding, we zien elkaar bij de volgende COIG... Marlies veel succes met het afronden van<br />

jouw promotie. Geachte Prof. Bijlsma, beste Hans, veel dank voor uw interesse en waardevolle<br />

suggesties gedurende de <strong>TI</strong>P meetings.<br />

Bij het uitvoeren van mijn onderzoek is de hulp van de research nurses onmisbaar geweest.<br />

Lieve Sandra, jeanette, Ingrid en Mariëlle. Ik kan oprecht zeggen dat het zonder jullie<br />

niet gelukt zou zijn. Enorm veel dank. Ook jennifer Benit, Ans en Nicolette, veel dank bij<br />

alle logistieke hulp op de research unit, het heeft het uitvoeren van mijn onderzoek veel<br />

eenvoudiger gemaakt! Ook Lilian en Louise, dank voor alle hulp over de afgelopen jaren!<br />

Furthermore, I would like to thank Dr. Mari. Dear Andrea, thanks for all your help and<br />

guidance in the different beta-cell projects that we did. The week that I spent in your lab in<br />

Padua (together with the dinner on your roof terrace and being witness of your qualities as a<br />

tour guide in different Padua churches) is something that I consider as of one the highlights of<br />

my PhD period. I have learned a great deal from your analytical mind and technical approach<br />

to interpret data. I hope that we can continue our collaboration in the future.<br />

Dr. Pouwels, beste Petra, dank je wel voor het mogelijk maken van de MRI metingen zowel qua<br />

ontwikkeling als uitwerking.<br />

Ook wil ik enkele mensen van de afdeling radiologie zelf bedanken. Vooral Ton Schweigmann<br />

(nogmaals sorry aan je vrouw dat we haar soms om 06.30 wakker belden bij onbegrijpelijke<br />

MRI storingen) en Astrid, zonder jou was het onmogelijk geweest de DEXA scans zo punctueel<br />

georganiseerd te krijgen!<br />

Erik Serné en Ed Eringa, dank voor de samenwerking op het microcirculatie manuscript.<br />

342


Dr. Kersten, beste Sander, veel dank voor de samenwerking op onze Angptl4 paper!<br />

In het AMC, dr. Ackermans, beste Mariëtte, dank voor je hulp bij het design van de isotopen<br />

infusie protocollen en voor het feit dat de verrijkingsgetallen altijd zo spoedig bepaald werden<br />

in je lab!<br />

Ook in het AMC, Prof. Karemaker, beste john, veel dank voor uw hulp bij de analyses van de<br />

portapres data (en uw gastvrijheid naar Kelly)!<br />

Daarnaast wil ik mijn directe collega’s uit het diabetes centrum en de research afdeling<br />

Interne Geneeskunde bedanken.<br />

Allereerst Nynke. Samen zijn we in de zomer van 2006 begonnen en we hebben van begin tot<br />

eind ons hele promotie traject gedeeld. Ik heb enorm veel aan je te danken gehad gedurende<br />

de jaren. je sociale vaardigheden alsmede organisatorisch talent zijn onnavolgbaar; het heeft<br />

voor mij flink wat deuren open gezet in het VUmc. Ook waardeerde ik onze gesprekken over<br />

wat buiten ons werk in ons leven speelde en hadden we gezellige etentjes met Boudewijn en<br />

Lisette. Fijn dat je mijn paranymf wilt zijn!<br />

Mijn andere roomie, Renate. Wat fijn dat jij het glucocorticoiden stokje hebt overgenomen!<br />

Never a dull moment met jou op de kamer, dance moves, weekend verhalen over wielrennen<br />

in de Limburgse heuvels en natuurlijk je messcherpe retoriek, met name na wat alcoholische<br />

consumpties! Succes met het afronden van jouw GC proefschrift!<br />

Bunckie, bedankt voor de mooie tijden, met name alle congressen all over the world die we<br />

hebben bezocht. What happens op de via pescarotto stays...<br />

Tus, enorm veel dank voor alle interesse in en het creatief meedenken met mijn protocol in<br />

de opstartfase. Als nestor voor jonge onderzoekers werd de overstap vanuit het AMC naar het<br />

VUmc makkelijk!<br />

Remco, ik heb de gezelligheid in de Meander gewaardeerd, gesprekken over wetenschap<br />

tijdens tennis spelletjes op de computer! En met name je gastvrijheid in Zuid-Afrika<br />

voor Lisette en mij. Game driven in Kruger en kamperen en braaien in Tsenzde onder de<br />

sterrenhemel; het kon slechter.<br />

Verder wil ik graag mijn andere collega’s bedanken voor de leuke tijd: Larissa, Weena, Luuk,<br />

Diane, Mariska, Roger, Michiel, Rick, Liselotte (ik hou van je humor), Eelco, Annemarie,<br />

343<br />

Dankwoord


Dankwoord<br />

Carolien, Mirjam en jennifer. Verder Kelly en Marcel: het was ontzettend leuk om met zulke<br />

gedreven studenten samen te werken!<br />

Myrte, we hebben heel wat uren geclampt in de metabole kamer! Je inzet, precisie en werklust<br />

zijn een voorbeeld voor velen; samen hebben we ondanks alle strubbelingen tussen de twee<br />

Amsterdamse academische centra de studie tot een succesvol einde weten te brengen!<br />

Ik wil graag mijn vrienden bedanken voor alle leuke dingen buiten het werk, hoewel de<br />

meesten niet alleen geneeskunde hebben gestudeerd, maar ook nog eens gepromoveerd zijn<br />

en de gesprekken derhalve toch ook nog genoeg over ons werk gaan... Ons contact, de laatste<br />

2 jaar verrijkt met de komst van alle kids, waardeer ik enorm. De jaarlijkse wintersport,<br />

de wekelijkse zaalvoetbalwedstrijden in de middenmoot van de 4de klasse KNVB, de<br />

fietsweekenden waar de km’s steeds langer lijken te worden en dezelfde heuvels steeds steiler,<br />

en natuurlijk alle etentjes en borrels, zijn altijd erg gezellig!! Danny, top dat je mijn paranimf<br />

wil zijn! Jonathan, dank voor alle mooie happenings along the way en een gaaf stagejaar in<br />

Vancouver in onze studententijd!<br />

Schoonouders Frits en Lieke, stand-in oppas bij noodgevallen, klussen in ons huis, (race)<br />

fietsen tussen Amsterdam en Soesterberg, niets is jullie teveel. Dank voor jullie liefdevolle<br />

interesse en betrokkenheid afgelopen jaren! En tante Antoinette dank voor je fameuze<br />

verwenweekendjes in Raalte voor het hele gezin!<br />

Lieve Sander en joost, ik zou mijn jeugd zonder jullie als broers mij niet kunnen voorstellen.<br />

Oneindig veel hockeyen en voetballen zowel binnens- als buitenshuis en dat we af en toe wat<br />

bij elkaar braken hoorde er een beetje bij...<br />

Lieve Pap en Mam, enorm bedankt voor alle support en stimulatie van jongs af aan om<br />

mezelf te ontplooien. jullie afzonderlijke steun is onvoorwaardelijk en lijkt oneindig. Een<br />

meer vastere bodem had ik niet kunnen wensen. jan en Marianne, ik hoef geloof ik niet te<br />

benadrukken wat voor enorme aanwinst jullie voor de familie zijn!!<br />

Tenslotte jij lieve Lisette! Na alle verre reizen die we met z’n 2-en hebben gemaakt, zijn we<br />

nu met kleine Sam neergedaald in de Watergraafsmeer. Het kost jou weinig moeite om me te<br />

laten zien dat er veel belangrijkere en leukere dingen in het leven zijn dan werk. je zorgt altijd<br />

voor voldoende “jus” in ons leven...<br />

Lobby you!<br />

344


Biography


Biography<br />

Daniël Henri van Raalte werd geboren op 8 juli 1980 in het Amsterdamse Slotervaart<br />

Ziekenhuis. Na het afronden van de Amstelveense basisschool Martin Luther King School, ging<br />

hij naar het Vossius Gymnasium in Amsterdam. In het jaar na zijn eindexamen werkte hij bij<br />

belasting- en accounting specialist KPMG en sorteerde hij fruit in een Israelische kibbutz, en<br />

werd zo klaargestoomd voor de opleiding Geneeskunde aan de Universiteit van Amsterdam.<br />

Hij behaalde zijn propedeutisch- (2000), doctoraal- (2004) en artsexamen (2006) cum laude.<br />

Als student deed hij onderzoek bij de afdeling Vasculaire Geneeskunde van het Academisch<br />

Medisch Centrum (AMC) onder supervisie van Prof. Dr. E.S. Stroes en Dr. j.A. Kuivenhoven.<br />

Dit werd gevolgd door een stage bij het Atherosclerosis Specialty Laboratory/Faculty of<br />

<strong>Pharma</strong>ceutical Sciences van de University of British Columbia (UBC) in Vancouver, Canada<br />

onder begeleiding van Prof. Dr. P.H. Pritchard, Prof. Dr. K.M. Wasan en Dr. M. Li. Gedurende zijn<br />

studie speelde hij hockey bij Pinoké heren 1 en 2 en bij de Vancouver Hawks HC, en was hij<br />

chauffeur voor AFC Ajax in de Champions League in het seizoen 2003-2004. In 2006 startte<br />

hij als promovendus bij het Diabetes Centrum van het Vrije Universiteit Medisch Centrum in<br />

Amsterdam onder begeleiding van Prof. Dr. M. Diamant waarvan de meeste resultaten in dit<br />

proefschrift zijn beschreven. Hij ontving in 2010 een Keystone Fellowship. Tevens werd een<br />

beursaanvraag door de European Foundation for the Study of Diabetes (EFSD) gehonoreerd,<br />

waardoor het onderzoek naar de effecten van glucocorticoïden op de suikerstofwisseling<br />

momenteel wordt voortgezet. Sinds mei 2011 is hij in opleiding tot internist in het Haarlemse<br />

Kennemer Gasthuis onder supervisie van Prof. Dr. R.W. ten Kate en Prof. Dr. M.H.H. Kramer.<br />

Daniël woont samen met Lisette Klip en ze hebben een zoon genaamd Sam.<br />

348


Daniël Henri van Raalte was born on july 8 th , 1980 in Amsterdam. After finishing primary<br />

education at the Martin Luther King School in Amstelveen, he went on to the Vossius Gymnasium<br />

in Amsterdam. In the year following his final exams, he worked at the large tax and auditor<br />

specialist KPMG and sorted fruits in an Israeli Kibbutz, and was thus properly prepared to<br />

start his medical training at the University of Amsterdam. He completed his first year (2000),<br />

master’s (2004) and MD (2006) examinations cum laude. As a student, he participated in<br />

research projects at the department of Vascular Medicine of the Academic Medical Center<br />

(AMC) supervised by Prof. Dr. E.S. Stroes and Dr. j.A. Kuivenhoven. This was followed by a<br />

scientific internship at the Atherosclerosis Specialty Laboratory/Faculty of <strong>Pharma</strong>ceutical<br />

Sciences of the University of British Columbia (UBC) in Vancouver, Canada under guidance of<br />

Prof. Dr. P.H. Pritchard, Prof. Dr. K.M. Wasan and Dr. M. Li. During his studies, he played field<br />

hockey at a competitive level for Pinoké and Vancouver Hawks HC, and was driver for soccer<br />

club AFC Ajax in the 2003-2004 Champions League season. In 2006 he started his PhD at the<br />

Diabetes Center of the VU University Medical Center in Amsterdam under supervision of Prof.<br />

Dr. M. Diamant. Most of the research performed during this period is presented in this thesis.<br />

For his research, he received a Keystone Fellowship in 2010. In addition, he co-authored a<br />

grant awarded by the European Foundation for the Study of Diabetes (EFSD), enabling<br />

continuation of research into the effects of glucocorticoids on carbohydrate metabolism. In<br />

May 2011, he started his residency in Internal Medicine at the Kennemer Hospital, Haarlem,<br />

under supervision of Prof. Dr. R.W. ten Kate and Prof. Dr. M.H.H. Kramer.<br />

Daniël lives together with Lisette Klip and they have a son named Samuel.<br />

349<br />

Biography


List of<br />

Publications


List of Publications<br />

1. van Genugten RE, van Raalte DH, Diamant M. DPP-4 inhibition restores glucocorticoid-<br />

352<br />

induced pancreatic beta-cell dysfunction in human metabolic syndrome. Submitted<br />

2. van Genugten RE, Serne EH, Heymans MW, van Raalte DH, Diamant M. Postprandial<br />

microvascular function deteriorates along with gradual worsening of insulin sensitivity<br />

and glucose tolerance in men with metabolic syndrome and type 2 diabetes. Submitted.<br />

3. van Genugten RE, Möller-Goede DL, van Raalte DH, Diamant M. Extra-pancreatic effects<br />

of incretin-based therapies: potential benefit for cardiovascular risk management in<br />

type 2 diabetes. Submitted.<br />

4. van Raalte DH, Diamant M, Ouwens DM, Ijzerman RG, Linssen MML, Guigas B, Eringa<br />

EC, Serné EH. Glucocorticoid treatment impairs microvascular function in healthy<br />

males, in association with its adverse effects on glucose metabolism and blood pressure.<br />

Submitted.<br />

5. Brands M, van Raalte DH, joão Ferraz M, Sauerwein HP, Verhoeven Aj, Aerts jM,<br />

Diamant M, Serlie Mj. Glycosphingolipid metabolism and mitochondrial Function do not<br />

explain glucocorticoid-induced insulin resistance in healthy men. Submitted.<br />

6. van Raalte DH, Kwa KAA, van Genugten RE, Tushuizen ME, Holst jj, Deacon CF,<br />

Karemaker jM, Heine Rj, Mari, A, Diamant M. Islet-cell dysfunction induced by<br />

glucocorticoid treatment: potential role for altered sympathovagal balance? Metabolism,<br />

in revision.<br />

7. van Raalte DH, Brands M, Serlie Mj, Mudde K, Stienstra R, Sauerwein HP, Kersten S,<br />

Diamant M. Angiopoietine-like protein 4 is differentially regulated by glucocorticoids<br />

and insulin in vitro and in vivo in healthy humans. Exp Clin Endocrinol Diabetes. In press.<br />

8. van Raalte DH, Den Uyl D, Nurmohamed MT, Diamant M, Lems WF. Glucocorticoid<br />

treatment and hyperglycemia. Arthritis Rheum. 2012. july 25 Epub ahead of print.<br />

9. van Raalte DH, Diamant M. Goals of treatment in type 2 diabetes: beta-cell<br />

preservation. In: Evidence-based management of diabetes. TFM Publishing Ltd. 2012.<br />

10. van Raalte DH, van Leeuwen N, Simonis-Bik AM, Nijpels G, van Haeften TW, Boomsma<br />

DI, Kramer MHH, Heine Rj, Maassen jA, Slagboom PE, Willemsen G, de Geus Ej, Dekker<br />

jM, Eekhoff EM, Diamant M, ‘t Hart LM. Glucocorticoid gene receptor polymorphisms<br />

are associated with reduced first-phase glucose-stimulated insulin secretion and<br />

disposition index in women, but not in men. Diabet Med. 2012.


11. Den Uyl D, van Raalte DH, Nurmohamed MT, Lems WF, Bijlsma jW, Hoes jN, Dijkmans<br />

BA, Diamant M. Metabolic effects of high-dose prednisolone treatment in early<br />

rheumatoid arthritis: diabetogenic effects and inflammation reduction on the balance.<br />

Arthritis Rheum. 2012. 64(3):639-646.<br />

12. Hoes jN, van der Goes MC, van Raalte DH, van der Zijl Nj, den Uyl D, Lems WF, Lafeber<br />

FP, jacobs jW, Welsing PM, Diamant M, Bijlsma jW. Glucose tolerance, insulin sensitivity<br />

and beta-cell function in patients with rheumatoid arthritis treated with or without<br />

low-to-medium dose glucocorticoids. Ann Rheum Dis. 2011. 70(11):1887-94.<br />

13. van Raalte DH, Diamant M. Glucolipotoxicity and beta-Cells in type 2 diabetes: target<br />

for durable therapy? Diab Clin Res Pract. 2011. Suppl 1:S37-46.<br />

14. van Genugten RE, van Raalte DH, Diamant M. Dipeptidyl peptidase-4 inhibitors and<br />

preservation of pancreatic islet-cell dysfunction: a critical appraisal of the evidence.<br />

Diabetes Obes Metab. 2012. 14(2):101-11.<br />

15. Linssen MM, van Raalte DH, Toonen Ej, Alkema W, van der Zon GC, Dokter WH,<br />

Diamant M, Guigas B, Ouwens DM. Prednisolone-induced beta-cell dysfunction is<br />

associated with impaired endoplasmic reticulum homeostasis in INS-1E cells. Cell<br />

Signal. 2011. 23(11):1708-15.<br />

16. van Raalte DH, Brands M, van der Zijl Nj, Muskiet MH, Pouwels Pj, Ackermans MT,<br />

Sauerwein HP, Serlie Mj, Diamant M. Low-dose glucocorticoid treatment affects multiple<br />

aspects of intermediary metabolism in healty humans: a randomised controlled trial.<br />

Diabetologia. 2011. 54(8):2103-12.<br />

17. van Raalte DH, van Genugten RE, Linssen MM, Ouwens DM, Diamant M. Glucagonlike<br />

peptide-1 receptor agonist treatment prevents glucocorticoid-induced glucose<br />

intolerance and islet-cell dysfunction in humans. Diabetes Care. 2011. 34(2):412-7.<br />

18. van der Zijl Nj, Goossens GH, Moors CC, van Raalte DH, Muskiet MH, Pouwels Pj, Blaak<br />

EE, Diamant M. Ectopic fat storage in the pancreas, liver and abdominal fat depots:<br />

impact on beta-cell function in individuals with impaired glucose metabolism. J Clin<br />

Endocrinol Metab. 2011. 96(2):459-67.<br />

19. van Raalte DH, van der Zijl Nj, Diamant M. Pancreatic steatosis in humans: cause or<br />

marker of lipotoxicity? Curr Opin Clin Nutr Metab Care. 2010. 13(4):478-85.<br />

353<br />

List of Publications


List of Publications<br />

20. van Raalte DH, Nofrate V, Bunck MC, van Iersel T, Elassais Schaap j, Nässander UK,<br />

354<br />

Heine Rj, Mari A, Dokter WHA, Diamant M. Acute and 2-week exposure to prednisolone<br />

impair different aspects of beta-cell function in healthy men. Eur J Endocrinol. 2010.<br />

162(4):729-35.<br />

21. van Genugten RE, van Raalte DH, Diamant M. Does glucagon-like peptide-1 receptor<br />

agonist therapy add value in the treatment of type 2 diabetes? Focus on exenatide. Diab<br />

Clin Res Pract. 2009.86 Suppl 1:S26-34.<br />

22. van Raalte DH, Ouwens DM, Diamant M. Novel insights into glucocorticoid-mediated<br />

diabetogenic effects: towards expansion of therapeutic options? Eur J Clin Invest. 2009.<br />

39(2):81-93<br />

23. Rip j, Nierman MC, Sierts jA, Peterson W, Den Oever K, van Raalte DH, Ross Cj, Hayden<br />

MR, Bakker AC, Dijkhuizen P, Hermens WT, Twisk j, Stroes E, Kastelein jj, Kuivenhoven<br />

JA, Meulenberg JM. Gene therapy for lipoprotein lipase deficiency: working towards<br />

clinical application. Hum Gene ther. 2005. 16(11):1276-86.<br />

24. Nierman MC, Rip j, Kuivenhoven jA, van Raalte DH, Hutten BA, Sakai N, Kastelein jj,<br />

Stroes ES. Carriers of the frequent lipoprotein lipase S447X variant exhibit enhanced<br />

postprandial ApoB-48 clearance. Metabolism. 2005. 54(11):1499-503.<br />

25. van Raalte DH, Li M, Pritchard PH, Wasan KM. Peroxisome proliferator-activated<br />

Receptor (PPAR)-alpha, a pharmacological target with a promising future.<br />

<strong>Pharma</strong>ceutical Res. 2004. 21(9):1531-8.


List of<br />

co-author<br />

affiliations


List of co-author affiliations<br />

Andrea Mari, Institute of Biomedical Engineering, National Research Council, Padova Italy<br />

Andreas Fritsche, Department of Internal Medicine, Eberhard-Karls University of Tübingen,<br />

Tübingen, Germany<br />

Annemarie M. Simonis-Bik, Diabetes Center/Department of Internal Medicine, VU<br />

University Medical Center, Amsterdam, The Netherlands<br />

Arthur J. Verhoeven, Department of Medical Biochemistry, Academic Medical Center,<br />

Amsterdam, The Netherlands.<br />

Ben A.C Dijkmans, Department of Rheumatology, VU University Medical Center, Amsterdam,<br />

The Netherlands<br />

Bruno Guigas, Department of Molecular Cell Biology, Leiden University Medical Center,<br />

Leiden, The Netherlands<br />

Carolyn F. Deacon, Department of Biomedical Sciences, The Panum Institute, University of<br />

Copenhagen, Copenhagen, Denmark<br />

Debby den Uyl, Department of Rheumatology, VU University Medical Center, Amsterdam, The<br />

Netherlands<br />

D. Margriet ouwens, Institute of Clinical Biochemistry and Pathobiochemistry, German<br />

Diabetes Center, Düsseldorf, Germany<br />

Doret I. Boomsma, Biological Psychology, VU University, Amsterdam, The Netherlands<br />

Eco J. de Geus, Biological Psychology, VU University, Amsterdam, The Netherlands<br />

E. Marelise Eekhoff, Diabetes Center/Department of Internal Medicine, VU University<br />

Medical Center, Amsterdam, The Netherlands<br />

Erik J.M. Toonen, Department of Internal Medicine, Radboud University Nijmegen Medical<br />

Center, Nijmegen, The Netherlands<br />

Erik H. Serné, Department of Internal Medicine, VU University Medical Center, Amsterdam,<br />

The Netherlands<br />

Etto C. Eringa, Laboratory for Physiology, Institute for Cardiovascular Research, VU University<br />

Medical Center, Amsterdam, The Netherlands<br />

Fausto Machicao, Department of Internal Medicine, Eberhard-Karls University of Tübingen,<br />

Tübingen, Germany<br />

358


Floris P.G.J. Lafeber, Department of Rheumatology and Clinical Immunology, University<br />

Medical Center Utrecht, Utrecht, The Netherlands<br />

Gerard C. van der Zon, Department of Molecular Cell Biology, Leiden University Medical<br />

Center, Leiden, The Netherlands<br />

Giel Nijpels, The EMGO Institute for Health and Care Research, VU University Medical Center,<br />

Amsterdam, the Netherlands<br />

Gonneke Willemsen, Biological Psychology, VU University, Amsterdam, The Netherlands<br />

Hans P. Sauerwein, Department of Endocrinology and Metabolism, Academic Medical Center,<br />

Amsterdam, The Netherlands.<br />

Hans-U. Häring, Department of Internal Medicine, Eberhard-Karls University of Tübingen,<br />

Tübingen, Germany<br />

Harald Staiger, Department of Internal Medicine, Eberhard-Karls University of Tübingen,<br />

Tübingen, Germany<br />

Heidi Mueller, Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes<br />

Center, Düsseldorf, Germany<br />

Jacqueline M. Dekker, The EMGO Institute for Health and Care Research, VU University<br />

Medical Center, Amsterdam, the Netherlands<br />

Jens J. Holst, Department of Biomedical Sciences, The Panum Institute, University of<br />

Copenhagen, Copenhagen, Denmark<br />

Jeroen Elaissaiss-Schaap, Department of Early Clinical Research and Experimental Medicine,<br />

MSD Research Laboratories, Oss, The Netherlands<br />

Johannes F.G. Aerts, Department of Medical Biochemistry, Academic Medical Center,<br />

Amsterdam, The Netherlands.<br />

Johannes W.G. Jacobs, Department of Rheumatology and Clinical Immunology, University<br />

Medical Center Utrecht, Utrecht, The Netherlands<br />

Johannes W.J. Bijlsma, Department of Rheumatology and Clinical Immunology, University<br />

Medical Center Utrecht, Utrecht, The Netherlands<br />

John M. Karemaker, Department of Physiology, Academic Medical Center, University of<br />

Amsterdam, Amsterdam, The Netherlands<br />

359<br />

List of co-author affiliations


List of co-author affiliations<br />

Jos N. Hoes, Department of Rheumatology and Clinical Immunology, University<br />

Medical Center Utrecht, Utrecht, The Netherlands<br />

Karin Mudde, Nutrition, Metabolism and Genomics Group, Division of Human Nutrition,<br />

Wageningen University, Wageningen, The Netherlands.<br />

Kelly A.A. Kwa, Diabetes Center/Department of Internal Medicine, VU University Medical<br />

Center, Amsterdam, The Netherlands<br />

Maarten E. Tushuizen, Diabetes Center/Department of Internal Medicine, VU University<br />

Medical Center, Amsterdam, The Netherlands<br />

Marcel H. Muskiet, Diabetes Center/Department of Internal Medicine, VU University Medical<br />

Center, Amsterdam, The Netherlands<br />

Margot M.L. Linssen, Department of Molecular Cell Biology, Leiden University Medical<br />

Center, Leiden, The Netherlands<br />

Mariëtte T. Ackermans, Department of Clinical Chemistry, Laboratory of Endocrinology,<br />

Academic Medical Centre, Amsterdam, The Netherlands<br />

Maria Joao Ferrez, Department of Medical Biochemistry, Academic Medical Center,<br />

Amsterdam, The Netherlands.<br />

Mark H.H. Kramer, Department of Internal Medicine, VU University Medical Center,<br />

Amsterdam, The Netherlands<br />

Marlies C. van der Goes, Department of Rheumatology and Clinical Immunology, University<br />

Medical Center Utrecht, Utrecht, The Netherlands<br />

Mathijs C. Bunck, Diabetes Center/Department of Internal Medicine, VU University Medical<br />

Center, Amsterdam, The Netherlands<br />

Michaela Diamant, Diabetes Center/Department of Internal Medicine, VU University Medical<br />

Center, Amsterdam, The Netherlands<br />

Mike T. Nurmohamed, Department of Rheumatology, VU University Medical Center,<br />

Amsterdam, The Netherlands<br />

Mireille J. Serlie, Department of Endocrinology and Metabolism, Academic Medical Center,<br />

Amsterdam, The Netherlands.<br />

360


Myrte Brands, Department of Endocrinology and Metabolism, Academic Medical Center,<br />

Amsterdam, The Netherlands<br />

Leen M. ‘t Hart, Section Molecular Epidemiology, Department of Medical Statistics and<br />

Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands.<br />

Nienke van Leeuwen, Department of Molecular Cell Biology, Leiden University Medical<br />

Center, Leiden, The Netherlands.<br />

Nynke J. van der Zijl, Diabetes Center/Department of Internal Medicine, VU University<br />

Medical Center, Amsterdam, The Netherlands<br />

Paco M.J. Welsing, Department of Rheumatology and Clinical Immunology, University<br />

Medical Center Utrecht, Utrecht, The Netherlands<br />

P. Eline Slagboom, Section Molecular Epidemiology, Department of Medical Statistics and<br />

Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands.<br />

Petra J.W. Pouwels, Department of Physics and Medical Technology, VU University Medical<br />

Center, Amsterdam, The Netherlands<br />

Renate E. van Genugten, Diabetes Center/Department of Internal Medicine, VU University<br />

Medical Center, Amsterdam, The Netherlands<br />

Richard G. IJzerman, Department of Internal Medicine, VU University Medical Center,<br />

Amsterdam, The Netherlands<br />

Rinke Stienstra, Nutrition, Metabolism and Genomics Group, Division of Human Nutrition,<br />

Wageningen University, Wageningen, The Netherlands.<br />

Rob J. Heine, Diabetes Center/Department of Internal Medicine, VU University Medical<br />

Center, Amsterdam, The Netherlands<br />

Sander Kersten, Nutrition, Metabolism and Genomics Group, Division of Human Nutrition,<br />

Wageningen University, Wageningen, The Netherlands.<br />

Stefan Lehr, Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes<br />

Center, Dusseldorf, Germany<br />

Silke A. Schäfer, Department of Internal Medicine, Eberhard-Karls University of Tübingen,<br />

Tübingen, Germany<br />

Thijs van Iersel, Xendo Drug Development, Groningen, The Netherlands<br />

361<br />

List of co-author affiliations


List of co-author affiliations<br />

Timon W. van Haeften, Department of Internal Medicine, Utrecht University Medical Center,<br />

Utrecht, the Netherlands<br />

Ton J.A. Maassen, Leiden University Medical Center, Leiden<br />

Ulla K. Nässander, Department of Early Clinical Research and Experimental Medicine, MSD<br />

Research Laboratories, Oss, The Netherlands<br />

Valentina Nofrate, Institute of Biomedical Engineering, National Research Council, Padova,<br />

Italy<br />

Willem F. Lems, Department of Rheumatology, VU University Medical Center, Amsterdam,<br />

The Netherlands<br />

Wim H. Dokter, Department of Immune Therapeutics, MSD Research Laboratories, Oss, The<br />

Netherlands<br />

Wynand Alkema, Department of Molecular Design and Informatics, MSD Research<br />

Laboratories, Oss, The Netherlands<br />

362


Color Figures


Color Figures<br />

Chapter 1 Figure 1. Overview of the therapeutic and adverse effects induced by glucocorticoid treatment.<br />

366


Chapter 15 Figure 1. Currently known mechanisms by which glucocorticoids (GCs) induce hyperglycemia<br />

in healthy humans.<br />

Chapter 15 Figure 2. Hypothetic representation of the balance between systemic inflammation severity<br />

and (high)-dose glucocorticoid (GC) treatment in relation to glucose tolerance.<br />

367<br />

Color Figures

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!