Elsevier

Metabolism

Volume 62, Issue 1, January 2013, Pages 21-33
Metabolism

Review
11beta-Hydroxysteroid dehydrogenase type 1 inhibitors: novel agents for the treatment of metabolic syndrome and obesity-related disorders?

https://doi.org/10.1016/j.metabol.2012.05.002Get rights and content

Abstract

Objective

Metabolic syndrome (MetS) and Cushing's syndrome share common features. It has been proposed that increased glucocorticoid activity at peripheral tissues may play a role in the pathogenesis of MetS and obesity-related disorders. It is well-known that intracellular cortisol concentrations are determined not only by plasma levels but also by the activity of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) which catalyzes the conversion of inactive cortisone to active cortisol, especially in the liver and adipose tissue. Another isoenzyme exists, the 11β-hydroxysteroid dehydrogenase type 2, which acts in the opposite direction inactivating cortisol to cortisone in the kidney.

This review considers the significance of the 11β-HSD1 inhibition in the treatment of several features of MetS and provides current data about the development of 11β-HSD1 inhibitors, as new agents for this purpose.

Materials/Methods

Using PubMed, we searched for publications during the last 20 years regarding the development of 11β-HSD1 inhibitors.

Results

Emerging data from animal and human studies indicate an association of 11β-HSD1 over-expression with obesity and disorders in glucose and lipid metabolism. This has led to the hypothesis that selective inhibition of 11β-HSD1 could be used to treat MetS and diabetes. Indeed, natural products and older agents such as thiazolidinediones and fibrates seem to exert an inhibitory effect on 11β-HSD1, ameliorating the cardiometabolic profile. In view of this concept, novel compounds, such as adamantyltriazoles, arylsulfonamidothiazoles, anilinothiazolones, BVT2733, INCB-13739, MK-0916 and MK-0736, are currently under investigation and the preliminary findings from both experimental and human studies show a favourable effect on glucose and lipid metabolism, weight reduction and adipokine levels.

Conclusions

Many compounds inhibiting 11β-ΗSD1 are under development and preliminary data about their impact on glucose metabolism and obesity-related disorders are encouraging.

Introduction

The metabolic syndrome (MetS) is a cluster of abnormalities including central obesity, impaired glucose tolerance, hypertension and dyslipidemia [1]. Insulin resistance is the main defect linking the individual components of MetS, although the strength of this correlation varies between, and even within, different populations [1]. However, MetS shares many of the features of Cushing's syndrome and it has been proposed that dysregulation of glucocorticoid action might contribute to the pathogenesis of MetS [2]. Indeed, some studies have shown that circulating cortisol levels are higher in patients with MetS compared with healthy controls [3], [4]. There is also evidence of increased activity of the hypothalamic–pituitary–adrenal (HPA) axis along with a perturbed feedback control in MetS [3].

Some studies showed a positive relationship between cortisol and waist circumference [4], [5] in contrast with the findings of others [6]. Similarly, increased urinary free cortisol excretion in patients with MetS has been reported [7] as well as increased urinary cortisone/cortisol ratio in subjects with increased abdominal fat compared with those with peripheral fat distribution, suggesting an increase in the peripheral metabolism of cortisol [6].

Cortisol excess seems to be more associated with insulin resistance, the major pathogenetic mechanism in MetS, rather than obesity per se. Increased cortisol (urinary free and serum overnight) levels are positively associated with insulin resistance [assessed using the homeostasis model assessment (HOMA)] [8], [9] and this association is independent of body weight. Furthermore, cortisol clearance seems to be inversely associated with insulin sensitivity irrespective of body fat [10]. Another finding indicating that higher cortisol levels promote the manifestation of MetS rather than obesity alone is that higher cortisol levels are associated with reduced insulin secretion [1].

Growing evidence suggests that MetS and central obesity may result from an increased availability of glucocorticoids at the tissue level (mainly liver and adipose tissue). A major determinant of glucocorticoid local action seems to be the expression of the enzyme 11-beta-hydroxysteroid dehydrogenase (11β-HSD) [2]. Two isoforms of 11β-HSD exist, the 11β-HSD type 1 (11β-HSD1) and 11β-HSD type 2 (11β-HSD2). The former is expressed in many tissues, such as liver, adipose tissue and central nervous system, as well as in skeletal and smooth muscles, fibroblasts and immune cells [11], [12]. 11β-HSD1 is a nicotinamide adenine dinucleotide phosphate (NADPH)-dependent enzyme, acting predominantly as a reductase, converting inactive cortisone to active cortisol, rather than a dehydrogenase (in the opposite direction). 11β-HSD1 facilitates the action of glucocorticoids in key-targets, such as liver and adipose tissue, which is mediated via glucocorticoid receptors (GR). Most studies have shown an increased expression of 11β-HSD1 in adipose tissue in obesity states, resulting in higher intracellular conversion of cortisone to cortisol [13].

In contrast, 11β-HSD2 is a high affinity NADPH-dependent dehydrogenase, which is expressed in mineralocorticoid target tissues, predominantly the kidney, but also in the colon, placenta, sweat and salivary glands [11]. This 11β-HSD isoenzyme catalyzes the inactivation of cortisol to cortisone, thus protecting the mineralocorticoid receptor from excess stimulation by cortisol [11], [12]. The significance of 11β-HSD2 can be further enhanced when looking in the pathogenesis of the rare syndrome of “apparent mineralocorticoid excess”, an autosomal recessive inherited disorder characterized by 11β-HSD2 deficiency. This disorder leads to inappropriate binding of cortisol to mineralocorticoid receptors in the distal tubule, resulting in low birth weight, short stature, hypertension, hypokalemia, metabolic acidosis and low renin and aldosterone levels [14].

The purpose of the present review is to provide current understanding about the role of 11β-HSD1 in glucose and lipid homeostasis and the existing data about its inhibition as a new therapeutic target in MetS and obesity-related disorders.

Section snippets

Data from animal studies

From preclinical studies, there is evidence for a beneficial effect on glucose homeostasis and weight reduction in diabetic and obese mouse models after inhibition of 11β-HSD1 [15]. In particular, 11β-HSD1-knockout mice demonstrate an attenuated activation of the key hepatic gluconeogenic enzymes glucose-6-phosphatase and phosphoenolpyruvate carboxykinase (PEPCK). Despite high-fat feeding, they are protected from hyperglycemia, obesity and dyslipidemia [16], [17].

Data from in vitro studies

11β-HSD1 expression has also

Non-selective 11β-HSD1 inhibitors

The data provided above indicate a key role of 11β-HSD1 in glucose and lipid metabolism and support the notion that inhibition of this enzyme may be a new therapeutic approach for MetS and obesity-related disorders. This role has been identified in some substances, although they are characterized by a concurrent inhibition of 11β-HSD2.

Selective 11β-HSD1 inhibitors

As indicated by the aforementioned data, there is a need for an ideal 11β-HSD1 inhibitor to be selective for this isoenzyme. Several products, antidiabetic and hypolipidemic agents have been shown to exert a selective inhibitory effect on 11β-HSD1, in addition to their well-known mode of action (Table 2).

Conclusions

11β-HSD1 is a key enzyme in corticosteroid metabolism at a peripheral tissue level. Its over-expression has been implicated in the pathogenesis of central obesity, MetS and dysregulation of glucose and lipid metabolism. Data from animal studies have demonstrated that 11β-HSD1 inhibition can improve several components of the MetS. Novel compounds with different mechanistic effect, are currently under investigation and the emerging data are encouraging. It remains for these promising preliminary

Author contributions

P.A., V.G.A., A.K. and D.P.M. designed the study. P.A. and F.A. condcuted and collected the data F.A., M.K. and D.P.M. analysed the study. P.A. wrote the manuscript.

Conflict of interest

This review was written independently. The authors did not receive any funding for the preparation of the manuscript. The authors have given talks, attended conferences and participated in advisory boards and trials sponsored by various pharmaceutical companies.

The authors declare that they have no conflict of interest to disclose.

References (114)

  • R. Mukherjee et al.

    Identification, characterization, and tissue distribution of human peroxisome proliferator-activated receptor (PPAR) isoforms PPARgamma2 versus PPARgamma1 and activation with retinoid X receptor agonists and antagonists

    J Biol Chem

    (1997)
  • J. Berger et al.

    Peroxisome proliferator-activated receptor-gamma ligands inhibit adipocyte 11beta -hydroxysteroid dehydrogenase type 1 expression and activity

    J Biol Chem

    (2001)
  • A. Hermanowski-Vosatka et al.

    PPARα agonists reduce 11β-hydroxysteroid dehydrogenase type 1 in the liver

    Biochem Biophys Res Commun

    (2000)
  • R.A. Srivastava

    Fenofibrate ameliorates diabetic and dyslipidemic profiles in KKAy mice partly via down-regulation of 11beta-HSD1, PEPCK and DGAT2. Comparison of PPARalpha, PPARgamma, and liver x receptor agonists

    Eur J Pharmacol

    (2009)
  • Y. Iwasaki et al.

    Is the metabolic syndrome an intracellular Cushing state? Effects of multiple humoral factors on the transcriptional activity of the hepatic glucocorticoid-activating enzyme (11beta-hydroxysteroid dehydrogenase type 1) gene

    Mol Cell Endocrinol

    (2008)
  • S. Olson et al.

    Adamantyl triazoles as selective inhibitors of 11β-hydroxysteroid dehydrogenase type 1

    Bioorg Med Chem Lett

    (2005)
  • C. Yuan et al.

    The discovery of 2-anilinothiazolones as 11β-HSD1 inhibitors

    Bioorg Med Chem Lett

    (2007)
  • T. Ishii et al.

    Augmentation of 11beta-hydroxysteroid dehydrogenase type 1 in LPS-activated J774.1 macrophages–role of 11beta-HSD1 in pro-inflammatory properties in macrophages

    FEBS Lett

    (2007)
  • M. Siu et al.

    N-(Pyridin-2-yl) arylsulfonamide inhibitors of 11beta-hydroxysteroid dehydrogenase type 1: discovery of PF-915275

    Bioorg Med Chem Lett

    (2009)
  • V.G. Athyros et al.

    Comparison of four definitions of the metabolic syndrome in a Greek (Mediterranean) population

    Curr Med Res Opin

    (2010)
  • P. Anagnostis et al.

    Clinical review: the pathogenetic role of cortisol in the metabolic syndrome: a hypothesis

    J Clin Endocrinol Metab

    (2009)
  • Y. Sen et al.

    Children and adolescents with obesity and the metabolic syndrome have high circulating cortisol levels

    Neuro Endocrinol Lett

    (2008)
  • E.S. Epel et al.

    Stress and body shape: stress-induced cortisol secretion is consistently greater among women with central fat

    Psychosom Med

    (2000)
  • J. Marniemi et al.

    Visceral fat and psychosocial stress in identical twins discordant for obesity

    J Intern Med

    (2002)
  • M.J. Weigensberg et al.

    Association between the metabolic syndrome and serum cortisol in overweight Latino youth

    J Clin Endocrinol Μetab

    (2008)
  • M. Misra et al.

    Lower growth hormone and higher cortisol are associated with greater visceral adiposity, intramyocellular lipids, and insulin resistance in overweight girls

    Am J Physiol Endocrinol Metab

    (2008)
  • T. Reinehr et al.

    Cortisol and its relation to insulin resistance before and after weight loss in obese children

    Horm Res

    (2004)
  • H.B. Holt et al.

    Cortisol clearance and associations with insulin sensitivity, body fat and fatty liver in middle-aged men

    Diabetologia

    (2007)
  • M.S. Cooper et al.

    11Beta-hydroxysteroid dehydrogenase type 1 and its role in the hypothalamus-pituitary-adrenal axis, metabolic syndrome, and inflammation

    J Clin Endocrinol Metab

    (2009)
  • J.W. Tomlinson et al.

    11beta-hydroxysteroid dehydrogenase type 1: a tissue-specific regulator of glucocorticoid response

    Endocr Rev

    (2004)
  • P. Anagnostis et al.

    Endocrine hypertension: diagnosis and management of a complex clinical entity

    Curr Vasc Pharmacol

    (2010)
  • P. Alberts et al.

    Selective inhibition of 11beta-hydroxysteroid dehydrogenase type 1 decreases blood glucose concentrations in hyperglycaemic mice

    Diabetologia

    (2002)
  • N.M. Morton et al.

    Novel adipose tissue-mediated resistance to diet-induced visceral obesity in 11 beta-hydroxysteroid dehydrogenase type 1-deficient mice

    Diabetes

    (2004)
  • Y. Kotelevtsev et al.

    11beta-hydroxysteroid dehydrogenase type 1 knockout mice show attenuated glucocorticoid-inducible responses and resist hyperglycemia on obesity or stress

    Proc Nat Acad Sci U S A

    (1997)
  • C.B. Whorwood et al.

    Regulation of glucocorticoid receptor alpha and beta isoforms and type I 11beta-hydroxysteroid dehydrogenase expression in human skeletal muscle cells: a key role in the pathogenesis of insulin resistance?

    J Clin Endocrinol Metab

    (2001)
  • B. Davan et al.

    Type 1 11beta-hydroxysteroid dehydrogenase mediates glucocorticoid activation and insulin release in pancreatic islets

    J Biol Chem

    (2000)
  • A. Swali et al.

    11beta-Hydroxysteroid dehydrogenase type 1 regulates insulin and glucagon secretion in pancreatic islets

    Diabetologia

    (2008)
  • H. Masuzaki et al.

    Transgenic amplification of glucocorticoid action in adipose tissue causes high blood pressure in mice

    J Clin Invest

    (2003)
  • H. Masuzaki et al.

    A transgenic model of visceral obesity and the metabolic syndrome

    Science

    (2001)
  • D.E. Livingstone et al.

    Understanding the role of glucocorticoids in obesity: tissue-specific alterations of corticosterone metabolism in obese Zucker rats

    Endocrinology

    (2000)
  • S.S. Prasad et al.

    A novel genetically-obese rat model with elevated 11 beta-hydroxysteroid dehydrogenase type 1 activity in subcutaneous adipose tissue

    Lipids Health Dis

    (2010)
  • H. Hauner et al.

    Promoting effect of glucocorticoids on the differentiation of human adipocyte precursor cells cultured in a chemically defined medium

    J Clin Invest

    (1989)
  • I.J. Bujalska et al.

    Differentiation of adipose stromal cells: the roles of glucocorticoids and 11β-hydroxysteroid dehydrogenase

    Endocrinology

    (1999)
  • R.S. Lindsay et al.

    Subcutaneous adipose 11beta-hydroxysteroid dehydrogenase type 1 activity and messenger ribonucleic acid levels are associated with adiposity and insulinemia in Pima Indians and Caucasians

    J Clin Endocrinol Metab

    (2003)
  • R.C. Andrews et al.

    Abnormal cortisol metabolism and tissue sensitivity to cortisol in patients with glucose intolerance

    J Clin Endocrinol Metab

    (2002)
  • M. Sjöstrand et al.

    Repeated measurements of 11β-HSD-1 activity in subcutaneous adipose tissue from lean, abdominally obese, and type 2 diabetes subjects—no change following a mixed meal

    Horm Metab Res

    (2010)
  • K. Kannisto et al.

    Overexpression of 11beta-hydroxysteroid dehydrogenase-1 in adipose tissue is associated with acquired obesity and features of insulin resistance: studies in young adult monozygotic twins

    J Clin Endocrinol Metab

    (2004)
  • S. Engeli et al.

    Regulation of 11beta-HSD genes in human adipose tissue: influence of central obesity and weight loss

    Obes Res

    (2004)
  • J.W. Tomlinson et al.

    Weight loss increases 11beta-hydroxysteroid dehydrogenase type 1 expression in human adipose tissue

    J Clin Endocrinol Metab

    (2004)
  • K. Simonyte et al.

    Weight loss after gastric bypass surgery in women is followed by a metabolically favorable decrease in 11beta-hydroxysteroid dehydrogenase 1 expression in subcutaneous adipose tissue

    J Clin Endocrinol Metab

    (2010)
  • Cited by (0)

    View full text