jdm

Journal of Diabetes & Metabolism

ISSN - 2155-6156

Review Article - (2015) Volume 6, Issue 4

Glycyrrhizic Acid as the Modulator of 11β -hydroxysteroid dehydrogenase (Type 1 and 2) in Rats under Different Physiological Conditions in Relation to the Metabolic Syndrome

Hui Ping Yaw1*, So Ha Ton1 and Khalid Abdul Kadir2
1School of Science, Monash University Malaysia, Malaysia
2School of Medicine and Health Sciences, Monash University Malaysia, Malaysia
*Corresponding Author: Hui Ping Yaw, School of Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 46150, Selangor Darul Ehsan, Malaysia, Tel: 60355146000 Email:

Abstract

Worldwide increase in the prevalence of metabolic syndrome has raised great attention to this disorder. Despite the effectiveness of the currently available therapeutic agents, most of the drugs elicit harmful side effects. Glycyrrhizic acid (GA) found in the licorice shrub, Glycyrrhiza glabra has been shown to exert anti-hyperglycaemic and antidyslipidaemic effects on rats under different physiological conditions via various mechanisms. The main route being the non-selective inhibition of 11β-hydroxysteroid dehydrogenase, an enzyme catalyzing the inter-conversion of active and inactive glucocorticoids. Altered intracellular glucocorticoid metabolism shows a stronger correlation to the development of metabolic syndrome compared to circulating glucocorticoid. Hence, this review focus on the role of GA in modulating glucocorticoid production in different tissues via regulation of 11β- hydroxysteroid dehydrogenase (both type 1 and 2) activities under different physiological conditions.

Keywords: Glycyrrhizic acid; Licorice shrub; Glucocorticoid

Introduction

Metabolic syndrome

Metabolic syndrome (MetS) is a cluster of risk factors including hyperglycaemia, dyslipidaemia, insulin resistance (IR), visceral obesity and hypertension of which an individual diagnosed with three out of five of the above factors will have increased susceptibility towards type 2 diabetes mellitus (T2DM) and cardiovascular diseases (CVD) [1]. Several studies have shown that the prevalence of MetS is increasing worldwide [2]. The adaptation of a sedentary lifestyle and increased consumption of high-calorie foods have contributed to this global epidemic [3]. The huge impact exerted by MetS on health and the economy has raised great public concern and attention not only towards understanding the pathogenesis of this disorder but also in identifying new and more effective therapeutic agents.

Glucocorticoids

Glucocorticoids (GC) is a group of hormones produced and secreted by the adrenal cortex [4]. The active and inactive form of GC in humans and rodents are cortisol and cortisone; corticosterone and 11-dehydrocorticosterone respectively [5]. GC is involved in various metabolic activities such as carbohydrate and lipid metabolism, regulation of blood pressure and control of the stress and inflammatory responses [6]. GC secretion is tightly regulated by the hypothalamopituitary- adrenal (HPA) axis following a circadian rhythm that is sensitive to light, sleep, stress and disease [6].

The main function of GC is to increase blood glucose level via multiple actions opposite to that of insulin [4]. GC stimulates lipolysis in the adipose tissues [7,8] and proteolysis in the skeletal muscles [9- 12]. These lead to increased release of glycerol, free fatty acids and amino acids which are transported to the liver for gluconeogenesis [13]. GC also reduces glucose uptake in the adipocytes [14,15] and muscle cells [16-18] and increase gluconeogenic enzymes such as phosphoenolpyruvate carboxykinase (PEPCK) [19] and glucose-6- phosphatase (G6Pase) activities [20]. Consequently, the increased glucose production promotes glycogenesis in the liver and since glycogen storage is limited, excess glucose is transported into the blood circulation. These result in hyperglycaemia and hyperinsulinaemia which could develop into T2DM.

Excessive GC production has been recognized as the primary contributor to MetS due to its phenotypic similarities with patients diagnosed with Cushing’s syndrome (characterized by elevated GC level and typical features of MetS) [21]. However, patients with MetS often have normal circulating GC level but with altered GC metabolism in GC targeted tissues such as the liver and adipose tissues [22-25]. This indicates that intracellularly generated GC may play a more significant role in MetS.

11β-hydroxysteroid dehydrogenase (11β-HSD)

11β-hydroxysteroid dehydrogenase (11β-HSD) is an enzyme that catalyzes the inter- conversion of active and inactive GC [4]. It determines the availability of GC for binding and activation of Glucocorticoid Receptors (GR). 11β-HSD belongs to the short-chain dehydrogenases/reductases (SDRs) superfamily [26]. There are two isoforms of 11β-HSD i.e. 11β-HSD type 1 (nicotinamide adenine dinucleotide (NAD) and nicotinamide adenine dinucleotide phosphate (NADPH)-dependent) and 11β-HSD type 2 (NAD-dependent) [5].

11β-HSD1, a bi-directional enzyme which can act as both a reductase (activate GC) and dehydrogenase (inactivate GC) is mainly found in the liver, adipose tissues and skeletal muscles. 11β-HSD2, a dehydrogenase involved in blood pressure regulation can be found in the mineralocorticoid (MC) target tissues e.g. kidney (Table 1) [5]. Aldosterone is the ligand for mineralocorticoid receptor (MR) in vivo. However, due to structure similarity, cortisol and aldosterone have equal affinities towards MR [27] and over-stimulation of MR promotes sodium reabsorption and leads to hypokalaemia and hypertension [4]. MR is protected from exposure to cortisol by 11β -HSD2 that metabolizes cortisol to the inactive form cortisone upon its production [5]. The important roles of 11β -HSD2 in the regulation of blood pressure potassium level were identified through studies where patients with apparent mineralocorticoid excess (AME) [28] and licoriceinduced [29] hypertension were found to have low renal 11β -HSD2 activity.

  11β-hydroxysteroid dehydrogenase type 1 11β-hydroxysteroid dehydrogenase type 2
Chromosomal location 1q32.2 16q22
Gene and size HSD11B1 30kb, 6 exons HSD11B2 6.2kb, 5 exons
Protein size 292 amino acids, 34kDa 405 amino acids, 444kDa
Enzyme reactions Bi-directional: In vivo: oxoreductase In vitro: dehydrogenase Dehydrogenase
Action Converts inactive GC into active GC Converts active GC into inactive GC
Co-factor(s) Oxoreductase: NADPH Dehydrogenase: NADP+ NAD+
Distribution Mainly expressed in the liver, lungs, pituitary, brains and adipose tissues Mainly expressed in the kidney, colon,salivary gland and placenta
Substrate affinity Low affinity (Km in μM) Corticosterone 1.8μM Cortisol 17μM 11-dehydrocorticosterone ~0.12μ High affinity (Km in μM) Corticosterone 5nM Cortisol 50μM 11-dehydrocorticosterone negligible
Function (s) Regulation of intracellular active GC level Protection of mineralocorticoid binding of active GC receptors from

Table 1: Comparisons between 11β-hydroxysteroid dehydrogenase type 1 and 2 (NAD-nicotinamide adenine dinucleotide; NADPH- nicotinamide adenine dinucleotide phosphate; GC- glucocorticoids) [5,105].

Glycyrrhizic acid

Glycyrrhizic acid (GA) is a triterpenoid compound found in the root extract of the licorice plant, Glycyrrhiza glabra (also known as ‘Gan Cao’ in chinese herbal medicine) [30]. Upon oral consumption, GA is deglucuroninated into glycyrrhetic acid (GE) by intestinal microflora [30]. GE then undergoes conjugation and generates glucuronide and sulfate conjugates which are secreted into the bile and then hydrolyzed by the intestinal microflora [31]. The products of hydrolysis are reabsorbed and this completes the enterohepatic circulation of GE followed by elimination of GA via faeces [31].

GA is widely used as a sweetener and aromatizer in the food industries. The ammoniated form of GA is used in the pharmaceutical industry such as for cough syrup production to mask the bitter taste of the medicine [32]. Therapeutic usage of GA can be traced back to 4000 years ago where the ancient Greeks and Romans used GA as treatment for respiratory-related diseases. In modern medicine, GA has been found to exert anti- oxidative [33], anti-inflammatory [34] and antitumorigenic [35] effects in various in vivo and in vitro models [30]. The potential role of GA against metabolic diseases was discovered when GA and its active metabolite, GE were found to be potent inhibitors of 11β-HSD [36,37]. An elevated 11β-HSD1 level is strongly correlated to increased risk of developing MetS [30].

GA has been shown to lower blood glucose and lipid levels in rats under different physiological conditions via various mechanisms particularly via non-selective inhibition of 11β-HSD [38-42]. Since GC is recognized as the main hormone that stimulates gluconeogenesis which leads to increased glucose release into the circulation [6], modification of GC production via modulation of 11β -HSD activities is expected to improve blood glucose level hence preventing the development of MetS. Hence, this review focus on the effects of GA in manipulating intracellular GC production via modulation of 11β -HSD activities specifically in the liver, kidney, subcutaneous adipose tissues (SAT), visceral adipose tissue (VAT), abdominal muscle (AM) and quadriceps femoris (QF) under different physiological conditions.

Effects of GA on 11β-HSD Activities

Lean rats

Intraperitoneal injection of GA at 50 mg/kg for 24 hours significantly lowered 11β- HSD1 activities in the liver, kidneys, SAT, AM and QF (p<0.05) and insignificantly in the VAT (p>0.05) [38]. However, only kidneys of the GA-treated rats showed significant reduction in 11β-HSD2 activities (p<0.05) while insignificant decrease were found in the livers, AM, QF, SAT and VAT (p>0.05). These were accompanied by a significant decrease in blood glucose levels and homeostatic model assessment of insulin resistance (HOMA-IR) index (p<0.05) and insignificant reduction in serum insulin levels (p>0.05).

The duration and route of administration may affect the ability of GA to reach various tissues. This could be due to the fact that intraperitoneal (IP) administration allows more of the GA to by-pass first-pass hepatic metabolism thus enabling more GA to reach the target tissues [31]. In another study where 50 mg/kg GA was administered to rats orally for 7 days, a significant reduction in 11β-HSD1 activities can only be observed in the livers (p<0.05) whereas an insignificant decrease was observed in the kidneys, AM, QF, SAT and VAT (p>0.05) [43]. GA-treated rats had significantly lower 11β-HSD2 activities in both the livers and kidneys (p<0.05) but insignificant in the AM, QF, SAT and VAT (p>0.05). Insignificant reduction of blood glucose, serum insulin and HOMA-IR index (p>0.05) were consistently found in the GA-treated rats.

Rats administered with 50 mg/kg GE (with reported inhibitory effects 200-100 times more potent than GA on 11β-HSD1 [31]) for 24 hours intraperitoneally had lower blood glucose concentrations (p<0.01), insignificantly higher serum insulin (p>0.05) and insignificantly lower HOMA-IR index (p>0.05). GE-treated rats had significantly lower 11β- HSD1 activities in the liver (p<0.05) than the controls whereas kidneys, SAT, VAT, AM and QF demonstrated insignificant decrease in 11β-HSD1 activities (p>0.05) [42]. Significantly higher 11β-HSD2 activities were found in both the kidneys and VAT (p<0.01) while lower activities were found in the liver, AM, QF and SAT (p>0.05) [44].

High-fat diet (HFD) (fats originated from animal or plant)

Over-consumption of high-fat diet increases circulatory lipids levels which in excess of energy expenditure will be stored as fats in the adipose tissues [45]. Accumulation of fats particularly in the visceral region is detrimental due to its proximity to the hepatic portal vein [46]. Increased expression of 11β-HSD1 in the adipose tissues has been associated with the development of visceral obesity and other risk factors of the MetS [47]. Rodent models with over-expression of 11β-HSD1 within adipocytes had been generated to examine the effects of tissue-specific GC activation [48,49]. The transgenic mice had elevated adipose corticosterone levels with increased mass of VAT. Metabolically, these animals exhibit both hyperglycaemia and IR with concomitant increase in serum free fatty acids (FFA) and triacylglycerol (TAG) [47,50]. 11β-HSD1 controls regional fat distribution and promotes visceral over subcutaneous adipose depot expansion [51]. VAT is particularly responsive to GC-induced adipogenesis and hypertrophy due to the presence of higher concentration of GR [52].

Rats fed on a HFD (animal-based) had significantly higher blood glucose concentrations (p<0.01) accompanied by elevated serum insulin concentrations and HOMA- IR index (p<0.05) [53]. All tissues of rats fed on a HFD showed higher 11β-HSD1 activities than the controls with significant increases in the liver and SAT (p<0.05); AM, QF and VAT (p<0.01) and insignificant increase in the kidneys (p>0.05). 11β-HSD2 activities were elevated significantly in the liver and kidneys (p<0.05), VAT (p<0.01) and insignificantly in the AM and QF (p>0.05). However, insignificant reduction of 11β-HSD2 activities was found in the kidneys (p>0.05). GA-treated HFD-fed rats had significantly lower blood glucose and serum insulin concentrations (p<0.01 and p>0.05 respectively) and HOMA-IR indices (p<0.05). These were accompanied by significantly lower 11β-HSD1 activities in the liver (p<0.05) and QF (p<0.01) while the kidneys, AM, SAT and VAT showed insignificant reduction (p>0.05) [53]. Similarly, all tissues of GA-treated rats showed reduction in the 11β-HSD2 activities with significant decreases in the liver, kidneys, SAT (p<0.01) and AM (p<0.05) and insignificant reduction in the QF and VAT (p>0.05) [53].

Rats fed on a HFD (plant-based) had elevated blood glucose concentrations (p>0.05) and significant increases in serum insulin (p<0.05) and HOMA-IR indices (p<0.01) [54]. The liver, AM, QF, SAT and VAT of these rats had significant increase in 11β-HSD1 activities (p<0.01) while insignificant reduction was observed in the kidneys (p>0.05). Significant increases in 11β-HSD2 activities were found in the SAT, AM and QF (p<0.05); and VAT (p>0.05) while reductions were found in the livers (p>0.05) and kidneys (p<0.01). HFD-fed rats given GA at 100mg/kg per day orally had significant decreases in blood glucose concentrations and HOMA-IR indices (p<0.05) and insignificantly lower serum insulin (p>0.05). These were accompanied by insignificant decrease in 11β-HSD1 activities in the kidneys, AM, QF, SAT and VAT (p>0.05) while significant reduction was found in the liver (p<0.05) [54]. Reduction in 11β-HSD2 activities were found in the QF (p<0.01) and AM and the livers (p>0.05). VAT and the kidneys demonstrated significant increase (p<0.01) while insignificant increase in the SAT (p>0.05).

High-sucrose diet

Refined carbohydrate constitutes the major part of a modern day diet [39]. Excessive sugar intake e.g. sucrose leads to increased blood glucose levels and since glycogen storage is limited, excess glucose will be converted into FFA and stored as TAG in the adipose tissues. Ectopic lipid storage especially in the visceral depots has been recognized as the primary contributor to MetS [55]. A diet high in sucrose content shows a direct link to elevated 11β- HSD1 activities which lead to increased active GC that has been associated with T2DM and CVD [56].

HSD-fed rats showed significant increases in 11β-HSD1 activities in the liver, kidneys, SAT, VAT (p<0.01); and AM (p<0.05) while insignificant increases were found in the QF (p>0.05) [39]. Significant increases in 11β-HSD2 activities were also found in the liver and VAT (p<0.01); AM and QF (p<0.05) while insignificant increases were found in the kidneys and SAT (p>0.05). The elevated 11β-HSD1 activities particularly in the liver contributed to the significant increase in blood glucose, serum insulin and HOMA-IR indices (p<0.05) [39].

HSD-fed rats given GA orally at 100 mg/kg per day showed significant reduction in 11β-HSD1 activities in the liver, SAT and VAT (p<0.01); kidneys, AM and QF (p<0.05). The kidneys; liver, VAT and AM demonstrated significant reduction in 11β-HSD2 activities (p<0.01 and p<0.05 respectively) while insignificant reduction was found in the SAT and QF (p>0.05). These contribute to the concomitant improvements in the blood glucose, HOMA-IR index (p<0.01); and serum insulin (p<0.05) [39].

High-calorie diet (HCD) and exposure to stress

Stress, together with over-consumption of high-calorie foods has been associated with the development of MetS [57]. GC is an important class of hormones which are elevated in response to stress [58]. Chronic over-secretion of GC leads to hyperglycaemia and hyperinsulinaemia thus causing visceral adiposity, hypertension and dyslipidaemia leading to MetS. Since 11β-HSD is involved in the regulation of GC, it would be expected that it will increase under stress to increase the active GC levels. However, stress induced by constant light exposure to 300-400 lux for 28 days on rats fed on a HCD did not have any effect on the 11β-HSD1 activities in the liver, kidneys, SAT, VAT and QF whereas a significant decrease was found in the AM (p<0.05). This may be related to adaptation towards stress [41]. Stress did not affect 11β-HSD2 activities in the kidneys, AM, QF, SAT and VAT but it did elicit a significant increase in the liver (p<0.05). Rats given GA orally at 100mg/kg did not show any difference in the blood glucose, serum insulin and HOMA-IR index (p>0.05) [41]. GA- treated rats demonstrated reduction in 11β-HSD1 activities in the kidneys (p<0.05); liver and SAT (p>0.05) but insignificant increases in the VAT, AM and QF (p>0.05). Reduction in11β-HSD2 activities (p<0.05) were found in the liver (p<0.05); kidneys and VAT (p>0.05) while insignificant increases were found in the SAT (p>0.05).

Adrenalectomized rats

The adrenal gland is the main site of hormones production involved in carbohydrate, fat and protein metabolism i.e. GC. Hence, dysfunction of the adrenal glands can lead to imbalances in the adrenal hormones and have been associated with the development of various diseased states particularly MetS [59]. Since excess GC production leads to diet- or stress-induced obesity due to excess energy intake [60], prevention of GC production by adrenalectomy is expected to reduce gluconeogenesis and food intake [61], slow body weight gain [62] and increases energy expenditure [63].

A study conducted by Ng et al., showed that adrenalectomized (ADX) rats had significant reduction in blood glucose levels (p<0.01) accompanied by insignificant increase in serum insulin (p>0.05) and insignificant decrease in HOMA-IR indices (p>0.05). Comparing the ADX and sham rats, the liver, kidneys, SAT, VAT and QF demonstrated insignificant decrease in 11β-HSD1 activities (p>0.05) while AM showed insignificant increase (p>0.05). ADX rats had lower 11β-HSD2 activities in the livers and AM (p<0.05); kidneys, QF, SAT and VAT (p>0.05) [64]. GC, glucagon and adrenaline are the main hormones that stimulate gluconeogenesis and glycogenolysis which contribute to elevated blood glucose level in the postabsorptive stage [65,66]. In the ADX rats, there was decreased production of the above hormones following removal of adrenal glands which contribute to reduced gluconeogenic enzymes activities and glycolysis that contribute to an overall lower blood glucose level.

Administration of GA orally at 100 mg/kg to the ADX rats restored the blood glucose, serum insulin levels and HOMA-IR indices of the ADX rats to a level similar to that of the sham rats (p>0.05). GA-treated ADX rats had significant reductions in 11β-HSD1 activities only in the QF (p<0.05) while insignificant decrease in the liver, kidneys, SAT, VAT and AM (p>0.05). GA-treated ADX rats had significantly higher 11β-HSD2 activities than the ADX rats only in the liver (p<0.05) while the kidneys, AM, QF, SAT and VAT demonstrated insignificant increase (p>0.05) [64].

Discussion

11β -HSD1 activities in the:

Liver: High-calorie diet (HCD) has been recognized as the main contributor to various metabolic diseases. Stress, for example, originating from workplace has also been shown to exhibit strong correlation with the development of MetS [67]. Modulation of 11β-HSD activities under different physiological conditions (Table 2) have been shown to lower blood glucose and lipid levels with concomitant improvement in insulin sensitivity thus alleviating the development of MetS.

Treatment Treatment period Route of administration Compound Concentration (mg/kg) 11β-HSD1 activities 11β-HSD2 activities Refs.
Tissues Tissues
Liver Kidney AM QF SAT VAT Liver Kidney AM QF SAT VAT
Leanrats 24 hours IP GA 50 * * * * * * [38]
7days Oral GA 50 * * * [43]
24 hours IP GE 50 * ** ** [42]
High-fat diet
(animal-based)
28-days Oral GA 100 * ** ** ** * ** [53]
High-fat diet
(plant-based)
28-days Oral GA 100 * * [54]
High-sucrosediet 28-days Oral GA 100 ** * * * ** ** * ** * * [39]
High-fat+high- sucrose+stress 28-days Oral GA 100 * * Not available Not available [41]
Adrenalectomized 28-days Oral GA 100 * * * * * [40]

Table 2: 11β-hydroxysteroid dehydrogenase type 1 and 2 (11β -HSD1 and 2) activities in different tissues under different physiological conditions (IP- intraperitoneal injection; AM: Abdominal muscle; QF- quadriceps femoris; SAT- subcutaneous adipose tissues; VAT- visceral adipose tissues; * and ** indicate significant changes with p-value of 0.05 and 0.01 respectively).

Elevated 11β-HSD1 activities particularly in the liver are deleterious as transgenic mice with 11β-HSD1 over-expression develop IR, hyperglycaemia, hepatic steatosis and dyslipidaemia via different mechanisms [68]. Morbidly obese patients with MetS also showed significantly higher hepatic 11β -HSD1, PEPCK, hexose-6-hosphate dehydrogenase and GR expressions than the obese counterparts without MetS [69]. Rats fed on a HCD (high in fat or sugar content) showed elevated blood glucose, serum insulin and circulating TAG levels accompanied by reduced insulin sensitivity with elevated hepatic 11β-HSD1 activities [39,53,70]. Increased 11β-HSD1 activities in the liver promote hepatic gluconeogenesis via induction of PEPCK and G6Pase activities [71]. This leads to elevated circulating blood glucose levels and in the long-run may develop into hyperglycaemia and subsequently IR. GC also increases fatty acids synthesis via increased activities of fatty acid synthase and acetyl-CoA carboxylase. This will lead to increased very-low-density lipoprotein (VLDL) production and triacylglycerol (TAG) synthesis [72]. These events contribute to elevated circulating lipid levels leading to dyslipidaemia. Furthermore, GC decreases β-oxidation of FFA by interfering with acyl-CoA dehydrogenase activities. This subsequently leads to TAG accumulation in the liver resulting in pathogenic fatty liver [73].

GA-treated HCD-fed rats showed significant reduction in their hepatic 11β-HSD1 activities with concomitant improvements in their blood glucose and TAG levels and insulin sensitivity [39,53,70]. These could be associated with improved lipid metabolism and reduced gluconeogenesis. 11β-HSD1 null mice have lowered TAG levels accompanied by an overall increase in HDL-cholesterol which can be associated with increased expressions of genes involved in fat metabolism such as the fatty-acid binding protein [74] and oxidative enzymes including carnitine-palmitoyl transferase 1 (CPT-1), acyl-CoA oxidase and uncoupling protein 2 (UCP2) [73,75]. Obese or diabetic mice with 11β-HSD1 inhibition or deficiency have lower fasting blood glucose levels with concomitant reduced GC action particularly gluconeogenesis in the liver as indicated by the lower hepatic PEPCK and G6Pase activities [36,76-78]. Hence, inhibition of 11β-HSD1 by GA in the liver of rats fed on a HCD may decrease the active GC levels in the hepatocytes and promote TAG uptake by the oxidative tissues e.g. liver and skeletal muscles followed by increased lipid oxidation. Hence, via inhibition of hepatic 11β-HSD1, GA improves dyslipidaemia and hyperglycaemia via inhibition of gluconeogenesis and reduction of VLDL secretion as well as TAG accumulation.

Adipose tissues: Adipose tissues act as the main energy storage site in the body system [79]. GC control lipid metabolism in the adipose tissues by regulating the breakdown and synthesis of fatty acids depending on the body’s energy status. During fasting, GC promote lipid mobilization to increase FFA release into the circulation via inhibition of lipogenesis [5] and reduction of the rate of glyceroneogenesis and fatty acid re-esterification [80,81]. These anti- lipogenic effects of GC increase the release of FFA into the circulation as an energy source to the body system. During satiety, GC promotes lipogenesis and lipid uptake into the adipose tissues for storage in response to insulin [25]. Dysregulated 11β-HSD1 level has been found in the adipose tissues of various rodent models of obesity and diabetes. For example, an elevated 11β-HSD1 level was found in the visceral adipose tissues (VAT) of obese Zucker rats [82]. 11β-HSD1(-/-) mice were shown to have a beneficial metabolic phenotype. These include resistance towards diet-induced obesity, storage of fat in the subcutaneous rather than the visceral region and improvement in glucose level and insulin sensitivity with lower circulating FFA level [78,83]. In human studies, increase in adipose 11β -HSD1 activity has been constantly found in both subcutaneous and visceral adipose tissues of obese and/or insulinresistant individuals and shows strong correlation to the development of MetS [52,84-89]. HCD-fed rats had elevated 11β-HSD1 activities in their adipose tissues [39,53,70] which can be attributed to the elevated blood glucose concentration that causes an increase in the level of glucose-6-phosphate level, a substrate for hexose-6-phosphate dehydrogenase. This is followed by increased NADPH supply in the adipose tissues that promote oxo-reductase activities of 11β-HSD1 thus increasing the active GC concentration [90].

However, among the GA-treated HCD-fed rats, only rats fed on a high-sucrose diet had significant reduction in their 11β-HSD1 activities in both the subcutaneous and visceral adipose tissues [39]. This is accompanied by significant reduction in blood glucose and improved insulin sensitivity. Indeed, mice with site-specific 11β-HSD1 deletion in the adipose tissues had lower fasting blood glucose level with concomitant insulin sensitization [78]. Furthermore, 11β-HSD1- deficient mice fed on a HFD showed preferred accumulation of fat in the subcutaneous tissues while promoting fat loss from the visceral depots [83]. These beneficial effects conferred upon 11β-HSD1 inhibition had been associated with lower active GC levels in the adipose tissues which was supported by mice with transgenic over- expression of 11β-HSD2 (inactivates active corticosterone) that demonstrated improved insulin sensitivity and were protected from obesity [91]. PEPCK activities are induced by active GC in the liver and kidneys but being suppressed in the adipose tissues [80]. Thus, inhibition of 11β-HSD1 by GA reduces active GC that inhibit PEPCK in the adipose tissues.

This promotes glyceroneogenesis which is the process of reesterification of unused FFA in the circulation with glycerol to regenerate TAG [81,92]. This is important as mice with elevated fat depots without elevated circulating FFA levels do not develop IR or T2DM [93]. Hence induction of PEPCK by inhibiting GC is important in reducing circulating FFA levels thereby preventing the development of T2DM.

Skeletal muscles: Skeletal muscle is the major site of action of insulin and carbohydrate and lipid metabolism [94]. 11β -HSD1 is expressed in the skeletal muscle at a lower level than liver whereas 11β -HSD2 is absent from the muscles [95,96]. Although less is known about the role of 11β -HSD1 in glucose-insulin homeostasis in the muscles compared to liver and adipose tissues, there are indications that skeletal muscle 11β -HSD1 activity may play a significant role in the development of MetS. For example, rat model of T2DM demonstrated increased 11β -HSD1 activities in the gastronecmius muscle [97] while obese individuals with T2DM were found to ha ve increased 11β-HSD1 expression in the myotubes compared to the BMI-matched controls [96,98]. Elevated 11β-HSD1 levels were proposed to have caused IR in the skeletal muscles via different pathways. The increased concentration of active GC interferes with the insulin signalling pathway by promoting inhibitory phosphorylation of insulin receptor substrate-1(IRS-1) and inhibiting the translocation of GLUT-4 to the surface membrane. Both IRS-1 and GLUT-4 are the essential components of the insulinmediated glucose uptake mechanism [16,17]. GC also inhibit glycogen synthesis via inhibition of dephosphorylation (hence the activation) of glycogen synthase [99,100]. IP administration of 50 mg/kg GA to rats fed on standard rat chow for 24 hours reduced 11β-HSD1 activities in the AM and QF significantly [38] hence is expected to improve insulin sensitivity. Similar findings were also found in HSD-fed rats [39].

Furthermore, administration of GA at 100 mg/kg lowered 11β-HSD1 in the QF of adrenalectomized rats [40]. GC increase the rate of gluconeogenesis in the skeletal muscles hence stimulates proteolysis and inhibits protein synthesis in the skeletal muscles and provides increased gluconeogenic substrates to the liver [6]. Inhibition of 11β-HSD1 in the skeletal muscles of diabetic mice promote fatty acids uptake and oxidation via increased expression of the related genes e.g. carnithine palmitoyltransferase 1 and acyl-CoA oxidase [73]. These events prevent the development of dyslipidaemia and FFA induced IR in the skeletal muscles. The summary of actions of GA on 11β-HSD1 in different tissues is shown in Figure 1.

diabetes-metabolism-ameliorating-MetS-modulation

Figure 1: Proposed actions of GA in ameliorating MetS via modulation of GC in different tissues through inhibition of 11β-HSD1. [11β-HSD: 11beta-hydroxysteroid dehydrogenase; CVD: cardiovascular disease; G6Pase: glucose-6-phosphatase; GA: Glycyrrhizic acid; GC: glucocorticoids; H6PDH: hexose-6-phopshate dehydrogenase; IR: insulin resistance; PEPCK: phosphoenolpyruvate carboxykinase; MetS: metabolic syndrome; T2DM: Type 2 diabetes mellitus].

11β -HSD2 activities

Administration of GA orally at 50 mg/kg for 24 hours or 7 days consistently lowered 11β-HSD2 levels in the kidneys of normal Rattus norvegicus [38]. The kidney has high concentration of MR in which GC have a high-affinity to. Thus, a high expression of 11β- HSD2 is found in the kidney to prevent over-stimulation of the MR [5] thereby exerting protective effects against hypermineralocorticoid effects e.g. hypertension [101]. 11β-HSD2 converts active GC into inactive GC. Thus, elevated 11β-HSD2 activities in the kidney increases circulating inactive GC which could be activated by 11β-HSD1 in the hepatocytes and adipocytes [102,103]. The increased 11β-HSD1 activities promote lipolysis in the adipose tissues particularly the VAT. The FFA released from the VAT is then released into the hepatic portal vein and promotes IR in the liver [104]. This forms the vicious cycle of increased FFA supply and hepatic IR. Inhibition of 11β-HSD2 in the kidney has been associated with hypertension, hypokalaemia and hypernatraemia due to over-stimulation of MR. However, this effect was not seen in rats given GA orally at a dosage of 50 mg/kg for 7 days as the rats had sodium and potassium levels and systolic blood pressure comparable to the controls. Hence, GA is able to reduce renal 11β-HSD2 activities without inducing hypertension.

Conclusion

Under different physiological conditions, GA administration was found to lower 11β- HSD1 and 2 activities with effects found mainly in the liver and kidneys. These were accompanied by reduced blood glucose levels, serum insulin and improved insulin sensitivity. GA, via inhibition of 11β-HSD1 also improves lipid profiles and prevents ectopic lipid storage particularly in the liver and VAT. All these contribute to improved blood glucose levels and insulin sensitivity which makes GA a potential therapeutic compound for MetS.

References

  1. Alberti KG, Eckel RH, Grundy SM, Zimmet PZ, Cleeman JI, et al. (2009) Harmonizing the metabolic syndrome. A joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; and International Association for the Study of Obesity. Circulation120: 1640-1645.
  2. Sommer P, Sweeney G (2010) Functional and mechanistic integration of infection and the metabolic syndrome. SKorean Diabetes J 34: 71-76.
  3. Mohamud WN, Ismail AA, Sharifuddin A, Ismail IS, Musa KI, et al. (2011) Prevalence of metabolic syndrome and its risk factors in adult Malaysians: results of a nationwide survey. SDiabetes Res ClinPract 91: 239-245.
  4. Seckl JR1 (2004) 11beta-hydroxysteroid dehydrogenases: changing glucocorticoid action. SCurrOpinPharmacol 4: 597-602.
  5. Draper N, Stewart PM (2005) 11beta-hydroxysteroid dehydrogenase and the pre-receptor regulation of corticosteroid hormone action. SJ Endocrinol 186: 251-271.
  6. Vegiopoulos A,Herzig S (2007) Glucocorticoids, metabolism and metabolic diseases. SMol Cell Endocrinol 275: 43-61.
  7. Xu C, He J, Jiang H, Zu L, Zhai W, et al. (2009) Direct effect of glucocorticoids on lipolysis in adipocytes. SMolEndocrinol 23: 1161-1170.
  8. Campbell JE,Peckett AJ, D'souza AM, Hawke TJ, Riddell MC (2011) Adipogenic and lipolytic effects of chronic glucocorticoid exposure. SAm J Physiol Cell Physiol 300: C198-209.
  9. Auclair D,Garrel DR, ChaoukiZerouala A, Ferland LH (1997) Activation of the ubiquitin pathway in rat skeletal muscle by catabolic doses of glucocorticoids. SAm J Physiol 272: C1007-1016.
  10. Baehr LM,Furlow JD, Bodine SC (2011) Muscle sparing in muscle RING finger 1 null mice: response to synthetic glucocorticoids. SJ Physiol 589: 4759-4776.
  11. Hu Z, Wang H, Lee IH, Du J, Mitch WE (2009) Endogenous glucocorticoids and impaired insulin signaling are both required to stimulate muscle wasting under pathophysiological conditions in mice. Journal of Clinical Investigation, 119: 3059-3069.
  12. Shimizu N, Yoshikawa N, Ito N, Maruyama T, Suzuki Y, et al. (2011) Crosstalk between glucocorticoid receptor and nutritional sensor mTOR in skeletal muscle. SCellMetab 13: 170-182.
  13. Hadley M, Levine J (2007) Adrenal Steroid Hormones. Endocrinology.Pearson Prentice Hall. New Jersey, USA.
  14. Fain J, Scow R, Chernick S (1963) Effects of glucocorticoids on metabolism of adipose tissue in Vitro. J BiolChem 238: 54-58.
  15. Blair SC,Caterson ID, Cooney GJ (1995) Glucocorticoid deprivation alters in vivo glucose uptake by muscle and adipose tissues of GTG-obese mice. SAm J Physiol 269: E927-933.
  16. Dimitriadis G, Leighton B, Parry-Billings M, Sasson S, Young M, et al. (1997) Effects of glucocorticoid excess on the sensitivity of glucose transport and metabolism to insulin in rat skeletal muscle. SBiochem J 321 : 707-712.
  17. Morgan SA, Sherlock M, Gathercole LL, Lavery GG, Lenaghan C, et al. (2009) 11beta-hydroxysteroid dehydrogenase type 1 regulates glucocorticoid-induced insulin resistance in skeletal muscle. SDiabetes 58: 2506-2515.
  18. Weinstein SP, Wilson CM, Pritsker A, Cushman SW (1998) Dexamethasone inhibits insulin-stimulated recruitment of GLUT4 to the cell surface in rat skeletal muscle. SMetabolism 47: 3-6.
  19. Stafford JM, Waltner-Law M, Granner DK (2001) Role of accessory factors and steroid receptor coactivator 1 in the regulation of phosphoenolpyruvatecarboxykinase gene transcription by glucocorticoids. J BiolChem 276: 3811-3819.
  20. Vander Kooi BT,Onuma H, Oeser JK, Svitek CA, Allen SR, et al. (2005) The glucose-6-phosphatase catalytic subunit gene promoter contains both positive and negative glucocorticoids response elements. MolEndocrinol19: 3001-3022.
  21. Walker B,Seckel J (2001) International Textbook of Obesity, 200, John Wiley and Sons: Chichester, UK. p. 198-204.
  22. Mårin P, Darin N, Amemiya T, Andersson B, Jern S, et al. (1992) Cortisol secretion in relation to body fat distribution in obese premenopausal women. SMetabolism 41: 882-886.
  23. Fraser R, Ingram MC, Anderson NH, Morrison C, Davies E, et al. (1999) Cortisol effects on body mass, blood pressure, and cholesterol in the general population. SHypertension 33: 1364-1368.
  24. Gathercole LL,Lavery GG, Morgan SA, Cooper MS, Sinclair AJ, et al. (2013) 11β-Hydroxysteroid dehydrogenase 1: translational and therapeutic aspects. SEndocr Rev 34: 525-555.
  25. Gathercole LL, Morgan SA, Bujalska IJ, Hauton D, Stewart PM, et al. (2011) Regulation of lipogenesis by glucocorticoids and insulin in human adipose tissue. PLoS One 6: e26223.
  26. Jörnvall H,Persson B, Krook M, Atrian S, Gonzàlez-Duarte R, et al. (1995) Short-chain dehydrogenases/reductases (SDR). SBiochemistry 34: 6003-6013.
  27. Arriza JL,Simerly RB, Swanson LW, Evans RM (1988) The neuronal mineralocorticoid receptor as a mediator of glucocorticoid response. SNeuron 1: 887-900.
  28. Ulick S, Levine LS, Gunczler P, Zanconato G, Ramirez LC, et al. (1979) A syndrome of apparent mineralocorticoid excess associated with defects in the peripheral metabolism of cortisol. SJ ClinEndocrinolMetab 49: 757-764.
  29. Stewart PM, Wallace AM, Valentino R, Burt D, Shackleton CH, et al. (1987) Mineralocorticoid activity of liquorice: 11-beta-hydroxysteroid dehydrogenase deficiency comes of age. SLancet 2: 821-824.
  30. Isbrucker RA, Burdock GA (2006) Risk and safety assessment on the consumption of Licorice root (Glycyrrhiza sp.), its extract and powder as a food ingredient, with emphasis on the pharmacology and toxicology of glycyrrhizin. SRegulToxicolPharmacol 46: 167-192.
  31. Ploeger BA, Meulenbelt J, DeJongh J (2000) Physiologically Based Pharmacokinetic Modeling of Glycyrrhizic acid, a Compound Subject to presystemic Metabolism and Enterohepatic Cycling. ToxicolApplPharmacol 162: 177- 188.
  32. Størmer FC,Reistad R, Alexander J (1993) Glycyrrhizic acid in liquorice--evaluation of health hazard. SFoodChemToxicol 31: 303-312.
  33. Li XL, Zhou AG, Zhang L, Chen WJ (2011) Antioxidant status and immune activity of glycyrrhizin in allergic rhinitis mice. SInt J MolSci 12: 905-916.
  34. Honda H, Nagai Y, Matsunaga T, Saitoh S, Akashi-Takamura S, et al. (2012) Glycyrrhizin and isoliquiritigenin suppress the LPS sensor toll-like receptor 4/MD-2 complex signaling in a different manner. SJ LeukocBiol 91: 967-976.
  35. Paolini M,Barillari J, Broccoli M, Pozzetti L, Perocco P, et al. (1999) Effect of liquorice and glycyrrhizin on rat liver carcinogen metabolizing enzymes. SCancerLett 145: 35-42.
  36. Alberts P,Engblom L, Edling N, Forsgren M, Klingström G, et al. (2002) Selective inhibition of 11beta-hydroxysteroid dehydrogenase type 1 decreases blood glucose concentrations in hyperglycaemic mice. SDiabetologia 45: 1528-1532.
  37. Wamil M,Seckl JR (2007) Inhibition of 11beta-hydroxysteroid dehydrogenase type 1 as a promising therapeutic target. SDrugDiscov Today 12: 504-520.
  38. Chia YY, Yin YY, Ton SH, Kadir KB (2009) Effects of Glycyrrhizic acid on 11ß-Hydroxysteroid Dehydrogenase (11ßHSD1 and 2) Activities and HOMA-IR in Rats at Different Treatment Periods. Experimental Clinical Endocrinology Diabetes118: 617-624.
  39. Chandramouli C,Ting YS, Lyn LY, Ha TS, Kadir KA (2011)Glycyrrhizic Acid Improves Lipid and Glucose Metabolism in High-Sucrose-Fed Rats. J EndocrinolMetab 1: 125-141.
  40. Ng YQ, Chandramouli C, Ton SH, Kadir KA, Haque F, et al. (2012) Modulation of glucose and lipid metabolism in adrenalectomised rats given glycyrrhizic acid. J MolPathophysiol 1: 3-20.
  41. Fernando HA, Chin HF, Ton SH, Abdul Kadir K (2013) Stress and Its Effects on Glucose Metabolism and 11β-HSD Activities in Rats Fed on a Combination of High-Fat and High-Sucrose Diet with Glycyrrhizic Acid. SJ Diabetes Res 2013: 190395.
  42. Yaw HP, Ton SH, Kadir KA, Tan YT, Teo YW, et al., (2013) Effects of Glycyrrhetic Acid (GE) on Some Gluconeogenic Enzymes, Lipoprotein Lipase and Peroxisome Proliferator-Activated Receptors Alpha and Gamma. The Open Bioactive Compounds Journal 4: 14-24.
  43. Choh L (2008) 11ß-Hydroxysteroid Dehydrogenase Type 1 and 2 and HOMA-IR in Orally- adminsiteredGlycycrrhizic Acid, in School of Science.Monash University Malaysia: Malaysia.
  44. Yohanes M (2010) Effect of glycrrhetic acid with on 11beta-hydroxysteroid dehydrogenase type 1 and 2 in rats, in School of Science2010, Monash University Malaysia: Malaysia.
  45. Bergman RN,Ader M (2000) Free fatty acids and pathogenesis of type 2 diabetes mellitus. STrendsEndocrinolMetab 11: 351-356.
  46. Perrini S,Leonardini A, Laviola L, Giorgino F (2008) Biological specificity of visceral adipose tissue and therapeutic intervention. SArchPhysiolBiochem 114: 277-286.
  47. Masuzaki H, Paterson J, Shinyama H, Morton NM, Mullins JJ, et al. (2001) A transgenic model of visceral obesity and the metabolic syndrome. SScience 294: 2166-2170.
  48. Maeda K, Cao H, Kono K, Gorgun CZ, Furuhashi M, et al. (2005) Adipocyte/macrophage fatty acid binding proteins control integrated metabolic responses in obesity and diabetes. Cellular Metabolism 1: 107-119.
  49. Paterson JM,Seckl JR, Mullins JJ (2005) Genetic manipulation of 11beta-hydroxysteroid dehydrogenases in mice. SAm J PhysiolRegulIntegr Comp Physiol 289: R642-652.
  50. Masuzaki H, Yamamoto H, Kenyon CJ, Elmquist JK, Morton NM, et al. (2003) Transgenic amplification of glucocorticoid action in adipose tissue causes high blood pressure in mice. SJ Clin Invest 112: 83-90.
  51. De Sousa Peixoto RA, Turban S, Battle JH, Chapman KE, Seckl JR, et al. (2008) Preadipocyte 11beta-hydroxysteroid dehydrogenase type 1 is a keto-reductase and contributes to diet-induced visceral obesity in vivo. SEndocrinology 149: 1861-1868.
  52. Paulsen SK, Pedersen SB, Fisker S, Richelsen B (2007) 11Beta-HSD type 1 expression in human adipose tissue: impact of gender, obesity, and fat localization. SObesity (Silver Spring) 15: 1954-1960.
  53. Lai H(2010) 11 beta-hydroxysteroid dehydrogenase type 1 and 2 activities in rats fed on high animal fat diet with glycyrrhizica acid, in School of Science2010, Monash University Malaysia: Malaysia. p. 99.
  54. Lim AWK (2009) 11 beta-hydroxysteroid dehydrogenase type 1 and 2 in orally administered glycyrrhizic acid-treated rats fed on high-fat diet, in School of Science2009, Monash University Malaysia: Malaysia. p. 121.
  55. Bergman RN, Kim SP, Catalano KJ, Hsu IR, Chiu JD, et al. (2006) Why visceral fat is bad: mechanisms of the metabolic syndrome. SObesity (Silver Spring) 14 Suppl 1: 16S-19S.
  56. London E,Castonguay TW (2009) Diet and the role of 11beta-hydroxysteroid dehydrogenase-1 on obesity. SJ NutrBiochem 20: 485-493.
  57. Chrousos GP1 (2009) Stress and disorders of the stress system. SNat Rev Endocrinol 5: 374-381.
  58. Stinnett G, Seasholtz A (2010)Encyclopedia of Behavioral Neuroscience. Stress and emotionality2010: Elsevier, USA.
  59. Kalady MF,McKinlay R, Olson JA Jr, Pinheiro J, Lagoo S, et al. (2004) Laparoscopic adrenalectomy for pheochromocytoma. A comparison to aldosteronoma and incidentaloma. SSurgEndosc 18: 621-625.
  60. Coccurello R, D'Amato FR, Moles A (2009) Chronic social stress, hedonism and vulnerability to obesity: lessons from rodents. SNeurosciBiobehav Rev 33: 537-550.
  61. Saito M, Bray GA (1984) Adrenalectomy and food restriction in the genetically obese (ob/ob) mouse. SAm J Physiol 246: R20-25.
  62. Chavez M, Seeley RJ, Green PK, Wilkinson CW, Schwartz MW, et al. (1997) Adrenalectomy increases sensitivity to central insulin. SPhysiolBehav 62: 631-634.
  63. Thurlby P, Trayhurn P (1980) Regional blood flow in genetically obese (ob/ob) mice. the importance of brown adipose tissue to reduced energy expenditure on non- shivering thermogenesis. PfluegersArchieve385: 193-201.
  64. Tamotharan M (2010) 11 beta-hydroxysteroid dehydrogenase type 1 and 2 in adrenalectomized rats, in School of Science2010, Monash University Malaysia: Malaysia. p. 101.
  65. Kodama T, Hori SH (1982) Possible functional coupling of hexose-6-phosphate dehydrogenase to microsomal electron transport system in rat kidney and liver. SBiochimBiophysActa 715: 151-161.
  66. Weltan SM, Bosch AN, Dennis SC, Noakes TD (1998) Influence of muscle glycogen content on metabolic regulation. SAm J Physiol 274: E72-82.
  67. Chandola T, Brunner E, Marmot M (2006) Chronic stress at work and the metabolic syndrome: prospective study. SBMJ 332: 521-525.
  68. Liu Y, Nakagawa Y, Wang Y, Sakurai R, Tripathi PV, et al. (2005) Increased glucocorticoid receptor and 11{beta}-hydroxysteroid dehydrogenase type 1 expression in hepatocytes may contribute to the phenotype of type 2 diabetes in db/db mice. SDiabetes 54: 32-40.
  69. Torrecilla E,Fernández-Vázquez G, Vicent D, Sánchez-Franco F, Barabash A, Cabrerizo L, et al. (2012) Liver upregulation of genes involved in cortisol production and action is associated with metabolic syndrome in morbidly obese patients. Obesity Surgery 22: 478-486.
  70. Eu CH, Lim WY, Ton SH, bin Abdul Kadir K (2010) Glycyrrhizic acid improved lipoprotein lipase expression, insulin sensitivity, serum lipid and lipid deposition in high-fat diet-induced obese rats. SLipids Health Dis 9: 81.
  71. Cassuto H,Kochan K, Chakravarty K, Cohen H, Blum B, et al. (2005) Glucocorticoids regulate transcription of the gene for hosphoenolpyruvatecarboxykinase in the liver via an extended glucocorticoid regulatory unit.J BiolChem 280:33873-33884.
  72. Nuotio-Antar AM,Hachey DL, Hasty AH (2007) Carbenoxolone treatment attenuates symptoms of metabolic syndrome and atherogenesis in obese, hyperlipidemic mice. SAm J PhysiolEndocrinolMetab 293: E1517-1528.
  73. Berthiaume M,Laplante M, Festuccia WT, Cianflone K, Turcotte LP, et al.(2007) 11ß-HSD1 inhibition improves triglyceridemia through reduced liver VLDL secretion and partitions lipids toward oxidative tissues. Am J PhysiolEndocrinolMetab 293: E1045-E1052.
  74. Berthiaume M,Laplante M, Festuccia WT, Berger JP, Thieringer R, et al. (2010) Preliminary report: pharmacologic 11beta-hydroxysteroid dehydrogenase type 1 inhibition increases hepatic fat oxidation in vivo and expression of related genes in rats fed an obesogenic diet. SMetabolism 59: 114-117.
  75. Morton NM, Holmes MC, Fiévet C, Staels B, Tailleux A, et al. (2001) Improved lipid and lipoprotein profile, hepatic insulin sensitivity, and glucose tolerance in 11beta-hydroxysteroid dehydrogenase type 1 null mice. SJ BiolChem 276: 41293-41300.
  76. Alberts P,Rönquist-Nii Y, Larsson C, Klingström G, Engblom L, et al., (2005) Effect of high-fat diet on KKAy and ob/ob mouse liver and adipose tissue corticosterone and 11-dehydrocorticosterone concentrations. Hormone Metabolic Research37: 402-407.
  77. Alberts P, Nilsson C, Selen G, Engblom LO, Edling NH, et al. (2003) Selective inhibition of 11 beta-hydroxysteroid dehydrogenase type 1 improves hepatic insulin sensitivity in hyperglycemic mice strains. SEndocrinology 144: 4755-4762.
  78. Kotelevtsev, Holmes MC, Burchell A, Houston PM, Schmoll D, et al. (1997) 11ß-Hydroxysteroid dehydrogenase type 1 knockout mice show attenuated glucocorticoid-inducible responses and resist hyperglycemia on obesity or stress. ProcNatlAcadSci U S A94: 14924-14929.
  79. Ronti T,Lupattelli G, Mannarino E (2006) The endocrine function of adipose tissue: an update. SClinEndocrinol (Oxf) 64: 355-365.
  80. Nechushtan H, Benvenisty N, Brandeis R, Reshef L (1987) Glucocorticoids control phosphoenolpyruvatecarboxykinase gene expression in a tissue specific manner. SNucleic Acids Res 15: 6405-6417.
  81. Olswang Y, Blum B, Cassuto H, Cohen H, Biberman Y (2003) Glucocorticoids repress transcription of phosphoenolpyruvatecarboxykinase (GTP) gene in adipocytes by inhibiting its C/EBP-mediated activation. Journal of Biological Chemistry278: 12929-12936.
  82. Livingstone DE, Jones GC, Smith K, Jamieson PM, Andrew R, et al. (2000) Understanding the role of glucocorticoids in obesity: tissue-specific alterations of corticosterone metabolism in obese Zucker rats. Endocrinology. 141: 560-543.
  83. Morton NM, Paterson JM, Masuzaki H, Holmes MC, Staels B, et al. (2004) Novel adipose tissue-mediated resistance to diet-induced visceral obesity in 11 beta-hydroxysteroid dehydrogenase type 1-deficient mice. SDiabetes 53: 931-938.
  84. Veilleux A, Rhéaume C, Daris M, Luu-The V, Tchernof A (209) Omental adipose tissue type 1 11ß-hydroxysteroid dehydrogenase oxoreductase activity, body fat distribution, and metabolic alterations in women. Journal of Clinical Endocrinology and Metabolism. 94: 3550-3557.
  85. Engeli S,Böhnke J, Feldpausch M, Gorzelniak K, Heintze U, et al. (2004) Regulation of 11beta-HSD genes in human adipose tissue: influence of central obesity and weight loss. SObes Res 12: 9-17.
  86. Kannisto K,Pietiläinen KH, Ehrenborg E, Rissanen A, Kaprio J, et al. (2004) 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. SJ ClinEndocrinolMetab 89: 4414-4421.
  87. Lindsay R, Wake DJ, Nair S, Bunt J, Livingstone DEW, et al. (2003)Subcutaneous adipose 11ß-hydroxysteroid dehydrogenase type 1 activity and messenger ribonucleic acid levels are associated with adiposity and insulinemia in Pima Indians and Caucasians. Journal of Clinical Endocrinology and Metabolism 88:2738-2744.
  88. Paulmyer-Lacroix O,Boullu S, Oliver C, Alessi MC, Grino M (2002) Expression of the mRNA coding for 11beta-hydroxysteroid dehydrogenase type 1 in adipose tissue from obese patients: an in situ hybridization study. SJ ClinEndocrinolMetab 87: 2701-2705.
  89. Desbriere R,Vuaroqueaux V, Achard V, Boullu-Ciocca S, Labuhn M, et al. (2006) 11beta-hydroxysteroid dehydrogenase type 1 mRNA is increased in both visceral and subcutaneous adipose tissue of obese patients. SObesity (Silver Spring) 14: 794-798.
  90. Bánhegyi G, Benedetti A, Fulceri R, Senesi S (2004) Cooperativity between 11ß-hydroxysteroid dehydrogenase type 1 and hexose-6-phosphate dehydrogenase in the lumen of the endoplasmic reticulum. J BiolChem279: 27017-27021.
  91. Kershaw EE, Morton NM, Dhillon H, Ramage L, Seckl JR, Flier JS (2005) Adipocyte-specific glucocorticoid inactivation protects against diet-induced obesity. Diabetes. 54: 1023-1031.
  92. Hanson RW,Reshef L (2003) Glyceroneogenesis revisited. SBiochimie 85: 1199-1205.
  93. Franckhauser S, Muñoz S, Elias I, Ferre T, Bosch F (2006) Adipose overexpression of phosphoenolpyruvatecarboxykinase leads to high susceptibility to diet-induced insulin resistance and obesity. SDiabetes 55: 273-280.
  94. Chapman K, Holmes M, Seckl J (2013) 11β-hydroxysteroid dehydrogenases: intracellular gate-keepers of tissue glucocorticoid action. SPhysiol Rev 93: 1139-1206.
  95. Chapman K, Holmes M, Seckl J (2013) 11β-hydroxysteroid dehydrogenases: intracellular gate-keepers of tissue glucocorticoid action. SPhysiol Rev 93: 1139-1206.
  96. Whorwood CB, Donovan SJ, Flanagan D, Phillips DI, Byrne CD (2002) Increased glucocorticoid receptor expression in human skeletal muscle cells may contribute to the pathogenesis of the metabolic syndrome. SDiabetes 51: 1066-1075.
  97. Zhang M,Lv XY, Li J, Xu ZG, Chen L (2009) Alteration of 11beta-hydroxysteroid dehydrogenase type 1 in skeletal muscle in a rat model of type 2 diabetes. SMol Cell Biochem 324: 147-155.
  98. Whorwood CB, Donovan SJ, Wood PJ, Phillips DIW (2001) Regulation of glucocorticoid receptor a and ß isoforms and type I 11ß-hydroxysteroid dehydrogenase expression in human skeletal muscle cells: a key role in the pathogenesis of insulin resistance? Journal of Clinical Endocrinology and Metabolism 6: 229602308.
  99. Ruzzin J, Wagman AS, Jensen J (2005) Glucocorticoid-induced insulin resistance in skeletal muscles: defects in insulin signaling and the effects of a selective glycogen synthase kinase-3 inhibitor. Diabetologia. 48: 2119-2130.
  100. Salehzadeh F, Al-Khalili L, Kulkarni SS, Wang M, Lönnqvist F, et al. (2009) Glucocorticoid-mediates effects on metabolism are reversed by targeting 11ß-hydroxysteroid dehydrogenase type 1 in human skeletal muscles. Diabetes Metab Res Rev 2009. 25:250-258.
  101. vanUum SH,Hermus AR, Smits P, Thien T, Lenders JW (1998) The role of 11 beta-hydroxysteroid dehydrogenase in the pathogenesis of hypertension. SCardiovasc Res 38: 16-24.
  102. Livingstone DE, Jones GC, Smith K, Jamieson PM, Andrew R, et al. (2000) Understanding the role of glucocorticoids in obesity: tissue-specific alterations of corticosterone metabolism in obese Zucker rats. SEndocrinology 141: 560-563.
  103. Müssig K, Remer T, Haupt A, Gallwitz B, Fritsche A, et al. (2008) 11beta-hydroxysteroid dehydrogenase 2 activity is elevated in severe obesity and negatively associated with insulin sensitivity. SObesity (Silver Spring) 16: 1256-1260.
  104. Mai K,Kullmann V, Bobbert T, Maser-Gluth C, Möhlig M, et al. (2005) In vivo activity of 11beta-hydroxysteroid dehydrogenase type 1 and free fatty acid-induced insulin resistance. SClinEndocrinol (Oxf) 63: 442-449.
  105. Quinkler M, Stewart PM (2003) Hypertension and the cortisol-cortisone shuttle. SJ ClinEndocrinolMetab 88: 2384-2392.
Citation: Yaw HP, Ton SH, Kadir KA (2015) Glycyrrhizic Acid as the Modulator of 11β -hydroxysteroid dehydrogenase (Type 1 and 2) in Rats under Different Physiological Conditions in Relation to the Metabolic Syndrome. J Diabetes Metab 6:522.

Copyright: © 2015 Yaw HP, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.