You are on page 1of 8

Diabetes & Metabolic Syndrome: Clinical Research & Reviews 14 (2020) 1273e1280

Contents lists available at ScienceDirect

Diabetes & Metabolic Syndrome: Clinical Research & Reviews

journal homepage: www.elsevier.com/locate/dsx

Stress, glucocorticoid signaling pathway, and metabolic disorders


Roldan M. de Guia a, b, c, *
a
Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance and Network Aging Research, German Cancer Research Center (DKFZ), Heidelberg,
Germany
b
Keio Global Research Institute (KGRI) and Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan
c
Czech Centre for Phenogenomics (CCP), Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic

a r t i c l e i n f o a b s t r a c t

Article history: Background and aims: Glucocorticoids and the GR serve as an essential molecular mediator of stress and
Received 2 January 2020 different physiologic processes. This review summarizes main findings from studies on the role of the
Received in revised form GC/GR signaling in the modulation of genes for nutrient processing by the different organs involved in
15 June 2020
metabolic diseases.
Accepted 16 June 2020
Methods: Descriptive review of relevant papers known to the author was conducted.
Results: Several high-throughput screenings in the past 15 years have identified potential GR DNA-
Keywords:
binding regions in different cell types with genes that are annotated to be important for the control of
Glucocorticoids
Glucocorticoid receptor
metabolism. Transcriptional regulation of these GC-responsive genes provides links between the
Metabolism hypothalamic-pituitary-adrenal axis (HPA) and systemic energy homeostasis in both physiological and
Metabolic disorders pathophysiological states. Future studies must reconsider the use of agonist, the utilization of animal
models of stress and metabolic disorders, and validation in humans.
Conclusion: This review recapitulates the significant role of the GC/GR signaling in molecular metabolic
control and metabolic disorders. Potential future research focus and optimizations have also been
identified.
© 2020 Diabetes India. Published by Elsevier Ltd. All rights reserved.

1. Introduction this review that despite the pleiotropic effects of GCs in the body,
inter-organ differences and similarities may exist in the gamut of
Stress refers to any processes that put a strain to all levels of GC-responsive genes and that some of these genes might be
biological complexity. From population and groups down to the involved in metabolic diseases. Furthermore, important gaps in the
individual, organ-tissue, cellular, and genetic levels, stress has current knowledge of stress signaling in metabolism may be
become a connotation of the definition of life encompassing identified.
adaptation of the organism and capacity for metabolism. Stress
together with the mammalian HPA axis have been thoroughly and
widely studied for decades. Similar is true for the molecular basis of 2. The hypothalamic-pituitary-adrenal axis
stress response via the GC signaling pathway. The involvement of
GC signaling in the control of nutrient and energy metabolism GCs, as counter-regulatory hormone to insulin, serve a vital role
started from earlier studies on genes involved in gluconeogenesis. in metabolism and inflammatory response. The concentration of
With the current global epidemic of diabetes and obesity, the GCs in the body is correlated to the activity of the HPA axis [1].
contribution of the GC signaling to the pathophysiology of these Fasting, starvation, and diabetes are perceived as stress by the
metabolic diseases has become a focal point of research as lipid neuroendocrine system in response to changes in the levels of
overload and defective carbohydrate metabolism may alter the glucose, fatty acids, and hormones [2e4]. The stress signal causes
body’s stress response. It can be hypothesized for the purpose of the hypothalamus to secrete corticotrophin releasing factor (CRF)
which stimulates the anterior pituitary gland to produce
adrenocorticotrophin-releasing hormone (ACTH) (Fig. 1A) [5].
* Corresponding author. Czech Centre for Phenogenomics (CCP), Institute of
ACTH in the bloodstream then activates the adrenal glands to
Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic. produce hormones. The adrenal medulla secretes catecholamines
E-mail address: roldan.deguia@img.cas.cz. (norepinephrine and epinephrine) and the adrenal cortex produces

https://doi.org/10.1016/j.dsx.2020.06.038
1871-4021/© 2020 Diabetes India. Published by Elsevier Ltd. All rights reserved.
1274 R.M. de Guia / Diabetes & Metabolic Syndrome: Clinical Research & Reviews 14 (2020) 1273e1280

Fig. 1. Mechanism of glucocorticoid and receptor action. (A) The hypothalamic-pituitary-adrenal axis showing release of CRF from the hypothalamus and ACTH from the pituitary
gland. ACTH promotes synthesis of cortisol by the adrenal gland; (B) The pre-receptor function of hydroxysteroid dehydrogenase which interconverts cortisol and cortisone; (C) The
glucocorticoid receptor is rendered free from the complex allowing GC binding. The GC/GR complex moves into the nucleus to bind its target; (D) Types of glucocorticoid receptor-
DNA interaction with the target can be direct and composite allowing the use of glucocorticoid response elements in the DNA. GC/GR can likewise bind to other DNA-binding
proteins in tethering manner.

mineralocorticoids (from zona glomerulosa), androgens, and the in the cell [11] (Fig. 1B). Furthermore, the relative expression of
GCs (zona fasciculata & zona reticularis) [5]. ACTH stimulates fas- these two isozymes in different organs of the body contribute to the
ciculata and reticularis cells to synthesize cortisol from cholesterol. tissue-specific effect of GCs [12]. HSD1 is expressed predominantly
Cortisol is the major stress hormones that acts on every cell of the in the liver, lungs, gonads, brain, and adipose tissues. HSD2, on the
body including the brain where it negatively feedbacks on CRF and other hand, is expressed in the kidneys, colon, and salivary glands
ACTH secretion making the HPA a close neuroendocrine feedback [13,14].
loop [6]. Once secreted in the circulation, majority of cortisol (75%) The GR mediates the function of the cortisol (corticosterone in
bind to transcortin and corticosteroid-binding globulin (CBG) [7]. mice). It is ubiquitously expressed numbering from 2000e30,000
Levels of these GC-binding proteins are stress-regulated [8]. Free molecules/cell with mRNA levels particularly high in the lungs,
cortisol concentration is being regulated by the HPA axis since it is spleen, brain, and liver. It is a member of the nuclear receptor family
the unbound cortisol which is responsible for the biological func- characterized by the presence of a highly conserved, centrally
tion of GCs [9,10]. located zinc-finger DNA-binding domain, a less conserved C-ter-
minal ligand-binding domain which contains the GC-binding re-
3. Mechanism of glucocorticoid and receptor action gion, and amino acid residues for dimerization and transactivation
functions [15,16]. The inactive GR in the cytoplasm is phosphory-
The molecular mechanism of GCs can be divided into pre- lated and forms a complex with other proteins which include
receptor and receptor-mediated actions. The pre-receptor ligand HSP90, HSP70, p59 immunophilin, FKBP52, and p23 phosphopro-
control regulates the intracellular concentration of the active GC, tein (Fig. 1C). GR is activated after it binds GC, hyper-
cortisol. It depends on the activity of the enzyme 11b-hydroxyste- phosphorylated, and is freed from the inhibitory complex [17].
roid dehydrogenase (HSD) in the endoplasmic reticulum. The two The GC/GR complex then translocates into the nucleus where it can
isozymes HSD1 and HSD2 are responsible for reductive (cortisone bind to the promoter regions of genes which harbors (1) GC
/ cortisol) and oxidative (cortisol / cortisone) reactions, response elements (GRE) where the GR binds as a dimer to the
respectively, that alter the concentration of the bio-active cortisol pentadecameric palindrome GGTACAnnnTGTTCT, (2) half-GRE
R.M. de Guia / Diabetes & Metabolic Syndrome: Clinical Research & Reviews 14 (2020) 1273e1280 1275

sites, or (3) negative GC response elements [17]. These GREs can In adipose tissue, ANGPTL4 activates lipolysis and inhibits LPL ac-
also function in composite manner together with other transcrip- tivity resulting into elevation of free fatty acids in the circulation
tion factors and co-regulators. Tissue specificity of GC action also [41]. This process is likewise implicated in fatty acid redistribution
depends on the presence of these proteins which are produced in from adipose tissue to the liver which in turn increases triglyceride
accordance to the requirements by the tissue [18]. The GR action synthesis of hepatocytes [42,43]. GCs are known to affect lipopro-
can also be a secondary response where there is no direct GR-DNA tein metabolism. Studies done on primary hepatocytes and
interaction but protein-protein interaction of the GR with other dexamethasone-treated mice showed increased VLDL triglycerides
proteins [19] (Fig. 1D). An example of this is the trans-repression by and plasma concentration of HDL particles [44e46]. Promotion of
the GR in inflammatory response together with NF-kB and AP1 [20]. VLDL secretion by GCs is possibly due to increased production and
Apart from the physiologic stresses that activate the HPA which stabilization of Apolipoprotein B (ApoB) together with increased
ultimately lead into cortisol production and GR binding, several triglyceride synthesis [47]. ApoB is the major protein component of
factors have been identified affecting GC/GR signaling. Cortisol VLDL and LDL. At least 50% of cholesterol needed by the adrenal
production follows a circadian rhythm where it is highest during gland to produce GCs are shown to be provided by lipoproteins [48]
activity period (day for diurnal species, night for nocturnals) with adrenocortical LDLR playing an important role [49]. Elevated
peaking at ~6e8 am in humans and lowest 3e5 h after onset of plasma HDL, on the other hand, can be attributed to the inhibition
sleep [21]. Although the general pattern is circadian, endogenous of hepatic lipase and activation of LCAT activity by GCs [50]. GCs
cortisol level follows an ultradian pattern where GCs are being have also been shown to promote triglyceride synthesis and stea-
released every hour [22]. This pattern affects GR activity where tosis in the liver by inhibition of the hairy and enhancer of split 1
pulses in the levels drive transient activation of GR-dependent (HES1) repressor [51]. The GR also controls induction of the Naþ-
genes [23]. The subsequent nuclear import of the active GR dur- taurocholate cotransporter (NTCP or SLC10A1) important for
ing each pulse also affects GC response. Although still unclear, it transporting bile acids from the blood back to the liver [52].
was reported that GR nucleocytoplasmic shuttling might be regu- Over the years, high-throughput chromatin immunoprecipita-
lated by the p38 mitogen-activated protein kinase (MAPK) pathway tion (ChIP) sequencing has been applied to identify genes that are
[24]. Another factor that affects GR activity is chromatin accessi- potentially being directly targeted by a transcription factor. GR ChIP
bility. Regulatory regions of GR-responsive genes have been shown scanning studies revealed enrichment of GC-binding regions (GBR)
to have accessible DNA and acetylated histones where the GR can in the promoter of genes that function in transcription, cellular
interact with the chromatin remodeling complex made up of transport, biomolecular synthesis, cell surface receptor linked cell
coactivators CREB-binding protein (CBP), p300, activator of thyroid signaling, cell proliferation, metabolism, and inflammation, among
hormone and retinoid receptors (ACTR), SRC1, p300/CBP-associated others [58,59]. Although common targets can be pinpointed, it can
factor (PCAF), and SWI/SNF complex [15,17]. PCAF has histone be generalized that GCs regulate different genes in different cell
acetyltransferase (HAT) activity while the latter complex possesses types due to the tissue-specific action of the GR (Table 1). In the
the chromatin remodeling activity. Both of which are important for liver, adipose tissues, and muscles, most of the identified GC-
further binding of other proteins and the components of the tran- regulated genes in metabolism correlate with the results of
scriptional machinery [25,26]. in vivo and in vitro functional studies utilizing GR agonist (dexa-
methasone, prednisone, etc.) treatment [55,57,61]. Furthermore,
4. Regulation of metabolism by glucocorticoids enrichment analysis for transcription factors and cofactors in GBRs
revealed proteins that are possibly involved in composite or teth-
GCs are involved in the regulation of carbohydrate, fat, and ering regulation by the GR. Some of these transcription factors
protein metabolism in both the fed and fasted state (Fig. 2). GCs include AP1, C/EBP, FoxA, FOXP3, PXR, LXR, Med1, HNF3a, HNF6,
work with glucagon, growth hormone, and thyroid hormone in PPARa, PPARg, T3R, GABPA, ZFP281, PRRX2, GATA1, RelA, Jun, NF-
bringing about counter-regulatory effects of insulin in the cell kB, and STAT5. Just like the gene targets, tissue-specificity is
[27e29]. However, under normal physiologic fed state following observed in the set of transcription factors that are co-enriched
starvation, GCs work with insulin in a synergistic manner [13,17]. within GBRs [62].
GCs and insulin potentiate lipogenesis and glycogenesis in the liver Apart from enzymes and transcription factors that are being
and promotes differentiation, lipogenesis and increased activity of controlled by the GR directly or indirectly, non-coding RNAs,
LPL and GPDH in adipose tissues [30]. GCs also stimulate glycolysis particularly microRNAs (miRNAs), has been reported also to be
by activation of GCK, PFK, and PK [1]. involved in GC/GR signaling. The growth arrest-specific 5 (Gas5)
Fasting and starvation activate enzymes important for lipolysis, non-coding RNA has been reported to act as a decoy GRE in the cells
b-oxidation, protein degradation, glycogenolysis, and gluconeo- deprived of nutrients or growth factors [63] while in models of
genesis [1]. Most of the key enzymes involved in these processes acute lymphoblastic leukemia, GCs have been shown to regulate
are also regulated by glucagon like PEPCK & G6Pase in gluconeo- miR-223 and the miR-15/16 cluster [64]. Reciprocally, the GC
genesis, HSL in lipolysis, among others [31,32]. In peripheral biosynthetic pathway has been reported to be regulated by miRNAs
glucose uptake, GCs induce expression but inhibit the translocation [65]. In the liver, we previously reported miRNAs of the miR-379-
to the membrane of the GLUT4 transporter in both muscles and 410 cluster that are upregulated in diabetic-obese db/db mice
adipose tissues [33,34]. GCs have also been shown to affect glucose- suffering from chronically elevated corticosterone [66]. We were
stimulated insulin release in b-cells of the pancreas [35,36]. In both able to identify and validate GC-binding region in the putative
rats and humans, GCs are reported to stimulate mitochondrial promoter region of the miRNA gene locus [66]. It is in these disease
NADPH production, Ca2þ and PLC/InsP3/PKC pathways which are state of GC excess (or insufficiency) where the importance of GC
implicated in the release of insulin from b-cells [37]. and GR signaling in metabolism can further be justified.
In lipid metabolism under energy-deprived state, GCs activate
transcription of genes in lipolysis like ATGL, HSL, and MGL via in- 5. Glucocorticoids and metabolic disorders
duction of FoxO transcription [30,38]. GCs also promote secretion of
ANGPTL4 from the liver and white adipose tissues [39]. ANGPTL4 GC and GR signaling dysfunctions affect carbohydrate, lipid, and
has been demonstrated to be involved in dexamethasone- protein metabolism making the GC/GR pathway an attractive target
dependent, hepatic ceramide production and dyslipidemia [40]. for the management of metabolic diseases [67]. Disturbances in the
1276 R.M. de Guia / Diabetes & Metabolic Syndrome: Clinical Research & Reviews 14 (2020) 1273e1280

Fig. 2. Enzymes and cellular processes regulated by the glucocorticoid signaling during fasting and feeding. During the fasted state, glucocorticoids act together with other hor-
mones in favor of lipolysis, beta-oxidation, proteolysis, glycogenolysis and gluconeogenesis. In the fed state, glucocorticoids can act synergistically with insulin. Glucocorticoids can
also affect glycogenesis and lipoprotein production & processing by the liver.
NOTE: Up-regulated by GC/GR; Down-regulated; Atgl (adipocyte triglyceride lipase; also known as desnutrin or patatin-like phospholipase domain containing 2
(PNPLA2)); Hsl (hormone-sensitive lipase; also known as LIPE); Mgl (monoacylglyceride lipase); Angptl4 (angiopoietin-like 4); Lpl (lipoprotein lipase); Gpdh (Glycerol-3-
phosphate dehydrogenase); Ampk (AMP-associated protein kinase); Pdk4 (pyruvate dehydrogenase lipoamide kinase 4); Redd1 (regulated in development and DNA damage
response 1); Klf15 (Krüppel-like factor 15); Gys (glycogen synthase); Acc (acetyl-CoA carboxylase); Fasn (fatty acid synthase); Scd (stearoyl-CoA desaturase); Gpat (glycerol
phosphate acyltransferase); Agpat (1-acylglycerol-3-phosphate acyltransferase; also MOGAT, monoacylglycerol acyltransferase); Lpin (lipin or phosphatidate phosphatase); Dgat
(diacylglycerol acyltransferase); TAG hydrolase (triacylglyceride hydrolase); Ldlr (LDL receptor); LSR (lipolysis-stimulated lipoprotein receptor); Gck (glucokinase); Pfk (phos-
pofructokinase); Pk (pyruvate kinase); Pepck (phosphoenolpyruvate carboxykinase); G6Pase (glucose-6-phosphatase).

HPA axis, the pre-receptor function, and the GC/GR signaling have specificity of the GC response in the context of insulin resistance.
been reported in almost all the components of the metabolic syn- The bone g-carboxyglutamate protein (BGALP) or osteocalcin,
drome (MetSyn) e insulin resistance, hyperglycemia, dyslipidemia, which is important in bone turnover, has been reported to partic-
and hypertension [68e71]. The abnormal phenotypes of the Met- ipate in the regulation of insulin sensitivity [81]. Osteocalcin’s
Syn have been observed to occur with hyperactive HPA axis and ability to inhibit adiposity and hepatic steatosis has been shown to
elevated cortisol levels in humans, a state called “functional be blunted by GCs [82]. A recent study in humans indicated that
hypercortisolism” [67,72,73]. Chronic stress and low birth weight GC-induced insulin resistance in both basal and post-exercise
have been reported to be associated with increased HPA activity conditions is correlated with reduced serum undercarboxylated
and circulating cortisol [72] and affects levels of GR and MR re- osteocalcin levels [83]. In the muscles and liver, GCs can induce
ceptors in the brain [74]. Cortisol levels are also described to be insulin resistance by inhibiting the transcription or phosphoryla-
abnormal in people with central obesity where there is an tion of IRS-1 and increasing levels of tyrosine phosphatase (PTP1B)
increased rate of secretion by the adrenal glands and clearance by and p38MAPK. The opposite scenario has been reported for the
the kidneys [75]. It was shown that plasma cortisol increases with latter in 3T3-L1 adipocytes [84].
age in obese individuals [76]. BMI is likewise positively correlated GC-induced, selective insulin resistance is likewise implicated to
with chronically elevated cortisol levels in shift workers [77]. Rats the pre-receptor function of the HSD. Animal models over-
treated with dexamethasone for 4 weeks displayed significantly expressing 11b-HSD1 in adipocytes resulted in obesity and insulin
higher triglycerides, hematocrit, and insulin compared to controls resistance while gene knockout models showed the opposite
injected with the vehicle [78]. These findings further support the [85,86]. Humans suffering from morbid obesity have been reported
involvement of the GC/GR signaling in insulin secretion, lipid to have high levels of 11b-HSD1 in the liver and visceral adipose
metabolism, and insulin resistance. GC-induced insulin resistance tissues [87]. In this study, the liver had 20 times more of the
was also observed in human skeletal muscles, omental fat, but not enzyme than the visceral fats although the hepatic levels were
in subcutaneous adipose tissues [79,80]. This suggest tissue- negatively correlated with BMI suggesting possible secondary
R.M. de Guia / Diabetes & Metabolic Syndrome: Clinical Research & Reviews 14 (2020) 1273e1280 1277

Table 1
Genes, cellular processes, and pathways enriched in glucocorticoid receptor ChIP-Seq.

Tissue/Cell Line Selected Genes or Enriched Cell Processes/Pathways in GR ChIP-Seq References

Rat Neonatal Positive regulation of transcription; Positive regulation of cell death; Response to lipopolysaccharide; Regulation of gene Severinova et al.,
Cardiomyocytes expression; Cellular response to molecular stimulus; Apoptotic processes; Liver regeneration, Cellular response to insulin 2019 [53]
stimulus; Axon guidance; Focal adhesion; ECM-receptor interaction; PI3K-Akt signaling; Protein digestion and absorption;
Dilated and hypertrophic cardiomyopathy; Adrenergic signaling; Rap1 signaling; Pathways in cancer; Insulin signaling
Human Primary Molecular mechanisms of cancer; Role of macrophages, fibroblasts, and endothelial cells in rheumatoid arthritis; Hepatic Singh et al., 2015
Subcutaneous Fat fibrosis/hepatic stellate cell activation; IL-8 signaling; HGF signaling; B cell receptor signaling; GC receptor signaling [54]
AHSP, BIVM-ERCC5, C16orf3, CCKBR, CNKSR2, DDX11L5, DEPTOR, DSTYK, EDC3, EEF1A1, GH1, HBB, HIST1 (H2BC, H4B, H4C,
H4H,H4I, H4L), HIST4H4, ING3, KIF9-AS1, LRRCC1, MIR4271, MOV10, NAF1, NR2F1-AS1, NR3C1, NR3C2, PABPC1, PAIP2,
SGK1, SLC18B1, USP14, VPS26A, ZNF219, ZNF385B
C2C12 Myotubes Insulin signaling: Grb10, Sorbs1, Cblb, Pid1, p85a, Redd1, Sesn1, Depdc6 Kuo et al., 2012
[55]
PC12 Neurons Neuronal development, cytoplasmic vesicle, neuron projection, metal ion binding, angiogenesis, synapse, protein tyrosine Polman et al., 2012
kinase activity [56]
3T3-L1 Adipocytes Insulin action (Insig2, Lcn2, Pi3kr1 or p85a, Ptgds), adipocyte differentiation (Tsc22d3, Ctsl, Sgk1), triglyceride synthesis (Scd, Yu et al., 2010 [57]
Gpat, Agpat, Lpin1), lipolysis (Hsl, Mgll), lipid transport (Cd36, Lrp1, Vldlr, Slc27a2or Fatp2), lipid storage (S3-12)
A549 Lung Epithelial Inflammation (CCL20, COX-2, IL11), growth and apoptosis (GADD45B, FGFBP1), metabolism (ANGPTL4, B3GNT5, EKI2, MGAM), Wang et al., 2004
Carcinoma transport (MT1L, SLC19A2, SLC26A2) | anatomical structure development, stress response, regulation of gene expression, [58]
apoptosis, transcription factor activity, signal transduction Reddy et al., 2009
[59]
Rat Hypothalamus Gadd45g, Sgk, Pdk4, Nfkbia, Plekhf1, Redd1, Gpd1, Mt1a, Hes5 Sato et al., 2008
[60]
Mouse Liver Adfp, Adh1, Aldh3a2, ApoA2, ApoC1, Abca1, Arg1, Creb1, Ch25h, Clock, Cs, Cyp11a1, Dld, Facl2, Gys1, Gys3, Hmgcs2, Hsd17b12, Phuc Le et al., 2005
Hsd3b4, Igfbp1, Idh1, Isl2, Il11ra2, Klf15, Lztr1, Lipc, Lipe, Lipc, Oat, Pck1, Pygl, Lrp4, Mgll, Nqo2, Ndufa7, Odc, Pgd, Pik3cg, Pdsm, [61]
Prkag3, Ppm1g, Ppp2r3a, Ptp4a2, Pdk2, Retnla, Sepp1, Serpin, Slc1a4, Sra1, Tgfb2, Tnf

NOTE: Complete list of genes, cell processes, and pathways at specific and different time of agonist incubations can be found in each respective source.
Abca (ATP binding cassette subfamily A), Adfp (perilipin 2), Adh1 (alcohol dehydrogenase 1), Agpat (1-Acylglycerol-3-Phosphate O-Acyltransferase), AHSP (alpha hemoglobin
stabilizing protein), Aldh (aldehyde dehydrogenase), ANGPTL4 (angiopoietin like 4), Apo (apolipoprotein), Arg1 (arginase 1), B3GNT (UDP-GlcNAc:BetaGal Beta-1,3-N-
Acetylglucosaminyltransferase), BIVM-ERCC5 (BIVM-ERCC5 readthrough), Cblb (Cbl proto-oncogene B), CCKBR (cholecystokinin B receptor), CCL (CeC motif chemokine
ligand), Clock (clock circadian regulator), Ch25h (cholesterol 25-hydroxylase), CNKSR2 (connector enhancer of kinase suppressor of Ras 2), COX (cyclooxygenase), Creb (cAMP
responsive element binding protein), Cs (citrate synthase), Ctsl (cathepsin L), Cyp (cytochrome P450), DDX11L (DEAD/H-Box Helicase 11 Like), Depdc6 (DEPTOR), DEPTOR
(DEP domain containing MTOR-interacting protein), Dld (dihydrolipoamide dehydrogenase), DSTYK (dual serine/threonine and tyrosine protein kinase), ECM (extracellular
matrix), EDC3 (enhancer of mRNA decapping 3), EEF (eukaryotic translation elongation factor), EKI (ethanolamine kinase), Facl (fatty-acid-coenzyme A ligase long chain),
FGFBP (fibroblast growth factor binding protein), GADD45 (growth arrest and DNA damage inducible beta), GH (growth hormone), Gpat (glycerol-3-phosphate acyl-
transferase), Gpd (glycerol-3-phosphate dehydrogenase), Grb (growth factor receptor bound protein), Gys (glycogen synthase), HBB (hemoglobin subunit beta), Hes (hes
family bHLH transcription factor), HGF (hepatocyte growth factor), HIST (histone cluster), Hmgcs (3-hydroxy-3-methylglutaryl-CoA synthase), Hsd17b12 (hydroxysteroid 17-
beta dehydrogenase 12), Hsd3b (hydroxy-delta-5-steroid dehydrogenase), Hsl (hormone-sensitive lipase), Idh1 (isocitrate dehydrogenase (NADP(þ)) 1, cytosolic), Igfbp1
(insulin like growth factor binding protein), IL (interleukin), ING (inhibitor of growth), Insig (insulin induced gene), Isl2 (ISL LIM homeobox 2), KIF9-AS (KIF9 antisense RNA),
Klf (kruppel like factor), Lcn (lipocalin), Lipc/e (lipase c or e), Lpin1 (lipin 1), Lrp (LDL receptor related protein), LRRCC (leucine rich repeat and coiled-coil centrosomal
protein), Lztr (leucine zipper like transcription regulator), MGAM (maltase-glucoamylase), Mgll (monoglyceride lipase), MIR (microRNA), MOV10 (Mov10 RISC complex RNA
helicase), MT (metallothionein), NAF1 (nuclear assembly factor 1 ribonucleoprotein), Ndufa7 (NADH:ubiquinone oxidoreductase subunit A7), Nfkbia (NFKB inhibitor alpha),
Nqo (NAD(P)H quinone dehydrogenase), NR2F1-AS (NR2F1 antisense RNA), NR3C (nuclear receptor subfamily 3 group C), Oat (ornithine aminotransferase), Odc (ornithine
decarboxylase), PABPC (poly(A) binding protein cytoplasmic), PAIP (poly(A) binding protein interacting protein), Pck (phosphoenolpyruvate carboxykinase), Pdk (pyruvate
dehydrogenase kinase), Pdsm (phosphorylation-dependent sumoylation motif), Pgd (phosphogluconate dehydrogenase), PI3K-Akt (phosphatidylinositol 3-kinase - protein
kinase B), Pi3kr1 or p85a (phosphoinositide-3-kinase regulatory subunit 1), Pid (phosphotyrosine interaction domain containing), Pik3 (phosphatidylinositol-4,5-
bisphosphate 3-kinase), Plekhf (pleckstrin homology and FYVE domain containing), Ppm (protein phosphatase, Mg2þ/Mn2þ dependent), Ppp2r (protein phosphatase 2
regulatory subunit), Prkag (protein kinase AMP-activated non-catalytic subunit gamma), Ptgds (prostaglandin D2 synthase), Ptp4a (protein tyrosine phosphatase type IVA),
Pygl (phosphorylase, glycogen, liver), Rap (Ras-related protein), Redd1/Ddit4 (DNA Damage Inducible Transcript 4), Retnla (resistin-like alpha), S3-12/PLIN4 (perilipin 4), Scd
(stearoyl-CoA desaturase), Sepp1 (selenoprotein P), Serpin (serine protease inhibitor), Sesn1 (Sestrin 1), SGK1 (serum/glucocorticoid regulated kinase 1), SLC (solute carrier),
Sorbs (sorbin and SH3 domain containing), Sra (steroid receptor RNA activator), Tgf (transforming growth factor), Tnf (tumor necrosis factor), Tsc22d3 (TSC22 domain family
member 3), USP (ubiquitin specific peptidase), Vldlr (very-low density lipoprotein receptor), VPS26A (retromer complex component A), ZNF (zinc finger protein).

effects of visceral adiposity and portal hypercortisolemia. Indeed, therapy [90,91]. While GC deficiency is a characteristic of primary
mice overexpressing 11b-HSD1 in adipocyte had increased active (Addison’s disease, AD) or secondary adrenal insufficiency. The
corticosterone secretion from visceral fats which in turn lead into abnormally high levels of GCs in individuals suffering from
increased GC input into the liver without changing plasma levels endogenous CS can be attributed to the presence of ACTH-secreting
[85]. Hepatocyte-specific, 11b-HSD1 over-expression, on the other pituitary adenomas (75e80% of cases) or ectopic corticotropin
hand, did not lead into obesity but displayed phenotypes of syndrome [92]. Iatrogenic or exogenous CS, on the other hand, is
metabolic syndrome like insulin resistance, fatty liver, dyslipide- due to excessive use of topical or inhaled corticosteroids as part of
mia, and impaired hepatic lipid clearance [85]. All these reports one’s medications [93]. Hypercortisolemia in patients with CS is
made 11b-HSD1 a potential target in mitigating phenotypes of the responsible for the development of insulin resistance, diabetes,
MetSyn [88]. Besides the 11b-HSD1 activity, MetSyn phenotypes dyslipidemia, cardiovascular diseases, and osteoporosis [94].
have been implicated to the expression of the GR itself. High levels Obesity is common to 95% of CS cases which further contributes to
of both hepatic 11b-HSD1 & GR expression have been reported to the risk of cardiovascular morbidity [95]. Furthermore, proximal
correlate with hyperglucocorticoidemia, hyperinsulinemia, and muscle weakness, fatigue, menstrual irregularity, and neuropsy-
hyperglycemia in diabetic-obese db/db mice [89]. chological disturbances are also observed in individuals suffering
The vital role of the GC/GR axis in MetSyn syndrome is further from CS [92]. Adult AD, on the other hand, is clinically characterized
exemplified in conditions of GC excess and deficiency. Hyper- by low blood pressure, hyperpigmentation of the skin, weight loss,
cortisolemia is observed on individuals suffering from endogenous joint & muscle pains & weakness, fatigue, or in severe cases, pro-
and exogenous Cushing syndrome (CS) and people receiving GC gression into the so called ‘Addisonian or adrenal crisis’ [96]. The
1278 R.M. de Guia / Diabetes & Metabolic Syndrome: Clinical Research & Reviews 14 (2020) 1273e1280

latter manifests as mixed symptoms which may include severe Declaration of competing interest
vomiting & diarrhea, syncope (loss of consciousness & inability to
stand), hypoglycemia, lethargy, hyponatremia, hyperkalemia, hy- The author declares that there are no conflicts of interest.
percalcemia, and fever [97]. AD is caused by adrenal cortex auto-
immunity (80% of cases), tuberculosis (10e15%), among others [96]. Acknowledgments
Secondary adrenal deficiency can develop when the causes of the
problem arose from outside the adrenal gland like in faulty pitui- Support for this study was provided by the Helmholtz Interna-
tary ACTH production. tional Graduate School for Cancer Research, Heidelberg, Germany;
Keio Global Research Institute (KGRI), Tokyo, Japan; and the Czech
Centre for Phenogenomics (CCP), Institute of Molecular Genetics of
the Czech Academy of Sciences (LM2015040). Furthermore, the
6. Relevance of the review and future perspective author would like to thank Prof. Dr. Stephan Herzig of the Institute
for Diabetes and Cancer, Helmholtz Zentrum München and Prof.
Whether the problem is congenital, induced by xenobiotics, or Masato Yasui of the School of Medicine and Keio Global Research
from a secondary source, chronic disturbances in the HPA axis Institute, Keio University.
affect normal function of the neuroendocrine system in handling
stress. The resulting damage is usually mediated by GCs, mineral-
References
ocorticoids, and androgens and their respective receptors which
impacts the immune system and metabolism. Treatment regimens [1] Andrews RC, Walker BR. Glucocorticoids and insulin resistance: old hor-
for stress and GC-related pathologies include drug therapy, surgery, mones, new targets. Clin Sci (Lond). 1999;96:513e23.
or even replacement therapy depending on the associated patho- [2] Morton GJ. Hypothalamic leptin regulation of energy homeostasis and
glucose metabolism. J Physiol 2007;583:437e43.
physiologic phenotypes [98]. Diabetes & dyslipidemia arising from [3] Koch C, Augustine RA, Steger J, Ganjam GK, Benzler J, Pracht C, et al. Leptin
CS are treated in the same way as in the MetSyn [99]. Despite ev- rapidly improves glucose homeostasis in obese mice by increasing hypo-
idences supporting the importance of GCs in nutrient and energy thalamic insulin sensitivity. J Neurosci 2010;30:16180e7.
[4] Lam TK, Schwartz GJ, Rossetti L. Hypothalamic sensing of fatty acids. Nat
metabolism and the connection between hyperactive GC/GR Neurosci 2005;8:579e84.
signaling and metabolic dysfunction, knowledge is still limiting as [5] Marieb EN, Keller SM. Essentials of human anatomy & physiology. twelfth ed.
to how GR-dependent metabolic processes are being regulated or if Ny, Ny: Pearson; 2018.
[6] Chrousos GP, Kino T. Glucocorticoid signaling in the cell. Expanding clinical
GCs regulate expression of transcriptional or translational regula-
implications to complex human behavioral and somatic disorders. Ann N Y
tors. The census of high-throughput, ChIP-seq studies provided in Acad Sci 2009;1179:153e66.
this review, which tabulates putative GR targets, clearly shows or- [7] Perogamvros I, Aarons L, Miller AG, Trainer PJ, Ray DW. Corticosteroid-
gan specificity. Comparing one cell type to another in different GR binding globulin regulates cortisol pharmacokinetics. Clin Endocrinol (Oxf).
2011;74:30e6.
ChIP-seq studies is over simplistic. The list of genes and pathways [8] Lewis JG, Bagley CJ, Elder PA, Bachmann AW, Torpy DJ. Plasma free cortisol
compiled in Table 1 might demonstrate specificity but it is still a fraction reflects levels of functioning corticosteroid-binding globulin. Clin
major question whether this is truly GR-mediated or due to study Chim Acta 2005;359:189e94.
[9] Dunn JF, Nisula BC, Rodbard D. Transport of steroid hormones: binding of 21
design differences. Treatments of the cell lines or animals with the endogenous steroids to both testosterone-binding globulin and
agonist dexamethasone vary widely from one study to the next in corticosteroid-binding globulin in human plasma. J Clin Endocrinol Metab
terms of concentration and duration of treatments. Addressing the 1981;53:58e68.
[10] Mendel CM. The free hormone hypothesis: a physiologically based mathe-
acute and chronic implications of the agonist treatment must be matical model. Endocr Rev 1989;10:232e74.
done in future studies. Another important issue to consider is the [11] Draper N, Stewart PM. 11beta-hydroxysteroid dehydrogenase and the pre-
use of the agonist itself in the experiments. Dexamethasone is a receptor regulation of corticosteroid hormone action. J Endocrinol
2005;186:251e71.
very potent agonist of the GR and therefore might mask some of the
[12] Tomlinson JW, Walker EA, Bujalska IJ, Draper N, Lavery GG, Cooper MS, et al.
true effects of GR activation if dosage and duration of treatments 11beta-hydroxysteroid dehydrogenase type 1: a tissue-specific regulator of
are not optimized. glucocorticoid response. Endocr Rev 2004;25:831e66.
[13] Witchel SF, DeFranco DB. Mechanisms of disease: regulation of glucocorti-
In the context of energy metabolism, although many studies
coid and receptor levels–impact on the metabolic syndrome. Nat Clin Pract
have been done transcriptomics of GR activation, few studies have Endocrinol Metabol 2006;2:621e31.
validated the results in GR ChIP experiments to show the type of GR [14] Harris HJ, Kotelevtsev Y, Mullins JJ, Seckl JR, Holmes MC. Intracellular
action on the target. This review included GR Chip-seq studies from regeneration of glucocorticoids by 11beta-hydroxysteroid dehydrogenase
(11beta-HSD)-1 plays a key role in regulation of the hypothalamic-pituitary-
tissues like the lungs and the heart which might be of interest for adrenal axis: analysis of 11beta-HSD-1-deficient mice. Endocrinology
future metabolic studies on the GC signaling. Majority of the studies 2001;142:114e20.
presented on Table 1 were done in vitro. It will be more physio- [15] Kadmiel M, Cidlowski JA. Glucocorticoid receptor signaling in health and
disease. Trends Pharmacol Sci 2013;34:518e30.
logically relevant if future studies are to be performed in animal [16] Schaaf MJ, Cidlowski JA. Molecular mechanisms of glucocorticoid action and
models of stress and metabolic disorders. Validation of the animal resistance. J Steroid Biochem Mol Biol 2002;83:37e48.
studies in human intervention, cross-sectional, or prospective [17] Schoneveld OJ, Gaemers IC, Lamers WH. Mechanisms of glucocorticoid sig-
nalling. Biochim Biophys Acta 2004;1680:114e28.
studies will be valuable for translational relevance to human cases [18] Rose AJ, Vegiopoulos A, Herzig S. Role of glucocorticoids and the glucocor-
of metabolic and adrenal disorders. Our recent findings on GC- ticoid receptor in metabolism: insights from genetic manipulations. J Steroid
induced miRNAs of the miR-379-410 cluster might provide a Biochem Mol Biol 2010;122:10e20.
[19] Petta I, Dejager L, Ballegeer M, Lievens S, Tavernier J, De Bosscher K, et al. The
model for future studies to seek for the link between GC/GR interactome of the glucocorticoid receptor and its influence on the actions of
signaling and the dysfunctional phenotypes of the MetSyn in both glucocorticoids in combatting inflammatory and infectious diseases. Micro-
mice and humans [66]. Additional functional and pharmacological biol Mol Biol Rev 2016;80:495e522.
[20] Ling J, Kumar R. Crosstalk between NFkB and glucocorticoid signaling: a
studies are necessary to provide more light on this hypothesis.
potential target of breast cancer therapy. Canc Lett 2012;322:119e26.
Furthermore, efforts and resources must continue to be focused on [21] Chung S, Son GH, Kim K. Circadian rhythm of adrenal glucocorticoid: its
the growing interests on the potential pivotal contribution of epi- regulation and clinical implications. Biochim Biophys Acta 2011;1812:
genetics, sexual dimorphism, age at exposure to stressors, and the 581e91.
[22] Flynn BP, Conway-Campbell BL, Lightman SL. The emerging importance of
microbiome to the impact of GCs on metabolic, cardiovascular, ultradian glucocorticoid rhythms within metabolic pathology. Ann Endo-
immune, and neurological health [100e103]. crinol (Paris) 2018;79:112e4.
R.M. de Guia / Diabetes & Metabolic Syndrome: Clinical Research & Reviews 14 (2020) 1273e1280 1279

[23] Young EA, Abelson J, Lightman SL. Cortisol pulsatility and its role in stress lipase activity. Biochim Biophys Acta 1981;663:69e82.
regulation and health. Front Neuroendocrinol 2004;25:69e76. [51] Lemke U, Krones-Herzig A, Berriel Diaz M, Narvekar P, Ziegler A,
[24] Irusen E, Matthews JG, Takahashi A, Barnes PJ, Chung KF, Adcock IM. p38 Vegiopoulos A, et al. The glucocorticoid receptor controls hepatic dyslipi-
Mitogen-activated protein kinase-induced glucocorticoid receptor phos- demia through Hes1. Cell Metabol 2008;8:212e23.
phorylation reduces its activity: role in steroid-insensitive asthma. J Allergy [52] Rose AJ, Berriel Diaz M, Reimann A, Klement J, Walcher T, Krones-Herzig A,
Clin Immunol 2002;109:649e57. et al. Molecular control of systemic bile acid homeostasis by the liver
[25] Muratcioglu S, Presman DM, Pooley JR, Grontved L, Hager GL, Nussinov R, glucocorticoid receptor. Cell Metabol 2011;14:123e30.
et al. Structural modeling of GR interactions with the SWI/SNF chromatin [53] Severinova E, Alikunju S, Deng W, Dhawan P, Sayed N, Sayed D. Glucocor-
remodeling complex and C/EBP. Biophys J 2015;109:1227e39. ticoid receptor-binding and transcriptome signature in cardiomyocytes. J Am
[26] Blanco JC, Minucci S, Lu J, Yang XJ, Walker KK, Chen H, et al. The histone Heart Assoc 2019;8:e011484.
acetylase PCAF is a nuclear receptor coactivator. Genes Dev 1998;12: [54] Singh P, Brock CO, Volden PA, Hernandez K, Skor M, Kocherginsky M, et al.
1638e51. Glucocorticoid receptor ChIP-sequencing of subcutaneous fat reveals mod-
[27] Cryer PE, Gerich JE. Relevance of glucose counterregulatory systems to pa- ulation of inflammatory pathways. Obesity (Silver Spring) 2015;23:2286e93.
tients with diabetes: critical roles of glucagon and epinephrine. Diabetes [55] Kuo T, Lew MJ, Mayba O, Harris CA, Speed TP, Wang JC. Genome-wide
Care 1983;6:95e9. analysis of glucocorticoid receptor-binding sites in myotubes identifies gene
[28] Gorton LM, Khan AM, Bohland M, Sanchez-Watts G, Donovan CM, Watts AG. networks modulating insulin signaling. Proc Natl Acad Sci U S A 2012;109:
A role for the forebrain in mediating time-of-day differences in glucocorti- 11160e5.
coid counterregulatory responses to hypoglycemia in rats. Endocrinology [56] Polman JA, Welten JE, Bosch DS, de Jonge RT, Balog J, van der Maarel SM,
2007;148:6026e39. et al. A genome-wide signature of glucocorticoid receptor binding in
[29] Rhyu YA, Jang JY, Park S, An JH, Kim DL, Kim SK, et al. Impaired cortisol and neuronal PC12 cells. BMC Neurosci 2012;13:118.
growth hormone counterregulatory responses among severe hypoglycemic [57] Yu CY, Mayba O, Lee JV, Tran J, Harris C, Speed TP, et al. Genome-wide
patients with type 2 diabetes mellitus. Endocrinol Metab (Seoul) 2019;34: analysis of glucocorticoid receptor binding regions in adipocytes reveal gene
187e94. network involved in triglyceride homeostasis. PLoS One 2010;5:e15188.
[30] Macfarlane DP, Forbes S, Walker BR. Glucocorticoids and fatty acid meta- [58] Wang JC, Derynck MK, Nonaka DF, Khodabakhsh DB, Haqq C, Yamamoto KR.
bolism in humans: fuelling fat redistribution in the metabolic syndrome. Chromatin immunoprecipitation (ChIP) scanning identifies primary gluco-
J Endocrinol 2008;197:189e204. corticoid receptor target genes. Proc Natl Acad Sci U S A 2004;101:15603e8.
[31] Wang M. The role of glucocorticoid action in the pathophysiology of the [59] Reddy TE, Pauli F, Sprouse RO, Neff NF, Newberry KM, Garabedian MJ, et al.
Metabolic Syndrome. Nutr Metab (Lond) 2005;2:3. Genomic determination of the glucocorticoid response reveals unexpected
[32] de Guia RM, Rose AJ, Herzig S. Glucocorticoid hormones and energy ho- mechanisms of gene regulation. Genome Res 2009;19:2163e71.
meostasis. Horm Mol Biol Clin Invest 2014;19:117e28. [60] Sato H, Horikawa Y, Iizuka K, Sakurai N, Tanaka T, Shihara N, et al. Large-scale
[33] Munck A. Glucocorticoid inhibition of glucose uptake by peripheral tissues: analysis of glucocorticoid target genes in rat hypothalamus. J Neurochem
old and new evidence, molecular mechanisms, and physiological signifi- 2008;106:805e14.
cance. Perspect Biol Med 1971;14:265e9. [61] Phuc Le P, Friedman JR, Schug J, Brestelli JE, Parker JB, Bochkis IM, et al.
[34] Ngo S, Barry JB, Nisbet JC, Prins JB, Whitehead JP. Reduced phosphorylation Glucocorticoid receptor-dependent gene regulatory networks. PLoS Genet
of AS160 contributes to glucocorticoid-mediated inhibition of glucose uptake 2005;1:e16.
in human and murine adipocytes. Mol Cell Endocrinol 2009;302:33e40. [62] Vegiopoulos A, Herzig S. Glucocorticoids, metabolism and metabolic dis-
[35] Kawai A, Kuzuya N. On the role of glucocorticoid in glucose-induced insulin eases. Mol Cell Endocrinol 2007;275:43e61.
secretion. Horm Metab Res 1977;9:361e5. [63] Kino T, Hurt DE, Ichijo T, Nader N, Chrousos GP. Noncoding RNA gas5 is a
[36] Borelli MI, Garcia ME, Gomez Dumm CL, Gagliardino JJ. Glucocorticoid- growth arrest- and starvation-associated repressor of the glucocorticoid
induced changes in insulin secretion related to the metabolism and ultra- receptor. Sci Signal 2010;3:ra8.
structure of pancreatic islets. Horm Metab Res 1982;14:287e92. [64] Rainer J, Ploner C, Jesacher S, Ploner A, Eduardoff M, Mansha M, et al.
[37] Rafacho A, Marroqui L, Taboga SR, Abrantes JL, Silveira LR, Boschero AC, et al. Glucocorticoid-regulated microRNAs and mirtrons in acute lymphoblastic
Glucocorticoids in vivo induce both insulin hypersecretion and enhanced leukemia. Leukemia 2009;23:746e52.
glucose sensitivity of stimulus-secretion coupling in isolated rat islets. [65] Clayton SA, Jones SW, Kurowska-Stolarska M, Clark AR. The role of micro-
Endocrinology 2010;151:85e95. RNAs in glucocorticoid action. J Biol Chem 2018;293:1865e74.
[38] Wang JC, Gray NE, Kuo T, Harris CA. Regulation of triglyceride metabolism by [66] de Guia RM, Rose AJ, Sommerfeld A, Seibert O, Strzoda D, Zota A, et al.
glucocorticoid receptor. Cell Biosci 2012;2:19. microRNA-379 couples glucocorticoid hormones to dysfunctional lipid ho-
[39] Mandard S, Zandbergen F, van Straten E, Wahli W, Kuipers F, Muller M, et al. meostasis. EMBO J 2015;34:344e60.
The fasting-induced adipose factor/angiopoietin-like protein 4 is physically [67] Anagnostis P, Athyros VG, Tziomalos K, Karagiannis A, Mikhailidis DP. Clin-
associated with lipoproteins and governs plasma lipid levels and adiposity. ical review: the pathogenetic role of cortisol in the metabolic syndrome: a
J Biol Chem 2006;281:934e44. hypothesis. J Clin Endocrinol Metab 2009;94:2692e701.
[40] Chen TC, Lee RA, Tsai SL, Kanamaluru D, Gray NE, Yiv N, et al. An ANGPTL4- [68] Martins CS, Elias D, Colli LM, Couri CE, Souza MC, Moreira AC, et al. HPA axis
ceramide-protein kinase Czeta axis mediates chronic glucocorticoid dysregulation, NR3C1 polymorphisms and glucocorticoid receptor isoforms
exposure-induced hepatic steatosis and hypertriglyceridemia in mice. J Biol imbalance in metabolic syndrome. Diabetes Metab Res Rev 2017;33.
Chem 2019;294:9213e24. [69] Engeli S, Bohnke J, Feldpausch M, Gorzelniak K, Heintze U, Janke J, et al.
[41] Gray NE, Lam LN, Yang K, Zhou AY, Koliwad S, Wang JC. Angiopoietin-like 4 Regulation of 11beta-HSD genes in human adipose tissue: influence of
(Angptl4) protein is a physiological mediator of intracellular lipolysis in central obesity and weight loss. Obes Res 2004;12:9e17.
murine adipocytes. J Biol Chem 2017;292:16135. [70] Chen TC, Benjamin DI, Kuo T, Lee RA, Li ML, Mar DJ, et al. The glucocorticoid-
[42] Peckett AJ, Wright DC, Riddell MC. The effects of glucocorticoids on adipose Angptl4-ceramide axis induces insulin resistance through PP2A and PKCzeta.
tissue lipid metabolism. Metabolism 2011;60:1500e10. Sci Signal 2017;10.
[43] Koliwad SK, Gray NE, Wang JC. Angiopoietin-like 4 (Angptl4): a [71] Mueller KM, Hartmann K, Kaltenecker D, Vettorazzi S, Bauer M, Mauser L,
glucocorticoid-dependent gatekeeper of fatty acid flux during fasting. et al. Adipocyte glucocorticoid receptor deficiency attenuates aging- and
Adipocyte 2012;1:182e7. HFD-induced obesity and impairs the feeding-fasting transition. Diabetes
[44] Martin-Sanz P, Vance JE, Brindley DN. Stimulation of apolipoprotein secre- 2017;66:272e86.
tion in very-low-density and high-density lipoproteins from cultured rat [72] Phillips DI, Barker DJ, Fall CH, Seckl JR, Whorwood CB, Wood PJ, et al.
hepatocytes by dexamethasone. Biochem J 1990;271:575e83. Elevated plasma cortisol concentrations: a link between low birth weight
[45] Duerden JM, Bartlett SM, Gibbons GF. Long-term maintenance of high rates and the insulin resistance syndrome? J Clin Endocrinol Metab 1998;83:
of very-low-density-lipoprotein secretion in hepatocyte cultures. A model 757e60.
for studying the direct effects of insulin and insulin deficiency in vitro. [73] Walker BR, Phillips DI, Noon JP, Panarelli M, Andrew R, Edwards HV, et al.
Biochem J 1989;263:937e43. Increased glucocorticoid activity in men with cardiovascular risk factors.
[46] Cole TG, Wilcox HG, Heimberg M. Effects of adrenalectomy and dexameth- Hypertension 1998;31:891e5.
asone on hepatic lipid metabolism. J Lipid Res 1982;23:81e91. [74] Gadek-Michalska A, Spyrka J, Rachwalska P, Tadeusz J, Bugajski J. Influence of
[47] Wang CN, Hobman TC, Brindley DN. Degradation of apolipoprotein B in chronic stress on brain corticosteroid receptors and HPA axis activity.
cultured rat hepatocytes occurs in a post-endoplasmic reticulum compart- Pharmacol Rep 2013;65:1163e75.
ment. J Biol Chem 1995;270:24924e31. [75] Douyon L, Schteingart DE. Effect of obesity and starvation on thyroid hor-
[48] Hoekstra M, Korporaal SJ, Li Z, Zhao Y, Van Eck M, Van Berkel TJ. Plasma mone, growth hormone, and cortisol secretion. Endocrinol Metab Clin N Am
lipoproteins are required for both basal and stress-induced adrenal gluco- 2002;31:173e89.
corticoid synthesis and protection against endotoxemia in mice. Am J Physiol [76] Chalew SA, Zadik Z, Lozano RA, Nelson JC, Kowarski AA. Plasma cortisol
Endocrinol Metab 2010;299:E1038e43. levels increase with age in obese subjects. Obes Res 1993;1:199e202.
[49] van der Sluis RJ, Van Eck M, Hoekstra M. Adrenocortical LDL receptor func- [77] Manenschijn L, van Kruysbergen RG, de Jong FH, Koper JW, van Rossum EF.
tion negatively influences glucocorticoid output. J Endocrinol 2015;226: Shift work at young age is associated with elevated long-term cortisol levels
145e54. and body mass index. J Clin Endocrinol Metab 2011;96:E1862e5.
[50] Krausz Y, Bar-On H, Shafrir E. Origin and pattern of glucocorticoid-induced [78] Severino C, Brizzi P, Solinas A, Secchi G, Maioli M, Tonolo G. Low-dose
hyperlipidemia in rats. Dose-dependent bimodal changes in serum lipids dexamethasone in the rat: a model to study insulin resistance. Am J Physiol
and lipoproteins in relation to hepatic lipogenesis and tissue lipoprotein Endocrinol Metab 2002;283:E367e73.
1280 R.M. de Guia / Diabetes & Metabolic Syndrome: Clinical Research & Reviews 14 (2020) 1273e1280

[79] Hazlehurst JM, Gathercole LL, Nasiri M, Armstrong MJ, Borrows S, Yu J, et al. diabetes in db/db mice. Diabetes 2005;54:32e40.
Glucocorticoids fail to cause insulin resistance in human subcutaneous adi- [90] Klein J, Vuguin P, Hyman S. Cushing syndrome. Pediatr Rev 2014;35:405e7.
pose tissue in vivo. J Clin Endocrinol Metab 2013;98:1631e40. [91] Messazos BP, Zacharin MR. Lessons from iatrogenic Cushing syndrome in
[80] Geer EB, Islam J, Buettner C. Mechanisms of glucocorticoid-induced insulin children. J Paediatr Child Health 2016;52:1106e10.
resistance: focus on adipose tissue function and lipid metabolism. Endocrinol [92] Valassi E, Crespo I, Santos A, Webb SM. Clinical consequences of Cushing’s
Metab Clin N Am 2014;43:75e102. syndrome. Pituitary 2012;15:319e29.
[81] Ferris HA, Kahn CR. New mechanisms of glucocorticoid-induced insulin [93] Tempark T, Phatarakijnirund V, Chatproedprai S, Watcharasindhu S,
resistance: make no bones about it. J Clin Invest 2012;122:3854e7. Supornsilchai V, Wananukul S. Exogenous Cushing’s syndrome due to topical
[82] Brennan-Speranza TC, Henneicke H, Gasparini SJ, Blankenstein KI, corticosteroid application: case report and review literature. Endocrine
Heinevetter U, Cogger VC, et al. Osteoblasts mediate the adverse effects of 2010;38:328e34.
glucocorticoids on fuel metabolism. J Clin Invest 2012;122:4172e89. [94] Mancini T, Doga M, Mazziotti G, Giustina A. Cushing’s syndrome and bone.
[83] Parker L, Lin X, Garnham A, McConell G, Stepto NK, Hare DL, et al. Gluco- Pituitary 2004;7:249e52.
corticoid-induced insulin resistance in men is associated with suppressed [95] Chanson P, Salenave S. Metabolic syndrome in Cushing’s syndrome. Neuro-
undercarboxylated osteocalcin. J Bone Miner Res 2019;34:49e58. endocrinology 2010;92(Suppl 1):96e101.
[84] Saad MJ, Folli F, Kahn JA, Kahn CR. Modulation of insulin receptor, insulin [96] Napier C, Pearce SH. Autoimmune Addison’s disease. Presse Med 2012;41:
receptor substrate-1, and phosphatidylinositol 3-kinase in liver and muscle e626e35.
of dexamethasone-treated rats. J Clin Invest 1993;92:2065e72. [97] Mitchell AL, Pearce SH. Autoimmune Addison disease: pathophysiology and
[85] Paterson JM, Morton NM, Fievet C, Kenyon CJ, Holmes MC, Staels B, et al. genetic complexity. Nat Rev Endocrinol 2012;8:306e16.
Metabolic syndrome without obesity: hepatic overexpression of 11beta- [98] Tritos NA, Biller BM. Advances in medical therapies for Cushing’s syndrome.
hydroxysteroid dehydrogenase type 1 in transgenic mice. Proc Natl Acad Sci Discov Med 2012;13:171e9.
U S A 2004;101:7088e93. [99] Arnaldi G, Mancini T, Tirabassi G, Trementino L, Boscaro M. Advances in the
[86] Morton NM, Paterson JM, Masuzaki H, Holmes MC, Staels B, Fievet C, et al. epidemiology, pathogenesis, and management of Cushing’s syndrome com-
Novel adipose tissue-mediated resistance to diet-induced visceral obesity in plications. J Endocrinol Invest 2012;35:434e48.
11 beta-hydroxysteroid dehydrogenase type 1-deficient mice. Diabetes [100] Farrell C, O’Keane V. Epigenetics and the glucocorticoid receptor: a review of
2004;53:931e8. the implications in depression. Psychiatr Res 2016;242:349e56.
[87] Baudrand R, Carvajal CA, Riquelme A, Morales M, Solis N, Pizarro M, et al. [101] Quinn M, Ramamoorthy S, Cidlowski JA. Sexually dimorphic actions of glu-
Overexpression of 11beta-hydroxysteroid dehydrogenase type 1 in hepatic cocorticoids: beyond chromosomes and sex hormones. Ann N Y Acad Sci
and visceral adipose tissue is associated with metabolic disorders in 2014;1317:1e6.
morbidly obese patients. Obes Surg 2010;20:77e83. [102] Schroeder A, Notaras M, Du X, Hill RA. On the developmental timing of
[88] Hughes KA, Webster SP, Walker BR. 11-Beta-hydroxysteroid dehydrogenase stress: delineating sex-specific effects of stress across development on adult
type 1 (11beta-HSD1) inhibitors in type 2 diabetes mellitus and obesity. behavior. Brain Sci 2018;8.
Expet Opin Invest Drugs 2008;17:481e96. [103] Luo Y, Zeng B, Zeng L, Du X, Li B, Huo R, et al. Gut microbiota regulates mouse
[89] Liu Y, Nakagawa Y, Wang Y, Sakurai R, Tripathi PV, Lutfy K, et al. Increased behaviors through glucocorticoid receptor pathway genes in the hippo-
glucocorticoid receptor and 11{beta}-hydroxysteroid dehydrogenase type 1 campus. Transl Psychiatry 2018;8:187.
expression in hepatocytes may contribute to the phenotype of type 2

You might also like