ArticlePDF Available

A novel combined glucocorticoid-mineralocorticoid receptor selective modulator markedly prevents weight gain and fat mass expansion in mice fed a high-fat diet

Authors:

Abstract and Figures

Background: We have previously shown that antagonism of the mineralocorticoid receptor (MR) results in a potent antiadipogenic activity, in vitro and in vivo. Excessive glucocorticoid exposure is associated with obesity and related disorders in humans and mice. Methods: In this study responses to a novel combined glucocorticoid receptor (GR)/MR antagonist were investigated in a model of diet-induced obesity. Female 10-week-old C57BL/6J mice were fed with normal chow or a high fat diet (HFD) for 9 weeks. Mice fed a HFD were concomitantly treated for 9 weeks with the GR antagonist mifepristone (80 mg/kg/day) or the novel combined GR/MR antagonist CORT118335 (80 mg/kg/day). Male, juvenile 6-week-old C57BL/6J mice fed HFD were treated with CORT118335 for 4 weeks. Results: Mice fed a HFD showed a significant increase in total body weight and white fat mass, with impaired glucose tolerance and increased fat infiltration in livers. Interestingly, only CORT118335 completely prevented the HFD-induced weight gain and white fat deposition, whereas mifepristone showed no effect on body weight and modestly increased subcutaneous fat mass. Importantly, food intake was not affected by either treatment, and CORT118335 dramatically increased PGC-1α protein expression in adipose tissue, without any effect on UCP1. Both CORT118335 and mifepristone produced metabolic benefit, improving glucose tolerance, increasing adiponectin plasma levels, decreasing leptin and reducing mean adipocyte size. When tested in vitro, CORT118335 markedly reduced 3T3-L1 differentiation and reversed MR-mediated pro-adipogenic effects of aldosterone; differently, GR-mediated effects of dexamethasone were not antagonised by CORT118335, suggesting that it mostly acts as an antagonist of MR in cultured preadipocytes. Conclusions: Combined GR/MR pharmacological antagonism markedly reduced HFD-driven weight gain and fat mass expansion in mice through the increase in adipose PGC-1α, suggesting that both receptors represent strategic therapeutic targets to fight obesity. The effects of CORT118335 in adipocytes seem predominantly mediated by MR antagonism.International Journal of Obesity accepted article preview online, 02 February 2016. doi:10.1038/ijo.2016.13.
Content may be subject to copyright.
ORIGINAL ARTICLE
A novel combined glucocorticoid-mineralocorticoid
receptor selective modulator markedly prevents weight
gain and fat mass expansion in mice fed a high-fat diet
C Mammi
1
, V Marzolla
1
, A Armani
1
, A Feraco
1
, A Antelmi
1
, E Maslak
2
, S Chlopicki
2,3
, F Cinti
1,4
, H Hunt
5
, A Fabbri
6
and M Caprio
1,7
BACKGROUND: We have previously shown that antagonism of the mineralocorticoid receptor (MR) results in a potent
antiadipogenic activity, in vitro and in vivo. Excessive glucocorticoid exposure is associated with obesity and related disorders in
humans and mice.
METHODS: In this study, responses to a novel combined glucocorticoid receptor (GR)/MR antagonist were investigated in a model
of diet-induced obesity. Female 10-week-old C57BL/6J mice were fed with normal chow or a high-fat diet (HFD) for 9 weeks.
Mice fed a HFD were concomitantly treated for 9 weeks with the GR antagonist mifepristone (80 mg kg
1
per day) or the novel
combined GR/MR antagonist CORT118335 (80 mg kg
1
per day). Male, juvenile 6-week-old C57BL/6J mice fed HFD were treated
with CORT118335 for 4 weeks.
RESULTS: Mice fed a HFD showed a signicant increase in total body weight and white fat mass, with impaired glucose tolerance
and increased fat inltration in livers. Interestingly, only CORT118335 completely prevented the HFD-induced weight gain and
white fat deposition, whereas mifepristone showed no effect on body weight and modestly increased subcutaneous fat mass.
Importantly, food intake was not affected by either treatment, and CORT118335 dramatically increased PGC-1αprotein expression
in adipose tissue, without any effect on UCP1. Both CORT118335 and mifepristone produced metabolic benet, improving glucose
tolerance, increasing adiponectin plasma levels, decreasing leptin and reducing mean adipocyte size. When tested in vitro,
CORT118335 markedly reduced 3T3-L1 differentiation and reversed MR-mediated pro-adipogenic effects of aldosterone; differently,
GR-mediated effects of dexamethasone were not antagonized by CORT118335, suggesting that it mostly acts as an antagonist
of MR in cultured preadipocytes.
CONCLUSIONS: Combined GR/MR pharmacological antagonism markedly reduced HFD-driven weight gain and fat mass expansion
in mice through the increase in adipose PGC-1α, suggesting that both receptors represent strategic therapeutic targets to ght
obesity. The effects of CORT118335 in adipocytes seem predominantly mediated by MR antagonism.
International Journal of Obesity advance online publication, 22 March 2016; doi:10.1038/ijo.2016.13
INTRODUCTION
The incidence of obesity and type 2 diabetes has markedly
increased over recent decades and novel therapeutic strategies
seem to be necessary to ght such an epidemic. Unbalanced
corticosteroid metabolism in adipose tissue has a pivotal role in the
development of obesity and related cardiovascular and metabolic
disorders.
1,2
In fact, patients with glucocorticoid (GC) excess, such as
in Cushings Syndrome, develop insulin resistance and central
obesity, a phenotype characteristic of the metabolic syndrome.
3,4
Interestingly, treatment of obese Zucker rats, carrying a dysregula-
tion of 11 beta-hydroxysteroid dehydrogenase type 1, with the GC
receptor (GR) antagonist mifepristone or adrenalectomy reverses
the obese phenotype in these animals.
5,6
Notably, GCs regulate almost every aspect of adipose tissue
biology, from the early regulation of adipocyte differentiation
7
to
the ne control of lipolysis, glucose uptake and gluconeogenesis.
8
Hence, integrity of the HPA axis is of fundamental importance for
healthy metabolic homeostasis.
9
Interestingly, the effects of GCs in
adipose tissue are not exclusively mediated by the GR, but also by
the mineralocorticoid receptor (MR).
10
Recently, it has been found
that the MR is expressed in white adipocytes as well as in brown
adipocytes,
11,12
and has a key role in corticosteroid-induced
adipogenesis.
13,14
The MR binds not only to aldosterone but also
to GCs with equivalent afnity.
15
Moreover, the afnity of GCs
(cortisol in humans, corticosterone in rodents) for MR is 10-fold
higher than that for GR.
16
Importantly, GCs represent the major
MR ligand in adipocytes, and MR knockdown in primary murine
adipocytes as well as in 3T3-L1 cells blocks corticosteroid-induced
adipose differentiation.
13,17
In contrast to these observations,
recent data provide evidence that the GR has a more important
role than the MR in promoting the early steps of adipogenesis in
human preadipocytes.
18
However, it is clear that both receptors
have an important role in adipogenesis, and their reciprocal
interplay is crucial for adipocyte differentiation.
17
Benecial effects of MR antagonists have been demonstrated
in several animal models of genetic and diet-induced obesity. Guo
1
Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy;
2
Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland;
3
Chair of Pharmacology, Jagiellonian University, Medical College, Krakow, Poland;
4
Department of Experimental and Clinical Medicine, Center for Obesity, Università Politecnica
delle Marche, Ancona, Italy;
5
Corcept Therapeutics, Menlo Park, CA, USA;
6
Department of Systems Medicine, Endocrinology Unit, S. Eugenio & CTO A. Alesini Hospitals, University
Tor Vergata, Rome, Italy and
7
Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy. Correspondence: Professor M
Caprio, Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Via di Val Cannuta 247, Rome 00166, Italy.
E-mail: massimiliano.caprio@sanraffaele.it
Received 5 June 2015; revised 30 November 2015; accepted 27 December 2015; accepted article preview online 2 February 2016
International Journal of Obesity (2016), 19
© 2016 Macmillan Publishers Limited All rights reserved 0307-0565/16
www.nature.com/ijo
et al.
19
reported that systemic MR antagonism with eplerenone
reversed the adverse metabolic consequences of genetic obesity
in db/db mice, improving glucose tolerance, insulin resistance and
adipokine expression in adipose tissue. Similar effects were obtained
in a model of diet-induced obesity, in which MR blockade induced
a marked reduction of hypertrophic adipocytes and macrophage
inltration with a concomitant decrease of pro-inammatory
adipokine levels and excess of reactive oxygen species in adipose
tissue.
20
Importantly, our group recently showed that pharmaco-
logical MR antagonism with spironolactone was able to counter
metabolic dysfunctions in a model of HFD-induced obesity in
female mice, and that benecial effects on body fat composition
and glucose tolerance were associated with browningof adipose
tissue.
21
To date, the exact role, interaction and relative importance
of GR vs MR in murine adipogenesis as well as in adipose tissue
expansion during a high-fat challenge is still controversial.
For these reasons, we explored adipose tissue responses to the
novel combined GR/MR antagonist CORT118335 in a model of
diet-induced obesity in mice. CORT118335 displays eightfold
selectivity for GR over MR, as assessed in reporter gene assays.
22
MATERIALS AND METHODS
Animal model
Animal procedures were approved by the Italian National Institute of
Health Care and Use Committees. Female 10-week-old (study 1) and male
6-week-old (study 2) C57BL/6J mice were purchased from Charles River
Laboratories, Calco (LC), Italy. Mice were divided into four groups (n= 10)
as follows: (i) ND: mice fed a normal diet (10% kcal as fat; D12450B;
Research Diets, New Brunswick, NJ, USA) plus vehicle by oral gavage;
(ii) HFD: mice fed a high-fat diet (HFD; 45% kcal as fat; D12451; Research
Diets) plus vehicle by oral gavage; (iii) mifepristone: mice fed a HFD plus
mifepristone (80 mg kg
1
per day) by oral gavage; (iv) CORT118335: mice
fed a HFD, plus CORT 118335 (80 mg kg
1
per day) by oral gavage.
Glucose tolerance assessment
After 8 weeks of treatment, animals from study 1 (n= 10) were fasted for
6 h and a tail vein blood sample was collected to measure basal blood
glucose levels.
The animals were then injected with a glucose solution into the peritoneum
(0.09 g ml
1
glucose, 112 μl solution per 10 g body weight) and blood
glucose concentrations measured using a commercial glucometer (Contour
XT, BAYER Healthcare, Leverkusen, Germany) over the next 2 h at 20-min
intervals for the rst hour and at 30-min intervals in the second hour.
Body weight, food intake, caloric efciency and adipokine
secretion assessment
Total body weight was measured weekly and when the mice were killed
(day 63 for study 1 or day 28 for study 2). We assessed the wet weight
of dissected inguinal subcutaneous and visceral abdominopelvic fat
depots, as well as tibia and femur length. Food intake was measured
each week throughout the study in mice (n= 6) housed in individual cages,
by providing a xed amount of food and weighing uneaten food for ve
consecutive days. Quantication of food intake and change in body weight
over the 63-day treatment period were used to evaluate caloric efciency,
which is expressed as body weight gain per unit of energy intake.
23
At the
time the mice were killed, peripheral blood samples were collected via
retro-orbital bleeding. Leptin and adiponectin serum levels were measured
by a commercially available ELISA kit (RD System, Minneapolis, MN, USA),
sodium and potassium were measured by Hitachi Cobas C 311 analyzer
(Roche Diagnostic Ltd, Rotkreuz, Switzerland).
Gene expression analysis
Total RNA from fat depots was isolated using RNeasy lipid tissue mini kit
(Qiagen, Milan, Italy) following the manufacturers instructions. Total
RNA from skeletal muscles (tibialis anterior) and adipocyte cultures was
isolated in Trizol reagent (Life Technologies, Milan, Italy) according to the
manufacturers recommendations. Preparation of cDNA and real-time
qPCR were performed as previously described.
21
See Supplementary Table
1 for primer sequences.
Micro-CT studies
The femur samples from juvenile male mice (n= 4, study 2) were analyzed,
after careful dissection, with a microcomputed cone-beam X-ray system
(Skyscan 1072 μ-CT, Kartuizersweg 3B, 2550 Kontich, Belgium) without
addition of contrasting agent. Image reconstruction and analysis were
conducted using the software package provided by Skyscan, as already
described.
24
For each sample, an internal region of interest was selected
and histomorphometric parameters for the corresponding volume of
interest were calculated from the most proximal point of the trochanter
and were extended proximally for 4 mm; then, the volume of the cortical
bone of femur neck was analyzed.
Cell culture studies and Oil Red O staining
Murine 3T3-L1 preadipocytes were grown until conuence at 37 °C
in Dulbecco's modied Eagle's medium (Invitrogen, Milan, Italy) containing
4.5 g l
1
D-glucose, 10% fetal calf serum (Invitrogen), 100 U ml
1
penicillin
and 100 μgml
1
streptomycin. To achieve adipogenic differentiation
in the absence of serum steroids, conuent cells were transferred in
Dulbecco's modied Eagle's medium containing 10% dextran-coated
charcoal-stripped fetal calf serum, and treated as previously reported.
13
Aldosterone (10
8
M) or dexamethasone (10
7
M) were added from day 2
of differentiation, with or without CORT118335 (10
6
M).
Primary cultures of murine preadipocytes derived from the vascular
stromal fraction of inguinal fat depots of 10-week-old female C57/BL6J
were prepared as previously described.
25
Cells were allowed to differ-
entiate until day 6. Differentiating cells were treated from conuence until
day 6 with spironolactone (10
5
M), mifepristone (10
5
M) or CORT118335
(10
6
M). Unless otherwise indicated, all chemicals were obtained from
Sigma-Aldrich (Milan, Italy). For determination of lipid content, cells were
processed as previously published.
26
To quantify the lipid content, the
stained cells were permeabilized with 10% sodium dodecyl sulfate and the
eluate was measured using a spectrophotometer at 520 nm. For every cell
culture study, two independent experiments were performed in triplicate.
Western blot analysis
Specimens of inguinal subcutaneous fat (n= 6) were lysed in HNTG lysis
buffer containing 1% Triton X-100, 50 mMHepes, 10% glycerol, 150 mM
NaCl, 1 mMNaVO4 and 75 U of aprotinin and allowed to stand for 30 min.
After sodium dodecyl sulfate-polyacrylamide gel electrophoresis and
blotting,
21
membranes were probed with rabbit polyclonal anti-uncoupling
protein 1 (UCP1, Abcam 23841, Cambridge, MA, USA), rabbit polyclonal
anti-peroxisome proliferator-activated receptor gamma coactivator 1-alpha
(PGC-1α, Santa Cruz Biotechnology sc-13067, Heidelberg, Germany) or goat
polyclonal anti-actin (Santa Cruz Biotechnology sc-1615, Heidelberg,
Germany). Bound antibodies were visualized with horseradish peroxidase-
conjugated anti-rabbit IgG (Sigma-Aldrich) and immunoreactivity assessed
by chemiluminescence reaction, using the ECL Western detection system
(General Electric Healthcare, Milan, Italy). Densitometric scanning analysis
was performed by Mac OS X (Apple Computer International, Milan, Italy)
using NIH Image 1.62 software.
Histological analysis of fat depots and livers
Dissected adipose tissue depots (n= 6) were xed overnight in 10%
formaldehyde at 4 °C and rinsed with 0.1M phosphate buffer before
embedding in parafn. Each parafn-embedded depot was cut (3-μm
thick) and the cutting plane corresponding to the largest surface was used
for histological examination, morphometry (hematoxylin-eosin). Ten elds
at × 40 nal magnication for each slide were analyzed to determine mean
adipocyte area using a digital image system (Lucia imaging, version 4.82,
Praha, Czech Republic).
Liver samples (n=6)after xation in formalin solution were cryoprotected
by immersion in 30% sucrose solution overnight and afterwards embedded
in Tissue-Tek OCT (optimum cutting temperature) gel, frozen at 80 °C and
cut into 7-μm slides. Prepared slides were stained using the Oil Red O
method for fat deposition. The Oil Red O sections were photographed under
× 100 magnication. At least six images of each section were randomly
obtained. The images were subsequently analyzed by using Columbus
Image Data Storage and Analysis System (Perkin Elmer, Boston, MA, USA),
with an algorithm adapted for Oil Red O-stained sections.
GR/MR antagonism in adipose tissue
C Mammi et al
2
International Journal of Obesity (2016), 1 9 © 2016 Macmillan Publishers Limited
Statistical analysis
Ten mice per group were required to allow the assessment for primary
outcome (total body weight) of a standardized difference among any
pair of means of at least 0.65 s.d. at the signicance level alpha = 0.05
(two-tailed test), with power (1beta) = 0.80.
Data are reported as the mean +/ s.e.m. Data points greater or less than
two standard deviations from the mean were considered statistical outliers
and were excluded from all analyses. Statistical comparisons were made
by one- or two-way analysis of variance, followed by Bonferroni multiple
comparison post hoc analysis, using Prism 6.0 (GraphPad, San Diego, CA,
USA). Po0.05 was considered signicant.
RESULTS
Treatment with CORT118335 protects from HFD-induced weight
gain and fat storage, whereas mifepristone shows no effect on
body weight and fat mass
In study 1, female mice were fed a diet with 45% calories from fat,
which is similar in macronutrient composition to a typical western
diet,
27
and treated daily with vehicle, CORT118335 or mifepristone
for 9 weeks. Mice fed a HFD showed an increase in total body
weight of approximately 4 g, as compared with the ND group
(Figure 1a), with a parallel increase in the weight of inguinal and
abdominopelvic fat pads. Treatment with CORT118335 dramati-
cally attenuated the HFD-induced body weight gain (Figure 1a,
Po0.001 vs HFD), preventing the increase in fat pad weight
(Figure 1b); this effect was already signicant after 2 weeks
of treatment with CORT118335. In contrast, mifepristone had no
effect on total body weight (Figure 1a) and abdominopelvic fat
pad weight (Figure 1b, lower panel), and modestly increased
subcutaneous fat (Figure 1b, upper panel). Average kilocalorie
intake was not modied by mifepristone or CORT118335 at any
time point of the study (Figure 1c); interestingly, caloric efciency
was dramatically reduced only in the CORT118335 group, when
compared with the HFD group (Figure 1d), suggesting a different
metabolic prole in mice treated with the combined GR/MR
antagonist. To conrm these observations in the male gender, and
to exclude any potential adverse effects of CORT118335 on
growth, skeletal muscle and bone quality, we performed a shorter
study (4 weeks, study 2) on juvenile male mice. CORT118335
treatment did not affect body growth, tibia and femur length
(data not shown), but markedly blunted HFD-induced weight
gain; this effect was signicant after only 1 week of treatment
(Figure 2a). The marked difference in total body weight observed
at the end of the study was due to a dramatic reduction in visceral
and subcutaneous fat depots in treated mice, compared with
controls (Figure 2b). Importantly, transcript expression of Myosin
Heavy Chain and MyoD, two markers of skeletal muscle
differentiation, and atrogin-1 (a marker of muscular atrophy) in
Figure 1. CORT118335 counters body weight gain in female mice, prevents the increase in fat pad weight from inguinal and abdominopelvic
depots, and reduces caloric efciency in mice fed a HFD. (a) Body weight gain of female 10-week-old C57BL/6J mice during a 63-day period of ND or
HFD, treated or not with mifepristone (MIFE) or CORT118335 (Cort335). (b) Wet weight percentage increase vs ND of subcutaneous (inguinal, upper
panel) and visceral (abdominopelvic, lower panel) depots from mifepristone and CORT118335-treated mice, as compared with the HFD group
(n=10). (c) Average kilocaloric intake was measured in mice housed in individual cages (n=6pergroup).(d)Caloricefciency calculated over the
entire study period (n=6). Values are expressed as means ±s.e.m.
#
Po0.05,
##
Po0.01 vs ND group; *Po0.05, **Po0.01, ***Po0.001 vs HFD group.
GR/MR antagonism in adipose tissue
C Mammi et al
3
© 2016 Macmillan Publishers Limited International Journal of Obesity (2016), 1 9
tibialis anterior were not modied by treatment with CORT118335.
Moreover, trabecular thickness and cortical bone volume were not
affected by CORT118335, as shown by micro-CT analysis of femurs
(Figure 2d). Interestingly, porosity of trabecular bone was even
slightly decreased in CORT118335-treated animals, thus excluding
a potential adverse effect of the compound on bone develop-
ment. CORT118335 did not alter Na and K plasma levels (data not
shown) and treated mice never displayed any sign or symptom
suggestive of adrenal insufciency, throughout the study.
Treatment with CORT118335 improves glucose tolerance and
adipokine secretion prole in HFD-fed mice
After 8 weeks of treatment (study 1), an intraperitoneal glucose
tolerance test was performed. Before injection, CORT118335- and
mifepristone-treated groups showed lower plasma glucose levels
compared with untreated HFD mice. Interestingly, 20 min after the
intraperitoneal injection of glucose, CORT118335-treated mice
showed a marked reduction in plasma glucose levels as compared
with the HFD group (Figure 3a), which led to a signicant decrease
in the area under the curve of plasma glucose (Figure 3b).
In accordance with previous studies,
28
mifepristone-treated mice
also showed a signicant improvement in glucose tolerance,
which was slightly delayed compared with CORT118335 (Figure 3a),
but demonstrated a similar reduction in plasma glucose area under
the curve (Figure 3b). Accordingly, both treatments signicantly
increased plasma adiponectin levels (Po0.001, Figure 3c), and
concomitantly decreased leptin levels (Po0.001, Figure 3d), as
compared with the HFD group.
Treatment with CORT118335 prevents liver steatosis and
adipocyte hypertrophy in HFD-fed mice, but does not increase
brown adipocyte content in WAT
To gain more insights in the favorable metabolic effects of
CORT118335, we studied its effects on HFD-induced liver steatosis.
As expected, histological analysis of Oil Red O stained liver lipid
droplets showed moderate, microvesicular steatosis within the
liver of HFD-treated mice. Treatment with CORT118335 signi-
cantly reduced liver steatosis (Po0.05; Figures 4a and b), similarly
to results observed for mifepristone, as previously reported.
28,29
As expected, the average adipocyte cross-sectional area of the
inguinal and abdominopelvic depots was signicantly larger in the
HFD group compared with the ND group (data not shown). Cell
Figure 2. CORT118335 counters body weight gain in male juvenile mice, prevents the increase in abdominopelvic fat depots, without any
adverse effect on growth, skeletal muscle and bone quality. (a) Body weight gain of male 6-week-old C57BL/6J mice during a 28-day period
of ND or HFD, treated or not with CORT118335 (Cort335). (b) Exposed abdominopelvic and inguinal fat of two representative mice at the end
of the study. (c) qRT-PCR analysis of genes involved in skeletal muscle differentiation (MyHC and MyoD) and atrophy pathway (Atrogin-1)
performed in tibialis anterior of male 6-week-old mice fed a HFD, treated or not with CORT118335. (d) micro-CT analysis of femurs dissected
after killing the mice in study 2. The panel represents the following structural parameters: Tissue Volume (total volume of the volume-of-
interest), Trabecular thickness, Porosity and Cortical Bone Volume.
GR/MR antagonism in adipose tissue
C Mammi et al
4
International Journal of Obesity (2016), 1 9 © 2016 Macmillan Publishers Limited
size distribution analysis, performed in both depots, showed
a signicant reduction of cross-sectional area of adipocytes of
mice treated with either CORT118335 or mifepristone, as
compared with the HFD group (Figure 4c, upper panels).
In mice, browning of subcutaneous WAT has been observed to
improve metabolic prole, protecting from diet-induced obesity.
30
To determine whether alterations in brown fat content con-
tributed to the protective effect of both treatments on glucose
tolerance and body composition, we investigated the occurrence
of browning in hematoxylin-eosin-stained sections of inguinal
subcutaneous fat depots; we did not, however, observe any increase
in the number of brown adipocytes in either of the treated groups
(Figure4c,lowerpanel).Thesedatawereconrmed by western blot
analysis for UCP1 in subcutaneous fat, which did not show any
increase in UCP1 protein expression, by either CORT118335 or
mifepristone treatment (Figures 5a and c). These data indicate that
CORT118335 does not induce browning of white adipose tissue, in
contrast to observations with selective MR antagonists, as recently
published.
21
Treatment with CORT118335 markedly increases adipose PGC-1α
expression
Importantly, treatment with CORT118335 induced a dramatic
increase in PGC-1αprotein expression in subcutaneous fat, which
was also observed, although at a lower extent, in mice treated
with mifepristone (Figures 5a and b). Accordingly, both com-
pounds were able to upregulate transcript expression of PGC-1αin
primary cultured murine preadipocytes (Figure 5g).
We recently demonstrated that selective antagonism of adipocyte
MR determines browning of white adipose tissue, in vivo and
in vitro.
21
To explore whether MR antagonistic properties displayed
by CORT118335 were able to affect browning, murine preadipocytes
isolated from the stroma-vascular fraction of the inguinal sub-
cutaneous fat depot were differentiated in the absence or in the
presence of spironolactone (10
5
M), mifepristone (10
5
M) or
CORT118335 (10
6
M) for 6 days. As previously shown,
21
specicMR
antagonism by spironolactone signicantly increased UCP1,
PRDM16, CIDEA and PGC-1-αtranscript levels (Figures 5dg).
Interestingly, CORT118335 only modestly increased UCP1 gene
expression (Po0.05), whereas it did not modify the expression of
PRDM16 and CIDEA mRNA. None of the brown adipocyte-specic
transcripts were affected by GR antagonism with mifepristone.
Taken together, these data suggest that the modest MR antagonism
displayed by CORT118335 was not able to fully trigger the brown
adipogenic gene program. This is conrmed by the lack of increase
in UCP1 protein, observed in vivo, in the adipose tissue of mice
treated with CORT118335 (Figure 5a). We can therefore hypothesize
that the metabolically benecial effects of CORT118335 can be
explained by the increase in adipose tissue protein expression of
PGC-1α, a master regulator of mitochondrial biogenesis, rather than
browning of white adipose tissue.
CORT118335 inhibits lipids accumulation of 3T3-L1 preadipocytes
in vitro, mainly through MR antagonism
To further explore a potential direct role of adipocyte MR and GR in
the metabolic effects of the compound in vivo, we investigated the
effects of CORT118335 (10
6
M) in 3T3-L1 preadipocyte differentia-
tion. Treatment began when cells reached conuence and was
maintained for the entire duration of adipocyte differentiation
(8 days).
26
CORT118335 markedly reduced lipid accumulation as
determined by Oil Red O staining (Po0.001, Figure 6a). The
inhibitory effects of CORT118335 were observed also at concentra-
tions of 10
7
M, were maximal at 10
6
M and did not increase at
10
5
M (data not shown). As expected, mifepristone also showed a
marked inhibition of adipocyte differentiation. We next dissected
the mixed GR/MR antagonist activity of CORT118335 in this model.
3T3-L1 cells were maintained in a steroid-depleted medium from
conuence, omitting dexamethasone from the adipogenic cocktail
during induction of differentiation (day 02), and treated from day 2
with aldosterone (10
8
M) or dexamethasone (10
7
M), in the
presence or in the absence of CORT118335 (10
6
M). Under these
culture conditions, aldosterone and dexamethasone are known to
Figure 3. CORT118335 treatment improves glucose tolerance, increases plasma adiponectin levels and markedly reduces plasma leptin, as
compared with the HFD group. (a) Plasma glucose levels during an intraperitoneal glucose tolerance test following food withdrawal for 6 h
(n=10) in female 10-week-old C57BL/6J mice fed a HFD in the presence or absence of mifepristone or CORT118335 for 63 days. (b) Area under
the curve (AUC) glucose was calculated using the trapezoidal rule (n=10). (c) Measurement of plasma adiponectin and (d) leptin levels in all
groups of mice before killing after fasting for 6 h (n=10). Values are expressed as means ±s.e.m. ***Po0.001 vs HFD group.
GR/MR antagonism in adipose tissue
C Mammi et al
5
© 2016 Macmillan Publishers Limited International Journal of Obesity (2016), 1 9
display opposite effects on adipocyte differentiation.
13
We then
measured Oil Red O accumulation at day 8, and mRNA levels for
leptin and resistin, which are selectively activated by aldosterone
13
and angiotensinogen, which is specically induced by dexametha-
sone through GR. As expected, aldosterone signicantly increased
adipose differentiation (Figure 6b), and its effects were selectively
countered by CORT118335, indicating functional MR antagonism. As
previously shown,
13
dexamethasone displayed a GR-mediated
inhibitory effect on adipose differentiation, which was not reversed
by CORT118335 (Figure 6b), whereas coincubation with mifepris-
tone was able to counter the inhibitory action of dexamethasone,
thereby favoring adipose differentiation (data not shown). More-
over, we found that treatment with aldosterone increased gene
expression of leptin and resistin and co-treatment with CORT118335
signicantly inhibited this response (Po0.001 and Po0.05,
respectively, Figures 6c and d). Treatment with dexamethasone
decreased leptin and resistin mRNA levels, and co-treatment with
CORT118335 did not prevent this effect (Figures 6c and d). To
further investigate whether CORT118335 exhibited antagonistic
effects on adipocyte GR, we analyzed transcript levels of
angiotensinogen, a well-known GC target gene.
13
Importantly, co-
treatment with CORT118335 did not prevent the upregulation of
angiotensinogen mRNA by dexamethasone (Figure 6e), suggesting
that, in vitro, CORT118335 affects adipocyte function mainly through
MR antagonism.
DISCUSSION
In the present study, we investigated for the rst time the effects
of a novel combined MR/GR antagonist in a validated model of
HFD-induced obesity in mice, and showed a powerful inhibition of
fat mass expansion that was associated with benecial metabolic
effects on glucose tolerance, serum adipokines and liver steatosis.
Importantly, the compound did not cause any sign or symptom
suggestive of adrenal insufciency, nor altered growth, skeletal
muscle and bone quality, when administered to juvenile growing
male mice. GR antagonism with mifepristone had no effect on
body weight and fat deposition, even though mifepristone still
exhibited favorable metabolic effects. Our data conrm previous
results, showing that mifepristone improved insulin sensitivity
but did not affect body weight and fat mass in a model of HFD-
induced obesity in mice.
28
Importantly, the concomitant antagon-
ism of the MR, provided by the novel compound, demonstrated a
dramatic inhibition of adipose mass expansion, driven by a HFD.
We have recently shown that pharmacological antagonism
of the MR, obtained with spironolactone and drospirenone,
countered weight gain, as well as the increase in WAT mass
expansion and impaired glucose tolerance induced by a HFD in
mice.
21
Therefore, we hypothesize that the pharmacological MR
antagonistic properties of CORT118335 have an important role in
the marked inhibition of HFD-driven weight gain and white fat
mass expansion observed in mice treated with this compound.
Interestingly, the combined antagonism of GR and MR completely
Figure 4. Treatment with CORT118335 and mifepristone prevents liver steatosis and adipocyte hypertrophy in HFD-fed mice.(a)
Representative histological sections of livers obtained from six animals in each treatment condition, stained by Oil Red O; scale
bars =50 μm. (b) Percentage of Oil Red O-stained area, as compared with the HFD group (n=6). (c) Representative sections of inguinal fat
depots of all groups (n=6), stained with hematoxylin-eosin. Scale bars =40 μm(upperpanels);scalebar=200 μm(lowerpanels).(d)Mean
cross-sectional area of adipocytes in inguinal fat depots from all groups. Values are expressed as means ±s.e.m. *Po0.05, ***Po0.001 vs
HFD group.
GR/MR antagonism in adipose tissue
C Mammi et al
6
International Journal of Obesity (2016), 1 9 © 2016 Macmillan Publishers Limited
prevented HFD-induced increase in body weight and fat mass, at a
greater extent to those observed with specic MR antagonism by
spironolactone and drospirenone.
21
In fact, CORT118335-treated
mice showed a marked difference in total body weight, as
compared with vehicle-treated, after only 2 weeks of treatment,
whereas at least 9 weeks of treatment with MR antagonists were
required to obtain a signicant effect on body weight gain.
21
Moreover, CORT118335-treated mice never showed any signi-
cant increase in total body weight induced by HFD, throughout
the study. The marked difference in total body weight was mostly
due to fat mass, as showed by the dramatic effect of the
compound on both visceral and subcutaneous fat pads weight.
Importantly, such effects were not due to reduced food intake,
which was unchanged among treatment groups throughout the
study. Caloric efciency, estimated as body weight gain per unit of
energy intake, which represents a rough estimation of energy
expenditure,
23
was dramatically lower in mice treated with
CORT118335, as compared with the HFD and mifepristone groups.
This suggests a marked increase in energy expenditure in mice
treated with the combined GR/MR antagonist. The lack of
metabolic studies including energy expenditure through calori-
metry is a limitation of the present study. However, our results
suggest that combined GR/MR antagonism improves the meta-
bolic activity of mice and has a profound impact on adiposity,
glucose tolerance and resistance to HFD.
White fat mass reduction in CORT118335-treated mice was
associated with a signicant improvement in glucose tolerance after
an intraperitoneal glucose tolerance test, higher levels of plasma
adiponectin and reduced concentrations of leptin. Several mechan-
isms may contribute to the improvement in glucose tolerance
induced by CORT118335. Our study showed that CORT118335 was
also able to counteract the dysfunctional enlargement of adipocyte
size and the subsequent altered adipokine expression induced by
HFD. Therefore, CORT118335 may indirectly affect blood glucose
disposal through the increase in adiponectin and the decrease in
leptin secretion by adipose tissue.
19,20,31,32
The metabolic benets observed in mice treated with both
CORT118335 and mifepristone were conrmed by the protection
from liver steatosis induced by the HFD. Our data conrm previous
results showing that mifepristone improved liver injury,
28
and
indicate that the MR antagonistic activity of CORT118335 does
not add benet to the protection from liver steatosis obtained
with GR antagonism, in contrast to the effects observed for white
fat mass expansion.
Figure 5. CORT118335 increases adipose PGC-1αprotein expression in vivo, without any major effects on UCP1 protein and brown adipose
tissue (BAT)-specic transcripts in vitro. CORT118335 treatment in vivo induces a dramatic increase of adipose tissue PGC-1αprotein
expression, without any major effect on UCP1. (a) Western blot analysis performed in subcutaneous adipose depots of mice fed a HFD, treated
with mifepristone or CORT118335, by using polyclonal antibodies to PGC-1αor UCP1. A representative experiment among three is shown.
(b) Densitometric analysis for PGC-1αand (c) UCP1 (using actin as loading control). (dg) qRT-PCR analysis of genes involved in brown
adipogenesis in primary murine preadipocytes from inguinal fat depot differentiated for 6 days with or without spironolactone (SPIRO,
10
5
M), mifepristone (10
5
M) or CORT118335 (10
6
M). Values are expressed as % of control untreated cells and represent the mean ±s.e.m. of
two independent experiments performed in triplicate. CORT118335 weakly upregulates UCP1 transcript levels (d), without any effect on
PRDM16 (e) and CIDEA (f) mRNA. (g) CORT118335 and mifepristone markedly increase PGC-1αmRNA expression. Values are expressed as
means ±s.e.m. *Po0.05, **Po0.01, ***Po0.001 vs UT.
GR/MR antagonism in adipose tissue
C Mammi et al
7
© 2016 Macmillan Publishers Limited International Journal of Obesity (2016), 1 9
Importantly, histological examination of inguinal fat samples from
CORT118335-treated mice did not reveal any increase in brown
adipocyte content, as we recently observed in mice treated with
spironolactone.
21
In line with this, western blot analysis failed to
show any increase of UCP1 expression in inguinal fat of mice treated
with CORT118335. We have recently shown that pharmacological
MR blockade determines browning of the adipose organ.
21
These
effects were not observed with the GR antagonist mifepristone, nor
with CORT118335, probably because of the moderate antagonistic
activity of the compound at MR, as compared with spironolactone.
22
This could explain a reduced ability of CORT118335 in inducing
UCP1 protein expression in WAT. This is also conrmed by the lack
of major effects exerted by CORT118335 on brown adipose tissue-
specic transcripts in primary preadipocytes in vitro,suchas
PRDM16 and CIDEA, while UCP1 mRNA was only modestly
increased by CORT118335.
Conversely, we observed a dramatic increase of PGC-1αprotein
expression in inguinal fat from mice treated with CORT118335.
These data show that combined MR/GR blockade causes a marked
increase in adipose PGC-1αprotein, enhancing the effects obtained
with specic GR antagonism by mifepristone. PGC-1αis a
transcriptional coactivator and master regulator of mitochondrial
biogenesis.
33
We hypothesize that the marked upregulation of
adipose PGC-1αrepresents one of the leading mechanisms
explaining the effects of the novel compound on body fat. In fact,
recent ndings suggest that adipose tissue PGC-1αhas an important
role in glucose homeostasis and fatty acid oxidation,
34
and that the
adipose organ in several murine models of obesity shows a reduced
mitochondrial function.
35
PGC-1αis a master gene linking mito-
chondrial number and function to external environmental and
hormonal stimuli,
33
and directly controls oxygen consumption,
oxidative phosphorylation, uncoupling and mitochondrial prolifera-
tion. In our model, we did not observe any increase in UCP1 protein
in any of the studied adipose depots, suggesting that the marked
increase in adipocyte PGC-1αregulates the biogenesis and electron
transport systems of mitochondria in WAT, without stimulating
uncoupling, hence without induction of browning. A limitation of
the present work stems from the systemic effects of the
pharmacological receptor blockade, which affects the activity of
GR and MR expressed not only in adipocytes but also in other cell
types (pancreatic beta cells, skeletal muscle cells, neurons). There-
fore, we cannot exclude the possibility that receptor blockade in
tissues other than fat may generate signals to control the metabolic
response to a HFD. For this reason, we treated in vitro primary
murine preadipocytes and 3T3-L1 cells with CORT118335, conrm-
ing its direct effects on adipose tissue biology, and suggesting
that adipose tissue may represent a direct target for CORT118335
in vivo. CORT118335, in fact, markedly reduced 3T3-L1 lipid
accumulation as measured by Oil Red O staining. To dissect
the mixed GR/MR antagonist activity of CORT118335 in this
model, we tested its effects under steroid-deprived conditions in
3T3-L1 cells.
13
Interestingly, only the MR-mediated pro-adipogenic
effects of aldosterone were antagonized by CORT118335, whereas
GR-mediated effects of dexamethasone were not affected. This
suggests that CORT118335 mostly acts as an antagonist of MR,
rather than GR, in murine cultured preadipocytes, and highlights the
relative importance of MR signaling as a strategic target to modulate
adipocyte differentiation. On the basis of our data, we can infer that
reduction in white fat mass and induction of browning may not
necessarily occur together, in response to pharmacological MR
antagonism.
Figure 6. CORT118335 inhibits adipose differentiation of 3T3-L1 preadipocytes in vitro, mainly through MR antagonism. (a) Both CORT118335
and mifepristone markedly reduce 3T3-L1 differentiation, as measured by Oil Red O staining. (b) Under steroid-deprived conditions,
CORT118335 counters adipose differentiation induced by aldosterone but does not reverse dexamethasone (DEXA)-driven inhibitory action
on adipose conversion. CORT118335 counters the increase in transcript levels of leptin (c) and resistin (d) stimulated by aldosterone, but does
not prevent the upregulation of angiotensinogen mRNA (e) induced by dexamethasone. Values are expressed as means ±s.e.m. **Po0.01,
***Po0.001 vs UT;
#
Po0.05;
###
Po0.001 vs ALDO.
GR/MR antagonism in adipose tissue
C Mammi et al
8
International Journal of Obesity (2016), 1 9 © 2016 Macmillan Publishers Limited
In conclusion, this is the rst study showing that combined
GR/MR pharmacological blockade markedly prevents fat mass
expansion in a model of HFD-induced obesity both in female and
male mice, conrming that both MR and GR have a relevant role in
adipose tissue function, and that such combined pharmacological
approach could represent a novel strategy to prevent HFD-induced
obesity and its metabolic complications.
CONFLICT OF INTEREST
Dr Hazel Hunt is employed by CORCEPT Therapeutics, which developed and provided
CORT118335 and mifepristone for the experiments. All the other authors declare no
conict of interest.
ACKNOWLEDGEMENTS
We thank Dr Raffaella Pecci and Dr Rossella Bedini for precious technical assistance,
Prof Silvia Migliaccio and Prof Paolo Bianco for scientic support. This work
was supported by a grant from Ministero della Salute (BANDO 2011-2012 Progetti
Collaborazione Ricercatori Italiani allEstero; project grant PE-2011-02347070 to MC),
and from Corcept Therapeutics. This work was presented, in part, at the
COST-ADMIRE Annual Meeting, Padua, Italy, 16th17th October 2014. We would
like to acknowledge networking support by the COST Action ADMIRE BM1301. This
work is dedicated to the memory of Prof Paolo Bianco, outstanding scientist, shining
example of dedication to work and freedom of thought.
REFERENCES
1 Dallman MF, Pecoraro N, Akana SF, La Fleur SE, Gomez F, Houshyar H et al.
Chronic stress and obesity: a new view of "comfort food". Proc Natl Acad Sci USA
2003; 100: 1169611701.
2 Warne JP, Akana SF, Ginsberg AB, Horneman HF, Pecoraro NC, Dallman MF.
Disengaging insulin from corticosterone: roles of each on energy intake and
disposition. Am J Physiol Regul Integr Comp Physiol 2009; 296: R1366R1375.
3 Geer EB, Shen W, Gallagher D, Punyanitya M, Looker HC, Post KD et al. MRI
assessment of lean and adipose tissue distribution in female patients with
Cushing's disease. Clin Endocrinol (Oxf) 2010; 73:469475.
4 Geer EB, Shen W, Strohmayer E, Post KD, Freda PU. Body composition and car-
diovascular risk markers after remission of Cushing's disease: a prospective study
using whole-body MRI. J Clin Endocrinol Metab 2012; 97: 17021711.
5 Langley SC, York DA. Effects of antiglucocorticoid RU 486 on development of
obesity in obese fa/fa Zucker rats. Am J Physiol 1990; 259: R539R544.
6 Livingstone DE, Kenyon CJ, Walker BR. Mechanisms of dysregulation of 11 beta-
hydroxysteroid dehydrogenase type 1 in obese Zucker rats. J Endocrinol 2000;
167: 533539.
7 Chapman AB, Knight DM, Ringold GM. Glucocorticoid regulation of adipocyte
differentiation: hormonal triggering of the developmental program and induction
of a differentiation-dependent gene. J Cell Biol 1985; 101: 12271235.
8 Gathercole LL, Bujalska IJ, Stewart PM, Tomlinson JW. Glucocorticoid modulation
of insulin signaling in human subcutaneous adipose tissue. J Clin Endocrinol
Metab 2007; 92: 43324339.
9 Harrell CS, Gillespie CF, Neigh GN. Energetic stress: The reciprocal relationship between
energy availability and the stress response. Physiol Behav 2015; S0031-9384: 301396.
10 Feraco A, Armani A, Mammi C, Fabbri A, Rosano GM, Caprio M. Role of miner-
alocorticoid receptor and renin-angiotensin-aldosterone system in adipocyte
dysfunction and obesity. J Steroid Biochem Mol Biol 2013; 137:99106.
11 Rondinone CM, Rodbard D, Baker ME. Aldosterone stimulated differentiation of
mouse 3T3-L1 cells into adipocytes. Endocrinology 1993; 132: 24212426.
12 Zennaro MC, Le Menuet D, Viengchareun S, Walker F, Ricquier D, Lombes M.
Hibernoma development in transgenic mice identies brown adipose tissue as a
novel target of aldosterone action. J Clin Invest 1998; 101: 12541260.
13 Caprio M, Feve B, Claes A, Viengchareun S, Lombes M, Zennaro MC. Pivotal role of
the mineralocorticoid receptor in corticosteroid-induced adipogenesis. FASEB J
2007; 21: 21852194.
14 Hoppmann J, Perwitz N, Meier B, Fasshauer M, Hadaschik D, Lehnert H et al. The
balance between gluco- and mineralo-corticoid action critically determines
inammatory adipocyte responses. J Endocrinol 2010; 204: 153164.
15 Funder J, Myles K. Exclusion of corticosterone from epithelial mineralocorticoid
receptors is insufcient for selectivity of aldosterone action: in vivo binding stu-
dies. Endocrinology 1996; 137: 52645268.
16 Funder JW. Mineralocorticoid receptors: distribution and activation. Heart Fail Rev
2005; 10:1522.
17 Armani A, Marzolla V, Rosano G, Caprio M. Mineralocort icoid vs glucocorticoid
receptors: solo players or team mates in the control of adipogenesis? Int J Obes
(Lond) 2014; 38: 15801581.
18 Lee MJ, Fried SK. The glucocorticoid receptor, not the mineralocorticoid receptor,
plays the dominant role in adipogenesis and adipokine production in human
adipocytes. Int J Obes (Lond) 2014; 38:12281233.
19 Guo C, Ricchiuti V, Lian BQ, Yao TM, Coutinho P, Romero JR et al. Miner-
alocorticoid receptor blockade reverses obesity-related changes in expression of
adiponectin, peroxisome proliferator-activated receptor-gamma, and proin-
ammatory adipokines. Circulation 2008; 117: 22532261.
20 Hirata A, Maeda N, Hiuge A, Hibuse T, Fujita K, Okada T et al. Blockade of
mineralocorticoid receptor reverses adipocyte dysfunction and insulin resistance
in obese mice. Cardiovasc Res 2009; 84: 164172.
21 Armani A, Cinti F, Marzolla V, Morgan J, Cranston GA, Antelmi A et al. Miner-
alocorticoid receptor antagonism induces browning of white adipose tissue
through impairment of autophagy and prevents adipocyte dysfunction in high-
fat-diet-fed mice. FASEB J 2014; 28: 37453757.
22HuntHJ,RayNC,HyndG,SuttonJ,SajadM,O'ConnorEet al. Discovery of
a novel non-steroidal GR antagonist with in vivo efcacy in the olanzapine-
induced weight gain model in the rat. Bioorg Med Chem Lett 2012; 22:
73767380.
23 Zorrilla EP, Sanchez-Alavez M, Sugama S, Brennan M, Fernandez R, Bartfai T et al.
Interleukin-18 controls energy homeostasis by suppressing appetite and feed
efciency. Proc Natl Acad Sci USA 2007; 104: 1109711102.
24 Marinozzi F, Marinozzi A, Bini F, Zuppante F, Pecci R, Bedini R. Variability of
morphometric parameters of human trabecular tissue from coxo-arthritis and
osteoporotic samples. Ann Ist Super Sanita 2012; 48:1925.
25 Soukas A, Socci ND, Saatkamp BD, Novelli S, Friedman JM. Distinct transcrip-
tional proles of adipogenesis in vivo and in vitro. JBiolChem2001; 276:
3416734174.
26 Caprio M, Antelmi A, Chetrite G, Muscat A, Mammi C, Marzolla V et al. Anti-
adipogenic effects of the mineralocorticoid receptor antagonist drospirenone:
potential implications for the treatment of metabolic syndrome. Endocr inology
2011; 152: 113125.
27 Collins S, Martin TL, Surwit RS, Robidoux J. Genetic vulnerability to diet-induced
obesity in the C57BL/6 J mouse: physiologic al and molecular characteristics.
Physiol Behav 2004; 81:243248.
28 Hashimoto T, Igarashi J, Hasan AU, Ohmori K, Kohno M, Nagai Y et al. Mifepristone
promotes adiponectin production and improves insulin sensitivity in a mouse
model of diet-induced-obesity. PLoS One 2013; 8: e79724.
29 van den Heuvel JK, Boon MR, Mol IM, van de Velde N, Koorneef LL, Hunt H et al. A
novel mixed glucocorticoid/mineralocorticoid receptorselective modulator redu-
cesobesityand adipose tissue and liver inammation. Oral presentation COST-
ADMIRE Annual Meeting, Padua, Italy, October 16th-17th, 2014.
30 Seale P, Conroe HM, Estall J, Kajimura S, Frontini A, Ishibashi J et al. Prdm16
determines the thermogenic program of subcutaneous white adipose tissue
in mice. J Clin Invest 2011; 121:96105.
31 Wada T, Kenmochi H, Miyashita Y, Sasaki M, Ojima M, Sasahara M et al. Spir-
onolactone improves glucose and lipid metabolism by ameliorating hepatic
steatosis and inammation and suppressing enhanced gluconeogenesis induced
by high-fat and high-fructose diet. Endocrinology 2010; 151: 20402049.
32 Marroqui L, Gonzalez A, Neco P, Caballero-Garrido E, Vieira E, Ripoll C et al. Role of
leptin in the pancreatic beta-cell: effects and signaling pathways. J Mol Endocr inol
2012; 49:R917.
33 Wu Z, Puigserver P, Andersson U, Zhang C, Adelmant G, Mootha V et al.
Mechanisms controlling mitochondrial biogenesis and respiration through the
thermogenic coactivator PGC-1. Cell 1999; 98:115124.
34 Kleiner S, Mepani RJ, Laznik D, Ye L, Jurczak MJ, Jornayvaz FR et al. Development
of insulin resistance in mice lacking PGC-1alpha in adipose tissues. Proc Natl Acad
Sci USA 2012; 109: 96359640.
35 Choo HJ, Kim JH, Kwon OB, Lee CS, Mun JY, Han SS et al. Mitochondria are
impaired in the adipocytes of type 2 diabetic mice. Diabetologia 2006; 49:
784791.
Supplementary Information accompanies this paper on International Journal of Obesity website (http://www.nature.com/ijo)
GR/MR antagonism in adipose tissue
C Mammi et al
9
© 2016 Macmillan Publishers Limited International Journal of Obesity (2016), 1 9
... The effect of glucocorticoid treatment contributes to the decrease of neuroinflammation. However, the chronic use of glucocorticoids is associated with weight gain and increased obesity and related disorders [184,185]. ...
Article
Full-text available
Multiple Sclerosis (MS) is an autoimmune disorder of the Central Nervous System that has been associated with several environmental factors, such as diet and obesity. The possible link between MS and obesity has become more interesting in recent years since the discovery of the remarkable properties of adipose tissue. Once MS is initiated, obesity can contribute to increased disease severity by negatively influencing disease progress and treatment response, but, also, obesity in early life is highly relevant as a susceptibility factor and causally related risk for late MS development. The aim of this review was to discuss recent evidence about the link between obesity, as a chronic inflammatory state, and the pathogenesis of MS as a chronic autoimmune and inflammatory disease. First, we describe the main cells involved in MS pathogenesis, both from neural tissue and from the immune system, and including a new participant, the adipocyte, focusing on their roles in MS. Second, we concentrate on the role of several adipokines that are able to participate in the mediation of the immune response in MS and on the possible cross talk between the latter. Finally, we explore recent therapy that involves the transplantation of adipocyte precursor cells for the treatment of MS.
... Moreover, specific nutrients seem to have some influence in the white-brown transdifferentiation phenomenon [68,69]. Further, interesting data have paid attention to the role of mineralocorticoid receptor antagonism, providing a new potential browning inducer [70][71][72][73]. ...
Article
Full-text available
In the last 30 years the adipose cell has been object of several studies, turning its reputation from an inert cell into the main character involved in the pathophysiology of multiple diseases, including the ongoing COVID-19 pandemic, which has changed the clinical scenario of the last two years. Composed by two types of tissue (white and brown), with opposite roles, the adipose organ is now classified as a real endocrine organ whose dysfunction is involved in different diseases, mainly obesity and type 2 diabetes. In this mini-review we aim to retrace the adipose organ history from physiology to physiopathology, to provide therapeutic perspectives for the prevention and treatment of its two main related diseases (obesity and type 2 diabetes) and to summarize the most recent discoveries linking adipose tissue to COVID-19.
... The value of nuclear receptor ligand combination strategies will become clear in years to come and following more research into receptor crosstalk at the endogenous level. Nonsteroidal compounds such as CORT118335, which have combined GR modulator and MRA properties (Nguyen et al., 2017), may be an additional interesting route to further explore in the context of GR-MR heterodimers and have been so far studied in the context of brain (Atucha et al., 2015;Nguyen et al., 2017), obesity (Mammi et al., 2016) or liver disease (Koorneef et al., 2018). Of note, in the case of cortisol as ligand, GR-MR heterodimers will primarily be targeted in nonepithelial cells where the expression of 11βhydroxysteroid dehydrogenase (11β-HSD2), the enzyme that converts cortisol (active) into cortisone (inactive), is mostly low or absent and so the MR is also occupied by cortisol. ...
Article
Full-text available
Mineralocorticoid receptor antagonists (MRAs) are highly effective therapies for cardiovascular and renal disease. However, the widespread clinical use of currently available MRAs in cardiorenal medicine is hampered by an increased risk of hyperkalaemia. The mineralocorticoid receptor (MR) is a nuclear receptor responsible for fluid and electrolyte homeostasis in epithelial tissues, whereas pathophysiological MR activation in nonepithelial tissues leads to undesirable pro‐inflammatory and profibrotic effects. Therefore, new strategies that selectively target the deleterious effects of the MR but spare its physiological function are needed. In this review, we discuss recent pharmacological developments starting from novel non‐steroidal MRAs, such as finerenone or esaxerenone, that are now entering clinical use, to concepts arising from the current knowledge of the MR signalling pathway, aiming at receptor–coregulator interaction, epigenetics or downstream effectors of the MR. LINKED ARTICLES This article is part of a themed issue on Emerging Fields for Therapeutic Targeting of the Aldosterone‐Mineralocorticoid Receptor Signaling Pathway. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.13/issuetoc
... Impairment of glucose tolerance by decreasing glucose uptake and metabolism is induced by excess dexamethasone activating GR, excess corticosterone activating both GR and MR, and overexpression of MR in adipose tissue [4e7]. It has also been shown that combined antagonism of GR and MR improves glucose tolerance [39,40]; the effect of 20b-DHB demonstrated herein is therefore consistent with that of a mixed GR/MR agonist. Interestingly, the effects of dexamethasone on glucose uptake are more marked under basal conditions compared with insulin-stimulated conditions, which may also be the case for 20b-DHB given that overexpression of Cbr1 did not worsen glucose tolerance during highfat feeding [41]. ...
Article
Full-text available
Objective Carbonyl reductase 1 (Cbr1), a recently discovered contributor to tissue glucocorticoid metabolism converting corticosterone to 20β-dihydrocorticosterone (20β-DHB), is upregulated in adipose tissue of obese humans and mice and may contribute to the cardiometabolic complications of obesity. This study tested the hypothesis that Cbr1-mediated glucocorticoid metabolism influences glucocorticoid and mineralocorticoid receptor activation in adipose tissue, and impacts glucose homeostasis in the lean and obese state. Methods The actions of 20β-DHB on corticosteroid receptors in adipose tissue were investigated first using a combination of in silico, in vitro and transcriptomic techniques and then in vivo administration in combination with receptor antagonists. Mice lacking one Cbr1 allele and mice overexpressing Cbr1 in their adipose tissue underwent metabolic phenotyping before and after induction of obesity with high-fat feeding. Results 20β-DHB activated both the glucocorticoid and mineralocorticoid receptor in adipose tissue and systemic administration to wild-type mice induced glucose intolerance, an effect which was ameliorated by both glucocorticoid and mineralocorticoid receptor antagonism. Cbr1 haploinsufficient lean male mice had lower fasting glucose and improved glucose tolerance compared with littermate controls, a difference that was abolished by administration of 20β-DHB and absent in female mice with higher baseline adipose 20β-DHB concentrations than male mice. Conversely, overexpression of Cbr1 in adipose tissue resulted in worsened glucose tolerance and higher fasting glucose in lean male and female mice. However, neither Cbr1 haploinsfficiency nor adipose overexpression affected glucose dyshomeostasis induced by high-fat feeding. Conclusions Carbonyl reductase 1 is a novel regulator of glucocorticoid and mineralocorticoid receptor activation in adipose tissue which influences glucose homeostasis in lean mice.
... MR, but not GR, knockdown inhibits GCinduced adipogenesis and expression of pro-inflammatory adipokines [38,39]. A combined MRA/GRA provides improvements (predominantly in body fat) in diet-induced metabolic dysregulation compared to GRA alone in rodents [46]. In humans, we have found that MRA (spironolactone) exerts opposing effects to those of GC (prednisolone) on adipose tissue metabolism in vivo; prednisolone suppressed the function of BAT and promoted postprandial lipid synthesis [47] while spironolactone enhanced BAT function and suppressed lipid synthesis [36]. ...
Article
Metabolic syndrome is a cluster of conditions that increase the risk of cardiovascular diseases, and comprises obesity, hypertension, impaired glucose metabolism and dyslipidaemia. It is well recognised that the mineralocorticoid receptor (MR) plays an important role in blood pressure regulation via its effect on salt and water retention in renal tubules, with hypertension being a key feature in primary aldosteronism patients with excess adrenal production of aldosterone, the primary ligand for MRs in the epithelial tissues. MRs are also expressed in a number of non-epithelial tissues including adipose tissue; in these tissues, glucocorticoids or cortisol can also activate MRs due to low levels of 11-beta-hydroxysteroid-dehydrogenase type 2 (11-βHSD2), the enzyme which inactivates cortisol. There is increasing evidence suggesting that over-activation of MRs plays a role in the pathophysiology of the other components of metabolic syndrome, promoting adiposity, inflammation and glucose intolerance, and that MR antagonists may confer beneficial effects on energy and substrate homeostasis and cardiometabolic diseases. This review discusses the advances in the literature shedding light on the MR as an emerging player in metabolic syndrome.
... However, the side effects caused by improper use of dexamethasone significantly limit its clinical application [5,6]. Disordered lipid metabolism and oxidative stress injury are side effects of dexamethasone [7,8]. Metabolic disorders can cause changes in fat mass and distribution, non-alcoholic fatty liver disease (NAFLD), and insulin resistance [9]. ...
Article
Full-text available
In order to investigate efficacy of FGF21 combine dexamethasone (Dex) on rheumatoid arthritis (RA) meanwhile reduce side effects of dexamethasone. We used combination therapy (Dex 15 mg/kg + FGF21 0.25 mg/kg, Dex 15 mg/kg + FGF21 0.5 mg/kg or Dex 15 mg/kg + FGF21 1 mg/kg) and monotherapy (Dex 15 mg/kg or FGF21 1 mg/kg) to treat CIA mice induced by chicken type II collagen, respectively. The effects of treatment were determined by arthritis severity score, histological damage, and cytokine production. The levels of oxidative stress parameters, liver functions, and other blood biochemical indexes were detected to determine FGF21鈥檚 efficiency to side effects of dexamethasone. Oil red O was performed to detect the effects of FGF21 and dexamethasone on fat accumulation in HepG2 cells. The mechanism of FGF21 improves the side effects of dexamethasone which was analyzed by Western blotting. This combination proved to be therapeutically more effective than dexamethasone or FGF21 used singly. FGF21 regulates oxidative stress and lipid metabolism by upregulating dexamethasone-inhibited SIRT-1 and then activating downstream Nrf-2/HO-1and PGC-1. FGF21 and dexamethasone are highly effective in the treatment of arthritis; meanwhile, FGF21 may overcome the limited therapeutic response and Cushing鈥檚 syndrome associated with dexamethasone.
Article
A series of ursodeoxycholic acid (UDCA)-cinnamic acid hybrids were designed and synthesized. The anti-inflammatory activity of these derivatives was screened through evaluating their inhibitory effects of LPS-induced nitric oxide production in RAW264.7 macrophages. The preliminary structure-activity relationship was concluded. Among them, 2m showed the best inhibitory activity against NO (IC50 = 7.70 μM) with no significant toxicity. Further study revealed that 2m significantly decreased the levels of TNF-α, IL-1β, IL-6 and PGE2, down-regulated the expression of iNOS and COX-2. Preliminary mechanism study indicated that the anti-inflammatory activity of 2m was related to the inhibition of the Akt/NF-κB and MAPK signaling pathway. Furthermore, 2m reduced inflammation by a mouse model of LPS-induced inflammatory disease in vivo. In brief, our findings indicated that 2m might serve as a new lead compound for further development of anti-inflammatory agents.
Article
Full-text available
Purpose of review: Bone marrow adipose tissue (BMAT) in the skeleton likely plays a variety of physiological and pathophysiological roles that are not yet fully understood. In elucidating the complex relationship between bone and BMAT, glucocorticoids (GCs) are positioned to play a key role, as they have been implicated in the differentiation of bone marrow mesenchymal stem cells (BMSCs) between osteogenic and adipogenic lineages. The purpose of this review is to illuminate aspects of both endogenous and exogenous GC signaling, including the influence of GC receptors, in mechanisms of bone aging including relationships to BMAT. Recent findings: Harmful effects of GCs on bone mass involve several cellular pathways and events that can include BMSC differentiation bias toward adipogenesis and the influence of mature BMAT on bone remodeling through crosstalk. Interestingly, BMAT involvement remains poorly explored in GC-induced osteoporosis and warrants further investigation. This review provides an update on the current understanding of the role of glucocorticoids in the biology of osteoblasts and bone marrow adipocytes (BMAds).
Article
Full-text available
Introduction Nonalcoholic fatty liver disease (NAFLD) is the most common liver disorder in Western industrialized countries and may progress to liver injury. Cortisol is thought to play a role in the pathogenesis of NAFLD and cortisol modulation has shown efficacy in preclinical models. However, published reports of the clinical effects of glucocorticoid receptor (GR) antagonism in these patients are limited. Case report Two female patients (66 and 60 years old) with endogenous hypercortisolism presented with hepatic steatosis, hypertension, type 2 diabetes mellitus, and dyslipidemia. Both patients declined adrenalectomy or pituitary tumor surgery and mifepristone 300 mg daily was initiated. During mifepristone treatment (durations ranging from 10 months to 5 years), improvements in hypercortisolism-related cardiometabolic abnormalities were observed, including normalization of lipid levels and improvement of hyperglycemia. In both cases, follow-up imaging revealed resolution of fatty liver, which was supported by a decrease in liver enzymes on liver function tests. No adverse events were reported. Discussion NAFLD is frequently observed in patients with endogenous hypercortisolism. Improvement in liver function tests has previously been demonstrated in patients with hypercortisolism treated with mifepristone. These cases showed, for the first time, radiological improvement of liver steatosis following mifepristone use in hypercortisolemic patients with NAFLD. Conclusion This case series demonstrated improvements in biochemical and imaging parameters of NAFLD in patients with hypercortisolism treated with mifepristone. Further research is needed to investigate the effects of GR modulation in fatty liver disease.
Article
Full-text available
Drug-induced lipid accumulation in the liver may induce two clinically relevant conditions, drug-induced steatosis (DIS) and drug-induced steatohepatitis (DISH). The list of drugs that may cause DIS or DISH is long and heterogeneous and includes therapeutically relevant molecules that cannot be easily replaced by less hepatotoxic medicines, therefore making specific strategies necessary for DIS/DISH prevention or treatment. For years, the only available tools to achieve these goals have been antioxidant drugs and free radical scavengers, which counteract drug-induced mitochondrial dysfunction but, unfortunately, have only limited efficacy. In the present review we illustrate how in vitro preclinical research unraveled new key players in the pathogenesis of specific forms of DISH, and how, in a few cases, proof of concept of the beneficial effects of their pharmacological modulation has been obtained in vivo in animal models of this condition. The key issue emerging from these studies is that, in selected cases, liver toxicity depends on mechanisms unrelated to those responsible for the desired, primary pharmacological effects of the toxic drug and, therefore, specific strategies can be designed to overcome steatogenicity without making the drug ineffective. In particular, the hepatotoxic drug could be given in combination with a second molecule intended to selectively antagonize its liver toxicity whilst, ideally, potentiating its desired pharmacological activity. Although most of the evidence that we discuss is from in vitro or animal models and will need to be further explored and validated in humans, it highlights new avenues to be pursued in order to improve the safety of steatogenic drugs.
Article
Full-text available
The mineralocorticoid receptor (MR) controls adipocyte function, but its role in the conversion of white adipose tissue (WAT) into thermogenic fat has not been elucidated. We investigated responses to the MR antagonists spironolactone (spiro; 20 mg/kg/d) and drospirenone (DRSP; 6 mg/kg/d) in C57BL/6 mice fed a high-fat (HF) diet for 90 d. DRSP and spiro curbed HF diet-induced impairment in glucose tolerance, and prevented body weight gain and white fat expansion. Notably, either MR antagonist induced up-regulation of brown adipocyte-specific transcripts and markedly increased protein levels of uncoupling protein 1 (UCP1) in visceral and inguinal fat depots when compared with the HF diet group. Positron emission tomography and magnetic resonance spectroscopy confirmed acquisition of brown fat features in WAT. Interestingly, MR antagonists markedly reduced the autophagic rate both in murine preadipocytes in vitro (10(-5) M) and in WAT depots in vivo, with a concomitant increase in UCP1 protein expression. Moreover, the autophagy repressor bafilomycin A1 (10(-8) M) mimicked the effect of MR antagonists, increasing UCP1 protein expression in primary preadipocytes. Hence, we showed that adipocyte MR regulates brown remodeling of WAT through a modulation of autophagy. These results provide a rationale for the use of MR antagonists to prevent the adverse metabolic consequences of adipocyte dysfunction.-Armani, A., Cinti, F., Marzolla, V., Morgan, J., Cranston, G. A., Antelmi, A., Carpinelli, G., Canese, R., Pagotto, U., Quarta, C., Malorni, W., Matarrese, P., Marconi, M., Fabbri, A., Rosano, G., Cinti, S., Young, M. J., Caprio, M. Mineralocorticoid receptor antagonism induces browning of white adipose tissue through impairment of autophagy and prevents adipocyte dysfunction in high-fat-diet-fed mice.
Article
Full-text available
International Journal of Obesity is a monthly, multi-disciplinary forum for papers describing basic, clinical and applied studies in biochemistry, genetics and nutrition, together with molecular, metabolic, psychological and epidemiological aspects of obesity and related disorders
Article
Full-text available
Background Both the glucocorticoid receptor (GR) and the mineralocorticoid receptor (MR) are expressed in adipose tissue and assumed to mediate cortisol actions on adipose tissue. The relative significance of the two receptors in mediating glucocorticoid regulation of adipogenesis and adipokine expression in human adipocytes has not been addressed. Methods We investigated the differential roles of the GR and MR in mediating glucocorticoid actions on adipogenesis and adipokine production using RNA interference in primary cultures of human preadipocytes and adipocytes. RESULTS Both types of receptors are expressed, but levels of GR were several hundred fold higher than MR in both human preadipocytes and adipocytes. As expected, cortisol added during adipogenesis increased the differentiation of human preadipocytes. Silencing of GR, but not MR, blocked these proadipogenic actions of cortisol. In differentiated human adipocytes, addition of cortisol increased leptin and adiponectin, while suppressing IL-6, mRNA levels and protein secretion. Knockdown of GR by 65% decreased leptin and adiponectin while increasing IL-6 production. In addition, GR silencing blocked the effects of cortisol on adipokine expression. In contrast, although MR knockdown increased leptin, it did not affect adiponectin and IL-6 expression. Conclusion Our data demonstrate that although both GR and MR have roles in regulating leptin expression, GR plays more important roles in mediating the actions of cortisol to regulate adipogenesis and adipokine production in human adipocytes.
Article
Full-text available
The steroid receptor antagonist mifepristone is used as an anti-cancer agent, eliciting both cytostatic and cytotoxic effects on malignant cells. However, the metabolic effects of long-term treatment with mifepristone have remained unclear. The effects of mifepristone on insulin sensitivity and adiponectin secretion were evaluated both in in vivo and in vitro. First, we explored the effects of mifepristone, on metabolic functions in obese mice receiving a high-fat diet. When these mice were fed mifepristone, they exhibited a marked improvement in insulin sensitivity, attenuated hepatic injury, and decreased adipocyte size, compared with mice that received only the high-fat diet. Intriguingly, mifepristone-treated mice showed significantly elevated plasma adiponectin levels. Second, we tested the effects of mifepristone on differentiated 3T3-L1 adipocytes in vitro. When differentiated adipocytes were treated with mifepristone for 48 h, adiponectin was upregulated at both mRNA and protein levels. Collectively, these results reveal novel actions of mifepristone on metabolic functions, in vivo and in vitro, in which the drug exerts antidiabetic effects associated with an upregulation in adiponectin-secretion.
Article
The worldwide epidemic of metabolic syndromes and the recognized burden of mental health disorders have driven increased research into the relationship between the two. A maladaptive stress response is implicated in both mental health disorders and metabolic disorders, implicating the hypothalamic-pituitary-adrenal (HPA) axis as a key mediator of this relationship. This review explores how an altered energetic state, such as hyper- or hypoglycemia, as may be manifested in obesity or diabetes, affects the stress response and the HPA axis in particular. We propose that changes in energetic state or energetic demands can result in "energetic stress" that can, if prolonged, lead to a dysfunctional stress response. In this review, we summarize the role of the hypothalamus in modulating energy homeostasis and then briefly discuss the relationship between metabolism and stress-induced activation of the HPA axis. Next, we examine seven mechanisms whereby energetic stress interacts with neuroendocrine stress response systems, including by glucocorticoid signaling both within and beyond the HPA axis; by nutrient-induced changes in glucocorticoid signaling; by impacting the sympathetic nervous system; through changes in other neuroendocrine factors; by inducing inflammatory changes; and by altering the gut-brain axis. Recognizing these effects of energetic stress can drive novel therapies and prevention strategies for mental health disorders, including dietary intervention, probiotics, and even fecal transplant.
Article
We have analyzed the hormonal basis for the acceleration of differentiation by dexamethasone and insulin in the stable adipogenic cell line TA1. These cells, which were derived from 5-azacytidine-treated 10T1/2 mouse embryo fibroblasts, undergo differentiation in culture after reaching confluence. The ensuing morphological changes are accompanied by widespread alterations in the pattern of protein synthesis and the increased accumulation of specific mRNAs. Using cDNA clones corresponding to mRNAs that are induced during adipogenesis, we find that dexamethasone elicits the precocious accumulation of differentiation-specific gene products. This effect appears to be mediated by the glucocorticoid receptor, yet unlike standard steroid inductions, most of the RNAs reach the same maximal levels in the absence of dexamethasone. Glucocorticoids thus may increase the expression of a regulatory factor required for activating the entire set of differentiation-dependent genes. We also describe a gene whose transcription is not only activated during adipogenesis but is also specifically inducible by dexamethasone in the mature adipocyte. Moreover, the glucocorticoid responsiveness of this gene in differentiated cells appears to be dependent on its prior developmental activation.