ArticlePDF Available

Esculetin ameliorates hepatic fibrosis in high fat diet induced non-alcoholic fatty liver disease by regulation of FoxO1 mediated pathway

Authors:
  • St. Michael's Hospital (University of Toronto)

Abstract and Figures

Background: Non-alcoholic fatty liver disease (NAFLD), a chronic metabolic disorder is associated with oxidative stress, inflammation and fibrotic cascades. In this study, we aimed to examine the effects of Esculetin, a well-known anti-oxidant on TGF-β1 mediated liver fibrosis and FoxO1 activity. Methods: A non-genetic murine model for NAFLD was developed by chronic high fat diet (HFD) (58% calories from fats) feeding in Wistar rats. The plasma biochemical parameters, liver function tests, oxidative stress, and histopathological alterations were assessed. The alterations in extracellular matrix (ECM) deposition and FoxO1 activity were assessed by immunohistochemistry. Results: The aberrations in plasma parameters, liver functioning, morphometric and microscopic changes in liver structure of HFD fed rats were significantly improved by treatment with Esculetin. Liver fibrosis, identified in the form of collagen deposition and expression of fibrotic proteins like TGF-β1 and fibronectin was also markedly controlled by Esculetin. The expression of phospho-FoxO1 was found to be reduced in HFD fed rats' liver, showing an increase in activation of FoxO1 under insulin resistant and hyperglycemic states. Esculetin treatment could improve phospho-FoxO1 expression, thus showing its ability to act on Akt/PI3K/FoxO1 pathway. Conclusions: As per the previous studies, a potential therapy for NAFLD may be the one with multi-faceted actions on insulin resistance, oxidative stress, inflammation and fibrosis. This study demonstrates the efficiency of Esculetin in improving liver fibrosis in HFD induced NAFLD.
Content may be subject to copyright.
Original
article
Esculetin
ameliorates
hepatic
brosis
in
high
fat
diet
induced
non-
alcoholic
fatty
liver
disease
by
regulation
of
FoxO1
mediated
pathway
Anuradha
Pandey,
Priyank
Raj,
Santosh
Kumar
Goru,
Almesh
Kadakol,
Vajir
Malek,
Nisha
Sharma,
Anil
Bhanudas
Gaikwad*
Laboratory
of
Molecular
Pharmacology,
Department
of
Pharmacy,
Birla
Institute
of
Technology
and
Science
Pilani,
Pilani
Campus,
Rajasthan,
India
A
R
T
I
C
L
E
I
N
F
O
Article
history:
Received
22
September
2016
Received
in
revised
form
20
January
2017
Accepted
3
February
2017
Available
online
4
February
2017
Keywords:
Non-alcoholic
fatty
liver
disease
Insulin
resistance
Fibrosis
FoxO1
Esculetin
A
B
S
T
R
A
C
T
Background:
Non-alcoholic
fatty
liver
disease
(NAFLD),
a
chronic
metabolic
disorder
is
associated
with
oxidative
stress,
inammation
and
brotic
cascades.
In
this
study,
we
aimed
to
examine
the
effects
of
Esculetin,
a
well-known
anti-oxidant
on
TGF-
b
1
mediated
liver
brosis
and
FoxO1
activity.
Methods:
A
non-genetic
murine
model
for
NAFLD
was
developed
by
chronic
high
fat
diet
(HFD)
(58%
calories
from
fats)
feeding
in
Wistar
rats.
The
plasma
biochemical
parameters,
liver
function
tests,
oxidative
stress,
and
histopathological
alterations
were
assessed.
The
alterations
in
extracellular
matrix
(ECM)
deposition
and
FoxO1
activity
were
assessed
by
immunohistochemistry.
Results:
The
aberrations
in
plasma
parameters,
liver
functioning,
morphometric
and
microscopic
changes
in
liver
structure
of
HFD
fed
rats
were
signicantly
improved
by
treatment
with
Esculetin.
Liver
brosis,
identied
in
the
form
of
collagen
deposition
and
expression
of
brotic
proteins
like
TGF-
b
1
and
bronectin
was
also
markedly
controlled
by
Esculetin.
The
expression
of
phospho-FoxO1
was
found
to
be
reduced
in
HFD
fed
rats
liver,
showing
an
increase
in
activation
of
FoxO1
under
insulin
resistant
and
hyperglycemic
states.
Esculetin
treatment
could
improve
phospho-FoxO1
expression,
thus
showing
its
ability
to
act
on
Akt/PI3
K/FoxO1
pathway.
Conclusions:
As
per
the
previous
studies,
a
potential
therapy
for
NAFLD
may
be
the
one
with
multi-faceted
actions
on
insulin
resistance,
oxidative
stress,
inammation
and
brosis.
This
study
demonstrates
the
efciency
of
Esculetin
in
improving
liver
brosis
in
HFD
induced
NAFLD.
©
2017
Institute
of
Pharmacology,
Polish
Academy
of
Sciences.
Published
by
Elsevier
Sp.
z
o.o.
All
rights
reserved.
Introduction
Non-alcoholic
fatty
liver
disease
(NAFLD),
characterized
by
hepatic
steatosis
without
any
other
causes
of
hepatic
fat
accumulation
such
as
signicant
alcohol
consumption,
steatogenic
medication
or
hereditary
disorders
is
one
of
the
most
common
liver
diseases
globally
[1].
In
about
15%
of
all
NAFLD
cases,
steatosis
may
evolve
into
non-alcoholic
steatohepatitis
(NASH),
a
juncture
of
inammation,
insulin
resistance
(IR),
hepatocellular
injury,
and
brosis,
often
resulting
in
cirrhosis
and
even
cancer
[2].
The
most
important
risk
factors
for
NAFLD
are
male
gender,
age,
obesity,
IR
and
metabolic
syndrome
[1].
Its
pathogenesis
involves
multiple
hits-
rst
hit
includes
lipid
accumulation
in
the
liver,
followed
by
subsequent
hits
in
which
pro
inammatory
mediators
induce
inammation,
oxidative
stress,
hepatocellular
injury,
and
brosis.
Fibrosis,
excessive
accumula-
tion
of
extra
cellular
matrix
(ECM)
proteins
including
collagen
plays
a
signicant
role
in
pathogenesis
of
liver
diseases
[3].
Advanced
liver
brosis
culminates
into
cirrhosis,
liver
failure,
portal
hypertension
and
often
requires
liver
transplantation.
Activated
hepatic
stellate
cells
(HSC),
portal
broblasts,
and
myobroblasts
of
bone
marrow
origin
have
been
identied
as
major
collagen-producing
cells
in
the
injured
liver
[3].
Forkhead
boxcontaining
protein
O
subfamily-1
(FoxO1),
a
transcription
factor
controlling
the
expression
of
glucose-6-phosphatase,
catalytic
subunit
and
phosphoenolpyruvate
carboxykinase
also
plays
a
crucial
role
in
the
trans-differentiation
and
proliferation
of
HSCs
in
liver
brosis.
Hyperinsulinemia
has
been
known
to
inactivate
FoxO1
in
HSCs,
resulting
in
HSC
activation
and
may
result
in
the
brosis
in
NAFLD
[4].
Despite
vast
ongoing
research,
dietary
intervention
and
exercise
remain
the
rst-line
therapy
and
there
are
no
surgical
or
pharmacological
interventions
approved
for
its
treatment.
*
Corresponding
author.
E-mail
address:
anil.gaikwad@pilani.bits-pilani.ac.in
(A.B.
Gaikwad).
http://dx.doi.org/10.1016/j.pharep.2017.02.005
1734-1140/©
2017
Institute
of
Pharmacology,
Polish
Academy
of
Sciences.
Published
by
Elsevier
Sp.
z
o.o.
All
rights
reserved.
Pharmacological
Reports
69
(2017)
666672
Contents
lists
available
at
ScienceDirect
Pharmacological
Reports
journal
home
page
:
www.elsevier.com/locat
e/pharep
Recent
studies
show
that
insulin
sensitizers,
anti-oxidants,
and
statins
could
potentially
be
used
as
adjunctive
therapy
of
NAFLD
but
side
effects
and
skewed
risk
benet
ratio
linked
with
synthetic
molecules
make
the
natural
products
or
their
derivatives
an
important
alternative
source
of
high
therapeutic
value
[5].
Ginger,
coffee,
moringa,
eucalyptus
leaf
extract,
liquorice
roots
and
sylibium
have
been
known
to
prevent
NAFLD
or
blunt
its
progression
[5].
Silymarin,
a
lipophilic
extract
derived
from
Silybum
marianum
has
anti-brotic,
insulin
sensitizing,
antioxi-
dant,
antiviral
and
anti-inammatory
activities
which
in
turn
help
in
reduction
steatosis
severity,
liver
ballooning
and
brosis,
followed
by
lowered
aminotransferase
levels
in
both
short
and
long
lasting
therapies
[6].
Another
compound
from
natures
lap
with
profound
anti-inammatory
and
insulin
sensitizing
proper-
ties
is
Esculetin,
a
dihydroxy
coumarin
derivative.
Esculetin
has
displayed
a
signicant
hepatoprotective
effect
in
mice
fed
a
HFD
by
increasing
plasma
adiponectin
level
and
the
protein
and
mRNA
expression
of
hepatic
AdipoR2,
which
led
to
the
activation
of
5
0
AMP-activated
protein
kinase.
The
protein
and
mRNA
expression
of
hepatic
peroxisome
proliferator-activated
receptor
alpha
(PPARa)
were
up-regulated
by
Esculetin
and
those
of
Sterol
regulatory
element-binding
transcription
factor-
1c
were
down-regulated
[7].
Recently,
Choi
et
al.
that
Esculetin
supplementation
could
protect
against
development
of
NAFLD
in
diabetes
through
the
regulation
of
metabolism
of
glucose
and
lipids
and
also
through
reduction
of
inammatory
makers
[8].
Recent
reports
suggest
that
chronic
HFD
feeding
induces
the
pathological
changes
including
visceral
obesity,
inammation,
hyperlipidemia
and
increased
free
fatty
acids
which
are
quite
similar
to
that
of
the
human
NAFLD
[9,10].
A
study
reported
that
HFD
feeding
for
24
weeks
led
to
peri-sinusodial
brosis
which
progressed
from
36
to
48
weeks
with
insulin
resistance
[9].
These
reports
suggested
that
chronic
HFD
feeding
could
be
used
as
an
efcient
tool
to
simulate
clinical
NAFLD
and
associated
hepatic
brosis.
Based
on
the
above
literature
survey,
we
hypothesized
that
Esculetin
might
ameliorate
liver
brosis
associated
with
chronic
high
fat
diet
induced
NAFLD
in
Wistar
rats.
In
the
current
study,
we
aimed
to
study
the
possible
underlying
mechanism
through
which
Esculetin
ameliorates
NAFLD
and
the
associated
hepatic
brosis
in
a
chronic
HFD
fed
murine
model
for
NAFLD.
Materials
and
methods
Chemicals
Antibodies
against
phospho-FoxO1,
bronectin
and
TGF-b1
were
obtained
from
Cell
Signalling
Technology
(Danvers,
MA,
USA).
For
biochemical
estimation,
spectrophotometric
kits
(Accurex
Biomedical
Pvt.
Ltd.,
Mumbai,
India)
and
ultra-sensitive
rat
insulin
kit
obtained
from
Crystal
Chem
(Downers
Grove,
IL,
USA)
were
used.
All
the
other
chemicals
were
purchased
from
Sigma
Aldrich
(St.
Louis,
MO,
USA),
unless
otherwise
mentioned.
Animal
studies
Adult
male
Wistar
rats
(160180
g),
procured
from
the
Central
Animal
Facility
of
the
institute,
Birla
Institute
of
Technology
and
Science
Pilani
were
maintained
under
standard
environmental
conditions
and
were
provided
with
feed
and
water
ad
libitum.
All
the
animals
were
fed
on
normal
pellet
diet
(NPD)
(12%
calories
as
fat)
one
week
prior
to
the
start
of
experimental
protocol
which
is
in
accordance
with
the
guidelines
of
the
Committee
for
the
Purpose
of
Control
and
Supervision
of
Experiments
on
Animals,
Ministry
of
Social
Justice
and
Environment,
Government
of
India
based
on
the
guidelines
of
Institute
of
Laboratory
Animal
Resources,
(Wash-
ington,
DC,
USA).
A
prior
permission
was
sought
from
the
institutional
animal
ethics
committee,
BITS
Pilani,
for
conducting
the
study.
The
animal
model
was
developed
using
chronic
high
fat
diet
(HFD)
(58%
calories
as
fat)
and
the
normal
control
was
treated
with
vehicle
(0.5%
sodium
carboxy
methyl
cellulose)
as
described
previously
[1113].
Based
on
the
previous
reports,
Esculetin
(50
and
100
mg/kg/day,
po)
was
administered
to
the
rats
fed
with
HFD
after
22
weeks
of
initiation
of
the
study.
The
number
of
animals
maintained
in
each
experimental
group
was
6
[11,14].
Assessment
of
plasma
biochemical
parameters
The
blood
samples
were
collected
and
plasma
was
analyzed
for
glucose
(PGL),
triglycerides
(TG)
and
total
cholesterol
(PTC).
Insulin
determination
was
made
by
ELISA
kit
using
rat
insulin
as
standard
(Rat
ELISA
kit,
Crystal
Chem,
USA)
[11,12].
Assessment
of
liver
function
The
plasma
parameters
which
are
potential
indicators
of
liver
function
include
alkaline
phosphatase
(ALP),
lactate
dehydroge-
nase
(LDH),
aspartate
aminotransferase
(AST),
and
alanine
aminotransferase
(ALT)
[15,16].
These
parameters
were
assessed
using
commercially
available
kits
(Accurex,
Mumbai,
India)
[12].
AST/ALT
ratio,
also
known
as
De
Ritis
ratio
was
calculated
to
determine
development
of
IR
[17].
Homeostasis
model
assessment
(HOMA)
HOMA
is
a
mathematical
model
to
estimate
an
individuals
index
of
IR,
insulin
sensitivity
(%S)
and
steady
state
beta
cell
function
(%B)
based
on
simultaneous
measurements
of
fasting
plasma
glucose
and
insulin
or
C-peptide
concentrations
[18].
HOMA-IR,
%S
and%B
scores
were
calculated
at
the
end
of
the
experimental
period
[19].
Estimation
of
lipid
peroxide
and
reduced
glutathione
(GSH)
levels
in
liver
The
estimation
of
oxidative
stress
was
carried
out
by
assessing
the
lipid
peroxide
and
GSH
levels
in
the
liver.
Briey,
after
sacricing
the
animals,
the
liver
was
excised,
rinsed
with
normal
saline
and
weighed.
The
lipid
peroxide
level
in
animal
tissues
was
measured
according
to
method
described
by
Ohkawa
et
al..
After
weighing,
liver
tissue
was
minced
properly
and
the
homogenate
was
prepared
in
1
mL
of
cold
phosphate
buffered
saline
(pH
7.4)
and
centrifuged
at
700
g.
Supernatant
was
collected
and
used
for
estimation
of
thiobarbituric
acid
reacting
substances
(TBARS)
by
using
spectrophotometric
method
at
532
nm
[20].
The
GSH
content
was
estimated
according
to
Ellmans
method.
For
GSH,
liver
tissues
were
homogenized
in
10
mL
ice-cold
homogenizing
buffer
combined
with
sulphosalicylic
acid
and
the
homogenate
was
centrifuged.
Ellmans
reagent
was
added
to
the
supernatant
to
produce
yellow
colour
of
5-thio-2-nitrobenzoate-SH.
The
absor-
bance
was
taken
at
412
nm
[21].
Histopathological
evaluation
Histopathology
was
performed
as
per
the
protocol
described
previously
[11].
Briey,
the
liver
tissue
was
xed
in
10%
(v/v)
formalin
in
phosphate
buffered
saline
and
embedded
in
parafn.
5
mm
sections
were
stained
with
hematoxylin
and
eosin
(H&E)
to
study
glomerular
damage.
At
least
25
sections
from
each
group
(4-
5
sections
from
each
animal)
were
observed
for
any
histological
changes.
Histopathological
images
were
captured
by
using
Olympus
microscope
(Model
no.
BX41,
NY,
USA)
[22].
For
the
study
of
deposition
of
ECM
(collagen),
the
sections
were
stained
A.
Pandey
et
al.
/
Pharmacological
Reports
69
(2017)
666672
667
with
Picro
Sirius
Red.
The
percentage
brotic
area
was
analyzed
using
ImageJ
software
and
the
data
analysis
was
carried
out
by
using
GraphPad
5
Prism
software
[23].
Immunohistochemistry
Immunohistochemistry
for
checking
the
expression
of
phos-
pho-FoxO1,
bronectin
and
TGF-b1
was
performed
as
per
the
protocol
described
previously
[11,14].
Briey,
liver
sections
(5
mm)
were
taken
from
parafn
blocks
and
deparafnized
with
xylene,
followed
by
antigen
retrieval
by
heating
in
citrate
buffer
(10
mmol/
L).
The
following
primary
antibodies
were
used:
anti-phospho
FoxO1,
bronectin
and
TGF-b1
(rabbit,
1:200
dilution;
Cell
Signaling
Technology)
and
anti-rabbit
Horse
Radish
Peroxidase
conjugated
secondary
antibody
was
used,
followed
by
detection
with
diaminobenzidine
(DAB)
as
a
chromogen.
The
slides
were
counterstained
with
hematoxylin,
dehydrated
with
alcohol
and
xylene,
and
mounted
in
DPX
(Sigma-
Aldrich).
All
the
images
were
analyzed
using
ImageJ
software
for
calculating
DAB
positive
area.
Statistical
analysis
Experimental
values
were
expressed
as
means
SEM.
Statisti-
cal
comparison
between
different
groups
was
performed
using
one
way
analysis
of
variance
to
detect
the
difference
in
observations
between
all
groups.
If
F
value
was
signicant
then
multiple
comparisons
were
done
by
Tukey
test
using
Prism
software
(version
5.0;
GraphPad,
San
Diego,
CA,
USA)
for
Windows.
Data
was
considered
to
be
statistically
signicant
if
p
<
0.05.
Results
Esculetin
improves
the
liver
function
tests
in
HFD
rats
The
plasma
parameters
are
the
hallmarks
of
liver
dysfunction.
The
HFD
fed
rats
were
found
to
show
a
marked
elevation
in
plasma
triglycerides,
cholesterol
and
insulin
but
the
glucose
levels
were
not
altered
signicantly
as
compared
to
the
normal
control
(NC).
Esculetin
treatment
could
signicantly
ameliorate
these
aberrant
alterations
at
both
the
doses.
HOMA-IR
is
an
independent
predictor
of
cardiovascular
disorders
in
type
2
diabetes.
Low
HOMA-IR
values
indicate
high
insulin
sensitivity,
whereas
high
HOMA-IR
values
indicate
low
insulin
sensitivity
(IR)
[24].
The
HOMA
model
showed
that
Esculetin
induced
a
reduction
in
IR
and
an
improvement
in
insulin
sensitivity
in
HFD
fed
rats
at
both
the
doses
(Table
1).
The
normal
functioning
of
liver
is
indicated
by
various
liver
specic
enzymes
which
act
as
hallmarks
of
liver
injury
or
dysfunction.
The
liver
function
parameters,
including
ALP,
LDH,
AST
and
ALT
also
found
to
be
elevated
by
chronic
HFD
feeding.
The
elevation
in
ALT
was
more
prominent
than
AST
in
the
HFD
fed
rats.
Esculetin
treatment
could
control
this
elevation
by
a
signicant
margin,
thus
indicating
a
hepato-protective
aspect
of
Esculetins
therapeutic
effect.
De
Ritis
described
the
AST/ALT
ratio
as
being
a
useful
indicator
of
the
aetiology
of
hepatitis.
Both
serum
AST
and
ALT
increase
with
body
weight
but
this
is
more
prominent
for
ALT
rather
than
AST.
In
alcoholic
steatohepatitis
the
De
Ritis
ratio
is
always
over
1.0,
in
patients
with
NASH
the
AST/ALT
ratio
is
more
than
1.0
particularly
in
morbidly
obese
patients.
De
Ritis
value
of
less
than
1.0
indicates
progression
of
disease
towards
brosis
and
cirrhosis
[17].
In
this
study,
we
found
that
AST
and
ALT
levels
were
elevated
in
HFD
fed
rats,
but
AST/ALT
ratio
was
found
to
be
reduced
signicantly
as
compared
with
the
normal
control
and
the
treatment
with
Esculetin
improved
the
De
Ritis
ratio.
This
indicates
the
ability
of
Esculetin
to
ameliorate
hepatic
brosis
in
chronic
high
fat
diet
rats
(Table
1).
Esculetin
reverses
microscopic
and
morphometric
alterations
in
HFD
fed
rat
liver
HFD
is
widely
used
to
induce
hepatic
steatosis
and
NASH
in
experimental
animals.
Chronic
HFD
feeding
for
24
weeks
led
to
a
signicant
rise
in
the
body
weight
as
well
as
the
liver
weight.
Also,
there
was
a
notable
change
in
the
appearance
of
liver.
These
alterations
could
be
reversed
markedly
by
the
treatment
with
Esculetin
(both
at
50
and
100
mg/kg
doses).
The
relative
liver
weight
was
elevated
by
chronic
HFD
feeding
and
Esculetin
could
limit
this
aberration,
showing
its
ability
to
reduce
the
accumula-
tion
of
fats
in
the
liver
(Table
1).
The
H&E
staining
showed
that
liver
of
the
rats
fed
with
HFD
has
prominent
hypertrophy,
as
was
evidenced
by
the
reduction
in
Table
1
Morphometric
and
plasma
biochemical
parameters.
Normal
Control
(NC)
High
fat
diet
control
(HFD)
HFD+
Esculetin
(HFD
+
E1)
(50
mg/kg/
day,
po)
HFD+
Esculetin
(HFD
+
E2)
(100
mg/kg/day,
po)
Morphometric
parameters
Liver
weight
(gm)
6.675
0.301
9.533
0.393
*
8.400
0.204
7.133
0.145
**
Body
weight
(gm)
241
4.409
318
14.79
*
266
3.59
**
249
2.459
**
Liver
weight/body
weight
ratio
(%)
2.769
0.068
2.997
0.026
*
3.15
0.056
2.864
0.060
**
Plasma
biochemical
parameters
Plasma
glucose
(mmol/L)
6.157
0.2271
7.190
0.4818
*
6.820
0.4787
6.885
0.5629
Plasma
triglyceride
(mg/dL)
53.60
9.541
245.4
19.20
*
177. 8
12.08
**
122.6
6.427
**
Plasma
total
cholesterol
(mmol/L)
1.134
0.209
6.392
0.367
*
3.326
0.277
**
2.777
0.275
**
Plasma
insulin
(pmol/L)
125.3
5.494
267.2
17.90
*
178.7
11.45
**
155.4
8.599
**
HOMA-IR
2.42
0.538
5.1
1.10
*
3.48
0.769
**
3.02
1.30
**
HOMA-
b
113.9
8.072
159
10.68
*
120.8
5.923
**
97.8
9.01
**
HOMA-%
S
41.4
6.56
19.6
4.09
*
28.7
4.29
**
33.1
5.34
**
Alkaline
phosphatase
(IU/L)
120.3
13.57
583.6
49.62
*
375.6
28.18
**
243.1
14.88
**
Lactate
dehydrogenase
(IU/L)
301.0
33.33
644.3
46.79
*
506
23.44
396
25.1
**
Aspartate
aminotransferase-
AST
(IU/L)
24.13
5.875
70.06
5.934
*
49.23
6.856
**
49.19
2.926
**
Alanine
aminotransferase-
ALT
(IU/
L)
25.61
7.741
87.81
10.61
*
55.74
4.765
44.53
7.291
**
De
Ritis
ratio
(AST/ALT
ratio)
0.981
0.125
0.797
0.223
*
0.883
0.241
1.08
0.145
**
Note:
Data
represented
in
mean
SEM
(n
=
6).
*
p
<
0.001
vs.
NC,
**
p
<
0.001
vs.
HFD
668
A.
Pandey
et
al.
/
Pharmacological
Reports
69
(2017)
666672
number
of
nuclei
per
high
power
eld
(hpf),
which
indicates
an
increase
in
individual
cell
size.
The
number
of
nuclei
per
hpf
was
found
to
be
improved
signicantly
by
the
treatment
with
Esculetin.
There
was
no
signicant
difference
observed
between
the
two
doses
of
Esculetin
(Fig.
1A).
Esculetin
ameliorates
oxidative
stress
in
liver
of
HFD
fed
rats
Oxidative
stress
is
one
of
the
major
inammation
and
brosis
triggering
factors
for
development
of
NAFLD.
The
oxidative
stress
was
found
to
be
elevated
signicantly
in
HFD
fed
rats,
owing
to
a
an
increase
in
lipid
peroxidation
elevated
thiobarbituric
acid
reactive
substrate
(TBARS)
and
reduced
anti
oxidants
defence
mechanisms
(GSH
levels).
The
treatment
with
Esculetin
was
found
to
reduce
lipid
peroxidation
and
elevate
GSH
levels
in
the
liver
tissues
of
HFD
fed
rats.
This
result
is
in
line
with
the
previous
reports
which
had
demonstrated
anti-oxidant
potential
of
the
molecule
(Fig.
1B
and
C).
Esculetin
elevates
the
phosphorylation
of
FoxO1
in
liver
of
HFD
fed
rats
FoxO1
is
constitutively
located
in
the
nucleus
to
regulate
the
transcription
of
gluconeogenic
genes.
However,
under
fasting
state
its
phosphorylation
increases
leading
to
the
nucleo-cytoplasmic
transportation
which
results
in
activation
of
the
transcription
factor
[4].
The
phosphorylated
form
of
FoxO1
was
found
to
be
reduced
signicantly
in
rat
liver
upon
chronic
HFD
feeding,
thus
showing
an
increase
in
the
activation
of
FoxO1.
The
treatment
with
Esculetin
could
revert
this
alteration
thus
indicating
its
ability
to
regulate
Akt/phosphatidylinositol
3-kinase
(PI3
K)/FoxO1
pathway
(Fig.
2).
Esculetin
reduces
liver
brosis
in
HFD
fed
rats
As
per
the
previous
reports,
chronic
HFD
feeding
leads
to
hepatic
peri-sinosodial
brosis
from
week
36
to
48
with
insulin
resistance
[9].
In
this
study,
we
found
that
accumulation
of
ECM
proteins
in
liver
was
found
to
be
signicantly
elevated
by
long
term
HFD
feeding.
The
treatment
with
Esculetin
(both
50
and
100
mg/
kg)
could
signicantly
reduce
the
elevated
deposition
of
ECM,
as
shown
by
the
amelioration
of
perivascular
brosis
in
Picro
Sirius
red
staining
and
deposition
of
brotic
proteins,
TGF-b1
and
bronectin
in
hepatocytes.
However,
there
was
no
signicant
difference
observed
between
the
effects
of
the
two
doses
of
Esculetin
(Fig.
3).
Discussion
The
current
study
demonstrates
the
role
of
Esculetin
in
improving
the
TGF-b1
mediated
hepatic
brosis
associated
with
advancement
of
NAFLD
induced
by
long
term
HFD
feeding
(Fig.
4).
This
anti-brotic
effect
may
be
due
to
its
ability
to
activate
Akt/
PI3
K/FoxO1
pathway
as
indicated
by
the
reduction
in
expression
of
phosphorylated
FoxO1.
Esculetin,
at
both
50
and
100
mg/kg/day
po
treatment
for
14
days
showed
a
marked
improvement
in
metabo-
lism
associated
biochemical
parameters,
liver
function
tests,
insulin
resistance
and
brosis.
However,
no
signicant
difference
was
observed
between
the
effects
elicited
by
the
two
doses
of
Esculetin.
According
to
the
multiple
hit
hypothesis,
multiple
factors
interact
for
the
pathogenesis
of
NAFLD.
The
rst
hit
is
the
accumulation
of
fat
in
hepatocytes
mostly
due
to
IR
[3].
Chronic
HFD
feeding
leads
to
fat
accumulation
in
the
liver
which
is
closely
associated
with
metabolic
derangements
related
to
obesity
and
insulin
resistance
shown
by
metabolic
abnormalities
such
as
altered
plasma
biochemical
parameters
like
plasma
glucose,
triglyceride,
cholesterol
and
liver
function
tests.
Thus,
chronic
HFD
feeding
has
been
found
to
be
an
appropriate
method
for
induction
of
NAFLD
in
murine
model
[9].
In
this
study,
we
found
that
long
term
HFD
feeding
mimics
the
clinical
features
of
human
NAFLD.
These
pathological
features
could
be
attenuated
signi-
cantly
by
the
treatment
with
Esculetin,
thus
conrming
its
potential
to
be
an
efcient
anti-hyperglycemic,
insulin
sensitizing
and
hepato-protective
agent.
The
subsequent
hits
for
progression
of
the
liver
disease
involve
a
combination
of
oxidative
stress,
lipid
peroxidation,
and
release
of
Fig
1.
(A)
Microscopic
changes
in
the
rat
liver
fed
with
high
fat
diet
and
effect
of
Esculetin,
using
hematoxylin
and
eosin
staining
(40).The
oxidative
stress
parameters:
(B)
GSH
and
(C)
TBARS.
Note:
Data
represented
in
mean
SEM
(n
=
6).
a
p<0.001
vs.
NC,
b
p
<
0.001
vs.
HFD.
(For
interpretation
of
the
references
to
colour
in
this
gure,
the
reader
is
referred
to
the
web
version
of
this
article.)
A.
Pandey
et
al.
/
Pharmacological
Reports
69
(2017)
666672
669
inammatory
mediators
[25].
The
initiation
and
perpetuation
of
cell
injury
in
NAFLD
is
associated
with
the
increase
of
free
radicals
and
the
depletion
of
endogenous
anti-oxidant
defence
both
in
human
and
rodents.
In
the
current
study
we
found
that
Esculetin
could
control
the
abrupt
rise
in
free
radical
stress
by
limiting
lipid
peroxidation
and
improving
GSH
in
the
liver
of
the
HFD
fed
rats.
Increased
oxidative
stress
leads
to
the
activation
of
stress
kinases
that
induce
insulin
resistance
by
serine
phosphorylation
of
IRS
proteins
and
Akt
pathway
[26].
Activated
Akt
plays
an
essential
role
in
regulating
renal
cells
proliferation,
broblast
activation
and
matrix
production
culminating
into
brosis
in
chronic
kidney
diseases
[27].
NAFLD
encompasses
a
spectrum
of
histopathological
condi-
tions,
ranging
from
simple
steatosis
to
NASH
and
cirrhosis,
with
risk
of
hepatocellular
carcinoma.
NASH
is
considered
the
more
aggressive
part
of
NAFLD
spectrum,
associated
with
progressive
brosis
as
well
as
higher
risk
of
cardiovascular
disease
[28].
Fibrosis
characterized
by
derangements
in
synthesis
and
degradation
of
ECM,
macrophage/lymphocyte
inltration,
mesan-
gial
cells
and
podocyte
apoptosis,
accumulation
of
activated
broblasts,
epithelial-mesenchymal
transition
[27].
If
brosis
continues
unopposed,
it
disrupts
the
normal
architecture
of
the
liver
and
alters
its
normal
functioning
[29].
Recently,
brosis
stage
has
been
suggested
to
predict
liver-specic
mortality
more
reliably
than
NAFLD
activity
score
[30].
Previous
studies
demonstrated
that
PI3
K/Akt/mammalian
target
of
Rapamycin
(mTOR),
a
non-Smad
TGF-b
pathway,
plays
an
important
role
in
the
brosis
of
different
organs
such
as
the
lung,
kidney,
skin
and
liver.
The
activation
of
PI3
K/Akt/mTOR
pathway
also
promotes
collagen
production
and
scar
formation
in
the
acute
contused
skeletal
muscle
[31].
Akt
phosphorylates
FoxO1,
one
of
the
most
widely
expressed
sub-
families
of
the
winged
helix
forkhead
factors,
at
Ser-253
and
enhances
FoxO1
export
into
the
cytoplasm
[32,33].
The
protein
14-
3-3
changes
the
subcellular
localisation
by
affecting
the
structural
properties
of
the
target
protein
or
by
obstructing
the
scaffolding
[3436].
This
prompts
an
interaction
between
Exportin
and
the
Fig.
2.
Immunostaining
for
phospho-FoxO1
expression
in
high
fat
diet
rat
liver
and
its
evaluation
(40).
Note:
Data
represented
in
mean
SEM
(n
=
6).
a
p
<
0.001
vs.
NC,
b
p
<
0.001
vs.
HFD.
(For
interpretation
of
the
references
to
colour
in
this
gure,
the
reader
is
referred
to
the
web
version
of
this
article.)
Fig.
3.
Effect
of
Esculetin
on
brosis
in
HFD
fed
rats
liver
(A)
Picro
Sirius
red
staining
(B)
TGF-
b
1
immunostaining
and
evaluation
(C)
Fibronectin
immunostaining
and
evaluation
(40).
Note:
Data
represented
in
mean
SEM
(n
=
6).
a
p
<
0.001
vs.
NC,
b
p
<
0.001
vs.
HFD.
(For
interpretation
of
the
references
to
colour
in
this
gure
legend,
the
reader
is
referred
to
the
web
version
of
this
article.)
670
A.
Pandey
et
al.
/
Pharmacological
Reports
69
(2017)
666672
nuclear
export
signal
and
thereby
prevents
re-entry
of
FoxO
factors
into
the
nucleus
by
masking
the
nuclear
localisation
sequence
[36,37].
FoxO1,
is
biologically
omni-functional
owing
to
its
far-
ung
roles
in
metabolism,
cell
cycle,
tissue
differentiation
and
development
and
oxidative
stress
response
[38].
Existing
reports
indicate
an
important
link
between
oxidative
stress,
FoxO1
activity
and
TGF-b1
mediated
brotic
cascade.
A
study
showed
that
FoxO1
facilitates
reduction
of
oxidative
stress
in
keratinocytes
to
maintain
cell
migration
and
prevent
cell
death
in
a
TGF-b1
independent
manner
and
also
up
regulates
TGF-b1
to
promote
wound
healing
[39,40].
In
a
study
of
HSCs
and
liver
brosis,
inactivation
of
FoxO1,
prompted
by
hyperinsulinemia
was
found
to
promote
proliferation
and
trans-differentiation
of
HSCs
resulting
in
the
increase
in
susceptibility
of
FoxO1+/
mice
to
experimentally
induced
liver
brosis
[4].
The
current
study
demonstrated
that
Esculetin
signicantly
reduces
brosis
in
chronic
HFD
fed
rats
by
regulating
TGF-b1
expression,
which
may
be
orchestrated
through
its
effect
on
Akt/PI3
K/FoxO1
pathway.
As
per
the
previous
studies,
a
potential
therapy
for
NAFLD
may
be
the
one
with
multi-faceted
actions
on
insulin
resistance,
oxidative
stress,
inammation
and
brosis.
To
the
best
of
our
knowledge,
this
is
the
rst
report
showing
the
ability
of
Esculetin
to
ameliorate
hepatic
brosis
in
NAFLD
and
its
effect
on
FoxO1
activity.
Thus,
Esculetin,
having
a
prominent
anti-oxidant
and
insulin
sensitizing
effect
appears
to
be
a
drug
of
choice
for
the
treatment
of
NAFLD
and
this
applicability
is
further
promoted
by
the
results
of
this
study
which
prove
it
to
be
an
anti-brotic
and
hepato-protective
agent.
Further
studies
need
to
be
done
to
elaborate
the
detailed
mechanism
through
which
Esculetin
elicits
hepato-protection
in
HFD
induced
NAFLD.
Conict
of
interests
We
declare
that
we
have
no
conict
of
interest.
Acknowledgements
A.P.
acknowledges
a
Ph.D.
fellowship
from
DST
-
INSPIRE
[IF131160].
A.B.G
sincerely
acknowledges
the
nancial
support
obtained
from
the
Science
&
Engineering
Research
Board
Department
of
Science
&
Technology
(SERB-DST),
Govt.
of
India
(SB/EMEQ-053/2013)
and
University
Grant
Commission
Major
Research
Project(UGC-MRP)
[F.NO.
42-702/2013
(SR)]
for
the
research
work.
References
[1]
Bellentani
S,
Scaglioni
F,
Marino
M,
Marino
Gc.
Epidemiology
of
non-alcoholic
fatty
liver
disease.
Dig
Dis
2010;28:15561.
[2]
Marrero
JA,
Fontana
RJ,
Su
GL,
Conjeevaram
HS,
Emick
DM,
Lok
AS.
NAFLD
may
be
a
common
underlying
liver
disease
in
patients
with
hepatocellular
carcinoma
in
the
United
States.
Hepatology
2002;36:134954.
[3]
Bataller
R,
Brenner
DA.
Liver
brosis.
J
Clin
Investig
2005;115:20918.
[4]
Adachi
M,
Osawa
Y,
Uchinami
H,
Kitamura
T,
Accili
D,
Brenner
DA.
The
forkhead
transcription
factor
FoxO1
regulates
proliferation
and
transdifferentiation
of
hepatic
stellate
cells.
Gastroenterology
2007;132:143446.
[5]
Takahashi
Y,
Sugimoto
K,
Inui
H,
Fukusato
T.
Current
pharmacological
therapies
for
nonalcoholic
fatty
liver
disease/nonalcoholic
steatohepatitis.
World
J
Gastroenterol
2015;21:377785.
[6]
Del
Prete
A,
Scalera
A,
Iadevaia
MD,
Miranda
A,
Zulli
C,
Gaeta
L,
et
al.
Herbal
products:
benets,
limits,
and
applications
in
chronic
liver
disease.
Evid
Based
Complement
Alternat
Med
2012;2012:119.
[7]
Sim
O,
Lee
HI,
Ham
JR,
Seo
I,
Lee
MK.
Long-term
supplementation
of
esculetin
ameliorates
hepatosteatosis
and
insulin
resistance
partly
by
activating
AdipoR2-AMPK
pathway
in
diet-induced
obese
mice.
J
Funct
Food
2015;15:16071.
[8]
Choi
Y,
Ham
JR,
Lee
K.
Esculetin
prevents
non-alcoholic
fatty
liver
in
diabetic
mice
fed
high-fat
diet.
Chem
Biol
Interact
2016;260:1321.
[9]
Xu
J,
Fan
JG,
Ding
XD,
Qiao
L,
Wang
L.
Characterization
of
high-fat,
diet-
induced,
non-alcoholic
steatohepatitis
with
brosis
in
rats.
Dig
Dis
Sci
2010;55:93140.
[10]
Kucera
O,
Cervinkova
Z.
Experimental
models
of
non-alcoholic
fatty
liver
disease
in
rats.
World
J
Gastroenterol
2014;20:836476.
[11]
Kadakol
A,
Malek
V,
Goru
SK,
Pandey
A,
Gaikwad
AB.
Esculetin
reverses
histone
H2A/H2
B
ubiquitination,
H3
dimethylation,
acetylation
and
phosphorylation
in
preventing
type
2
diabetic
cardiomyopathy.
J
Funct
Foods
2015;17:12736.
Fig.
4.
Summary:
High
fat
diet
feeding
leads
to
an
increase
in
oxidative
stress
and
insulin
resistance
which
activate
Akt
pathway.
Activated
Akt
phosphorylates
FoxO1
leading
to
its
exclusion
from
the
nucleus
and
preventing
FoxO1-dependent
gene
expression
of
anti-oxidant
and
cell
cycle
arrest
genes.
This
leads
to
an
abrupt
increase
in
cell
proliferation
and
brosis
further
promoting
the
expression
collagen,
TGF-
b
1
and
bronectin.
Esculetin
treatment
prevents
oxidative
stress,
insulin
resistance
and
hyperinsulinemia
to
inhibit
the
triggering
of
Akt/FoxO1
pathway.
Thus,
the
study
concludes
that
Esculetin
ameliorates
hepatic
brosis
in
high
fat
diet
induced
non-alcoholic
fatty
liver
disease
by
regulation
of
FoxO1
mediated
pathway.
A.
Pandey
et
al.
/
Pharmacological
Reports
69
(2017)
666672
671
[12]
Pandey
A,
Goru
SK,
Kadakol
A,
Malek
V,
Gaikwad
AB.
Differential
regulation
of
angiotensin
converting
enzyme
2
and
nuclear
factor-
k
B
by
angiotensin
II
receptor
subtypes
in
type
2
diabetic
kidney.
Biochimie
2015;118:7181.
[13]
Srinivasan
K,
Viswanad
B,
Asrat
L,
Kaul
CL,
Ramarao
P.
Combination
of
high-fat
diet-fed
and
low-dose
streptozotocin-treated
rat:
a
model
for
type
2
diabetes
and
pharmacological
screening.
Pharmacol
Res
2005;52:31320.
[14]
Kadakol
A,
Malek
V,
Goru
SK,
Pandey
A,
Bagal
S,
Gaikwad
AB.
Esculetin
attenuates
alterations
in
Ang
II
and
acetylcholine
mediated
vascular
reactivity
associated
with
hyperinsulinemia
and
hyperglycemia.
Biochem
Biophys
Res
Commun
2015;461:3427.
[15]
Sattar
N,
Forrest
E,
Preiss
D.
Non-alcoholic
fatty
liver
disease.
BMJ
2014;349:1
8.
[16]
Kotoh
K,
Enjoji
M,
Kato
M,
Kohjima
M,
Nakamuta
M,
Takayanagi
R.
A
new
parameter
using
serum
lactate
dehydrogenase
and
alanine
aminotransferase
level
is
useful
for
predicting
the
prognosis
of
patients
at
an
early
stage
of
acute
liver
injury:
a
retrospective
study.
Comp
Hepatol
2008;7:16.
[17]
Botros
M,
Sikaris
KA.
The
de
ritis
ratio:
the
test
of
time.
Clin
Biochem
Rev
2013;34:11730.
[18]
Holman
R,
Hines
G,
Kennedy
I,
Stevens
R,
Matthews
D,
Levy
J.
A
calculator
for
HOMA.
Diabetologia
2004;47:A222.
[19]
Calculator
H.
The
oxford
center
for
diabetes.
Endocrinology
&
metabolism
Diabetes
Trial
Unit.
.
[20]
Ohkawa
H,
Ohishi
N,
Yagi
K.
Assay
for
lipid
peroxides
in
animal
tissues
by
thiobarbituric
acid
reaction.
Anal
Biochem
1979;95:3518.
[21]
Boyne
AF,
Ellman
GL.
A
methodology
for
analysis
of
tissue
sulfhydryl
components.
Anal
Biochem
1972;46:63953.
[22]
Ninichuk
V,
Clauss
S,
Kulkarni
O,
Schmid
H,
Segerer
S,
Radomska
E,
et
al.
Late
onset
of
CCl2
blockade
with
the
Spiegelmer
mNOX-E36-3
0
PEG
prevents
glomerulosclerosis
and
improves
glomerular
ltration
rate
in
db/db
mice.
Am
J
Path
2008;172:62837.
[23]
Tsuda
M,
Zhang
W,
Yang
GX,
Tsuneyama
K,
Ando
Y,
Kawata
K,
et
al.
Deletion
of
interleukin
(IL)
12p35
induces
liver
brosis
in
dominant
negative
TGF
b
receptor
type
II
mice.
Hepatology
2013;57:80616.
[24]
Bonora
E,
Formentini
G,
Calcaterra
F,
Lombardi
S,
Marini
F,
Zenari
L,
et
al.
HOMA-estimated
insulin
resistance
is
an
independent
predictor
of
cardiovascular
disease
in
type
2
diabetic
subjects.
Diabetes
Care
2002;25:113541.
[25]
Anstee
QM,
Goldin
RD.
Mouse
models
in
non
alcoholic
fatty
liver
disease
and
steatohepatitis
research.
Int
J
Exp
Pathol
2006;87:116.
[26]
Zhang
Y,
Yang
JH.
Activation
of
the
PI3
K/Akt
pathway
by
oxidative
stress
mediates
high
glucose-induced
increase
of
adipogenic
differentiation
in
primary
rat
osteoblasts.
J
Cell
Biochem
2013;114:259525602.
[27]
Lan
A,
Du
J.
Potential
role
of
Akt
signaling
in
chronic
kidney
disease.
Nephrol
Dial
Transplant
2014;30:38594.
[28]
Ekstedt
M,
Hagström
H,
Nasr
P,
Fredrikson
M,
Stål
P,
Kechagias
S,
et
al.
Fibrosis
stage
is
the
strongest
predictor
for
disease-specic
mortality
in
NAFLD
after
up
to
33
years
of
follow-up.
Hepatology
2015;61:154754.
[29]
Wanless
IR.
Pathogenesis
of
cirrhosis.
J
Gastroenterol
Hepatol
2004;19:S369
71.
[30]
Younossi
ZM,
Stepanova
M,
Raq
N,
Makhlouf
H,
Younoszai
Z,
Agrawal
R,
et
al.
Pathologic
criteria
for
nonalcoholic
steatohepatitis:
interprotocol
agreement
and
ability
to
predict
liver-related
mortality.
Hepatology
2011;53:187482.
[31]
Li
H-Y,
Zhang
Q-G,
Chen
J-W,
Chen
S-Q,
Chen
S-Y.
The
brotic
role
of
phosphatidylinositol-3-kinase/Akt
pathway
in
injured
skeletal
muscle
after
acute
contusion.
Int
J
Sports
Med
2013;34:78994.
[32]
Matsuzaki
H,
Daitoku
H,
Hatta
M,
Aoyama
H,
Yoshimochi
K,
Fukamizu
A.
Acetylation
of
Foxo1
alters
its
DNA-binding
ability
and
sensitivity
to
phosphorylation.
Proc
Natl
Acad
Sci
U
S
A
2005;102:1127883.
[33]
Daitoku
H,
Sakamaki
J-i,
Fukamizu
A.
Regulation
of
FoxO
transcription
factors
by
acetylation
and
proteinprotein
interactions.
BBA
Mol
Cell
Res
1813;2011:195460.
[34]
Fu
H,
Subramanian
RR,
Masters
SC.
14-3-3
proteins:
structure,
function,
and
regulation.
Annu
Rev
Pharmacol
Toxicol
2000;40:61747.
[35]
Benayoun
BA,
Caburet
S,
Veitia
RA.
Forkhead
transcription
factors:
key
players
in
health
and
disease.
Trends
Genet
2011;27:22432.
[36]
Daitoku
H,
Fukamizu
A.
FOXO
transcription
factors
in
the
regulatory
networks
of
longevity.
J
Biochem
2007;141:76974.
[37]
Yang
X,
Lee
WH,
Sobott
F,
Papagrigoriou
E,
Robinson
CV,
Grossmann
JG,
et
al.
Structural
basis
for
proteinprotein
interactions
in
the
14-3-3
protein
family.
Proc
Natl
Acad
Sci
U
S
A
2006;103:1723742.
[38]
Pandey
A,
Kumar
GS,
Kadakol
A,
Malek
V,
Bhanudas
Gaikwad
A.
FoxO1
inhibitors:
the
future
medicine
for
metabolic
disorders.
Curr
Diabetes
Rev
2016;12:22330.
[39]
Ponugoti
B,
Xu
F,
Zhang
C,
Tian
C,
Pacios
S,
Graves
DT.
FOXO1
promotes
wound
healing
through
the
up-regulation
of
TGF-
b
1
and
prevention
of
oxidative
stress.
J
Cell
Biol
2013;203:32743.
[40]
Hameedaldeen
A,
Liu
J,
Batres
A,
Graves
GS,
Graves
DT.
FOXO1,
TGF-
b
regulation
and
wound
healing.
Int
J
Mol
Sci
2014;15:1625769.
672
A.
Pandey
et
al.
/
Pharmacological
Reports
69
(2017)
666672
... Among them, esculetin is an important coumarin derivative with a common structure of 6,7-dihydroxy-2-H-1-benzopyran-2one. Studies have shown that esculetin has a variety of biological activities, such as anti-inflammatory [11,12], antibacterial [13,14], antiviral [15], antitumor [16], liver protection [17][18][19], antifungal [20]. In vivo studies of esculetin showed improvement in hepatic fibrosis by increasing phospho-Forkhead box protein O1 (FOXO1) expression at concentrations of 50 mg/kg or 100 mg/kg [19]. ...
... Studies have shown that esculetin has a variety of biological activities, such as anti-inflammatory [11,12], antibacterial [13,14], antiviral [15], antitumor [16], liver protection [17][18][19], antifungal [20]. In vivo studies of esculetin showed improvement in hepatic fibrosis by increasing phospho-Forkhead box protein O1 (FOXO1) expression at concentrations of 50 mg/kg or 100 mg/kg [19]. In vitro antiviral experiments with esculetin revealed that esculetin concentrations of 32 μM inhibited Newcastle disease virus replication well [15]. ...
Article
Full-text available
Previous in vivo and in vitro studies revealed that esculetin (Fig. 1) has anti-hepatitis B virus (anti-HBV) activity as well as a protective effect on liver damage caused by duck hepatitis B virus. We designed and synthesized a series of esculetin derivatives, introduced side chains containing various amino groups into site 7 of the parent structure, and synthesized C-4 and C-8 substituted derivatives with the goal of investigating their anti-HBV activities. In vitro anti-HBV activity was performed against HepG2.2.15 cells by using Enzyme-Linked Immunosorbent Assay(ELISA) kit and cytotoxicity was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay with lamivudine as the positive control. The results demonstrated that several compounds showed moderate anti-HBV activity, while the introduction of morpholine groups could significantly inhibit the expression of hepatitis B e antigen (HBeAg) and the introduction of the 2-methylimidazole group could significantly inhibit the expression of Hepatitis B surface antigen (HBsAg). Among all tested compounds, compound 4a demonstrated the best anti-HBeAg activity (IC50 = 15.8 ± 4.2 μM), while compound 6d demonstrated the best anti-HBsAg activity (IC50 = 21.4 ± 2.8 μM). Compounds 6b and 6c showed moderate anti-HBV activity and HBsAg inhibition. Compounds 4b showed moderate anti-HBV activity and an inhibitory effect on HBeAg. In addition, compounds 4a, 4c, 4d, 6b, 6c and 6d showed improved metabolic stability. This study provides useful guidance for the discovery of anti-HBV drugs, which merits further investigation. Graphical Abstract
... Among these constituents, alkaloids and coumarins are believed to constitute the principal material foundation of BTWD. Coptisine chloride, berberine, columbamine, phellodendrine, palmatine, obacunone, esculetin, fraxetin, esculin, fraxin, and pulchinenoside B 4 have been recognized as the primary active constituents, exhibiting a broad range of pharmacological efects including antioxidant [3], anti-infammatory [4], antigastrointestinal cancer [5,6], hepatic fbrosis amelioration [7], gastroprotective [8], and intestinal epithelial barrier protective activity [9]. ...
Article
Full-text available
Traditional Chinese medicine (TCM) serves as a significant adjunct to chemical treatment for chronic diseases. For instance, the administration of Baitouweng decoction (BTWD) has proven effective in the treatment of ulcerative colitis. However, the limited understanding of its pharmacokinetics (PK) has impeded its widespread use. Chinese Bama miniature pigs possess anatomical and physiological similarities to the human body, making them a valuable model for investigating PK properties. Consequently, the identification of PK properties in Bama miniature pigs can provide valuable insights for guiding the clinical application of BTWD in humans. To facilitate this research, a rapid and sensitive UPLC-MS/MS method has been developed for the simultaneous quantification of eleven active ingredients of BTWD in plasma. Chromatographic separation was conducted using an Acquity UPLC HSS T3 C18 column and a gradient mobile phase comprising acetonitrile and water (containing 0.1% acetic acid). The methodology was validated in accordance with the FDA Bioanalytical Method Validation Guidance for Industry. The lower limit of quantitation fell within the range of 0.60–2.01 ng/mL. Pharmacokinetic studies indicated that coptisine chloride, berberine, columbamine, phellodendrine, and obacunone exhibited low Cmax, while fraxetin, esculin, fraxin, and pulchinenoside B4 were rapidly absorbed and eliminated from the plasma. These findings have implications for the development of effective components in BTWD and the adjustment of clinical dosage regimens.
... Phytomedicines and nutraceuticals alone or as adjuvant therapies with conventional drugs have exhibited beneficial effects in various diseases along with minimal adverse effects [54][55][56]. Likewise, esculetin, a coumarin derivative (present in Citrus limonia, Cortex fraxini, as well as Artemisia capillaris), and phloretin, which is a dihydrogen chalcone flavonoid (abundantly found in apples and strawberries), have been reported with anti-diabetic, anti-inflammatory, and anti-oxidative activity [18,21,54,57,58]. However, it is unknown till date whether esculetin and phloretin in combination could have a protective effect against AKI-diabetes comorbidity. ...
Article
Full-text available
Acute kidney injury (AKI) is a global health concern with high incidence and mortality, where diabetes further worsens the condition. The available treatment options are not uniformly effective against the complex pathogenesis of AKI–diabetes comorbidity. Hence, combination therapies based on the multicomponent, multitarget approach can tackle more than one pathomechanism and can aid in AKI–diabetes comorbidity management. This study aimed to investigate the therapeutic potential of esculetin and phloretin combination against AKI–diabetes comorbidity by network pharmacology followed by validation by molecular docking and dynamics. The curative targets for diabetes, AKI, esculetin, and phloretin were obtained from DisGeNET, GeneCards, SwissTargetPrediction database. Further, the protein–protein interaction of the potential targets of esculetin and phloretin against AKI–diabetes comorbidity was investigated using the STRING database. Gene ontology and pathway enrichment analysis were performed with the help of the DAVID and KEGG databases, followed by network construction and analysis via Cytoscape. Molecular docking and dynamic simulations were performed to validate the targets of esculetin and phloretin against AKI–diabetes comorbidity. We obtained 6341 targets for AKI–diabetes comorbidity. Further, a total of 54 and 44 targets of esculetin and phloretin against AKI–diabetes comorbidity were retrieved. The top 10 targets for esculetin selected based on the degree value were AKR1B1, DAO, ESR1, PLK1, CA3, CA2, CCNE1, PRKN, HDAC2, and MAOA. Similarly, phloretin’s 10 key targets were ACHE, CDK1, MAPK14, APP, CDK5R1, CCNE1, MAOA, MAOB, HDAC6, and PRKN. These targets were enriched in 58 pathways involved in the pathophysiology of AKI–diabetes comorbidity. Further, esculetin and phloretin showed an excellent binding affinity for these critical targets. The findings of this study suggest that esculetin and phloretin combination as a multicomponent multitarget therapy has the potential to prevent AKI–diabetes comorbidity. Graphical abstract
... additionally, esculetin exhibited renoprotection in diabetic nephropathy by h3-hyperacetylation, and monoubiquitination of h2aK119 [19]. hepatic fibrosis in a high-fat diet-induced murine model of nonalcoholic fatty liver disease (NaFlD) was improved with esculetin treatment by regulating forkhead box protein O1 (FoxO1) signaling [20]. administration of mitochondria-targeted esculetin significantly prevented NaFlD to nonalcoholic steatohepatitis (Nash) progression via aMPK-siRt1 axis modulation [21]. ...
Article
Mitophagy maintains cellular homeostasis by eliminating damaged mitochondria. Accumulated damaged mitochondria can lead to oxidative stress and cell death. induction of the PINK1/Parkin-mediated mitophagy is reported to be renoprotective in acute kidney injury (AKI). Esculetin, a naturally available coumarin, has shown protective action against diabetic complications. However, its effect on AKI-diabetes comorbidity has not been explored yet. Therefore, we aimed to investigate the renoprotective effect of esculetin against AKI under diabetic conditions via regulating PINK1/Parkin-mediated mitophagy. For this, type 1 diabetic male Wistar rats were treated with two doses of esculetin (50 and 100 mg/kg/day orally) for five days followed by AKI induction by bilateral ischemic-reperfusion injury (IRI). NRK-52E cells grown in high glucose were exposed to sodium azide (10 mM) for induction of hypoxia/reperfusion injury (HRI) in-vitro. Esculetin (50 μM) treatment for 24 h was given to the cells before HRI. The in-vitro samples were utilized for cell viability and ΔΨm assay, immunoblotting, and immunofluorescence. Rats’ plasma, urine, and kidney samples were collected for biochemical analysis, histopathology, and western blotting. Our results showed a significant decrease in kidney injury-specific markers and increased expression of mitophagy markers (PINK1 and Parkin) with esculetin treatment. Moreover, esculetin prevented the HRI and hyperglycemia-induced decrease in ΔΨm and autophagosome marker. Also, esculetin therapy reduced oxidative stress via increased Nrf2 and Keap1 expression. Esculetin attenuated AKI under diabetic condition by preventing mitochondrial dysfunction via inducing PINK1/Parkin-mediated mitophagy, suggesting its potential as an effective therapy for preventing AKI-diabetes comorbidity.
... In the setting of liver disorders, the elevation of oxidative radicals and lipid peroxidation can stimulate the synthesis of the noncellular matrix. This process is marked by the heightened multiplication of hepatic perisinusoidal cells and the rapid deterioration of hepatic tissue (47). In hepatocytes, microsomal monooxygenase could metabolize 1,1-dimethylethyl hydroperoxide (also known as t-BHP), a lipid peroxides analogous substance with a shortened chain. ...
Article
Full-text available
Background: Esculetin, scientifically referred to as 6,7-dihydroxycoumarin, functions as the primary bioactive constituent found in Cortex Fraxini (commonly known as ash bark), an ancient Asian medicinal substance. Herbal practitioners utilize the outer layer of the branch or stem bark of Cortex Fraxini for its gentle and safe medicinal properties and its potential as a nutritional component. In contemporary times, the landscape has undergone a notable transformation due to the emergence of a wide range of innovative 6,7-dihydroxycoumarin derivatives. The recent surge of innovation has sparked a heightened interest in understanding the molecular mechanisms that underlie the effects of Cortex Fraxini and 6,7-dihydroxycoumarin in clinical applications. Aim: This succinct review seeks to build up the extensive knowledge accumulated in the past decade concerning the synthesis, pharmacological profiles and principles linked to 6,7-dihydroxycoumarin and its chemical analogues. Furthermore, we aim to provide a concise yet inclusive overview of the unique characteristics of 6,7-dihydroxycoumarin. Conclusion: Satisfying these aims can enhance the comprehension of the diverse possibilities presented by this chemical and its related compounds across different research and application domains.
... Our investigation revealed the presence of coumarins, sterols and flavonoids, compounds with established hepatoprotective and radical-scavenging properties (Mahmoud et al., 2017b;Sayed et al., 2020). For instance, aesculetin attenuated inflammation by inhibiting NF-rB both in vitro and in vivo (Wang et al., 2022), and ameliorated oxidative stress and liver injury caused by various agents (Pandey et al., 2017, Mohamadi-Zarch et al., 2021. Fucosterol showed antioxidant properties and prevented NF-rB activation and acute liver injury in mice (Mo et al., 2018). ...
Article
Full-text available
Cisplatin (CIS) is a chemotherapeutic medication for the treatment of cancer. However, hepatotoxicity is among the adverse effects limiting its use. Caroxylon salicornicum is traditionally used for treating inflammatory diseases. In this investigation, three flavonoids, four coumarins, and three sterols were detected in the petroleum ether fraction of C. salicornicum (PEFCS). The isolated phytochemicals exhibited binding affinity toward Keap1, NF-κB, and SIRT1 in silico. The hepatoprotective role of PEFCS (100, 200 and 400 mg/kg) was investigated in vivo. Rats received PEFCS for 14 days and CIS on day 15. CIS increased ALT, AST and ALP and caused tissue injury along with increased ROS, MDA, and NO. Hepatic NF-κB p65, pro-inflammatory mediators, Bax and caspase-3 were increased in CIS-treated animals while antioxidants and Bcl-2 were decreased. PEFCS mitigated hepatocyte injury, and ameliorated transaminases, ALP, oxidative stress (OS) and inflammatory markers. PEFCS downregulated pro-apoptosis markers and boosted Bcl-2 and antioxidants. In addition, PEFCS upregulated Nrf2, HO-1, and SIRT1 in CIS-administered rats. In conclusion, PEFCS is rich in beneficial phytoconstituents and conferred protection against liver injury by attenuating OS and inflammation and upregulating Nrf2 and SIRT1.
... Kadakol et al. reported the renoprotective role of nutraceutical esculetin against type 2 diabetic nephropathy and cardiomyopathy by reversing histone H3 acetylation and dimethylation, H2A lysine 119 monoubiquitination, and H2A/H2B ubiquitination [28,29]. Our group observed the morphological and microscopic alterations in liver structure, liver functioning, and liver fibrosis were significantly attenuated in non-alcoholic fatty liver disease (NAFLD) rats by esculetin treatment [30]. Additionally, esculetin is documented to reduce hyperglycemia and hyperinsulinemia-related modifications in angiotensin II and acetylcholine-mediated vascular responsiveness [31]. ...
Article
Acute kidney injury (AKI) has become a global health issue, with ~12 million reports yearly, resulting in a persistent increase in morbidity and mortality rates. AKI pathophysiology is multifactorial involving oxidative stress, mitochondrial dysfunction, epigenetic modifications, inflammation, and eventually, cell death. Hence, therapies able to target multiple pathomechanisms can aid in AKI management. To change the drug discovery framework from "one drug, one target" to "multicomponent, multitarget," network pharmacology is evolving as a next-generation research approach. Researchers have used the network pharmacology approach to predict the role of nutraceuticals against different ailments including AKI. Nutraceuticals (herbal products, isolated nutrients, and dietary supplements) belong to the pioneering category of natural products and have shown protective action against AKI. Nutraceuticals have recently drawn attention because of their ability to provide physiological benefits with less toxic effects. This review emphasizes the nutraceuticals that exhibited renoprotection against AKI and can be used either as monotherapy or adjuvant with conventional therapies to boost their effectiveness and lessen the adverse effects. Additionally, the study sheds light on the application of network pharmacology as a cost-effective and time-saving approach for the therapeutic target prediction of nutraceuticals against AKI.
Article
Full-text available
Excessive accumulation of extracellular matrix (ECM) components within the liver leads to a pathological condition known as liver fibrosis. Alcohol abuse, non-alcoholic fatty liver disease (NAFLD), autoimmune issues, and viral hepatitis cause chronic liver injury. Exploring potential therapeutic targets and understanding the molecular mechanisms involved in liver fibrosis are essential for the development of effective interventions. The goal of this comprehensive review is to explain how the PI3K/AKT signaling pathway contributes to the reduction of liver fibrosis. The potential of this pathway as a therapeutic target is investigated through a summary of results from in vivo and in vitro studies. Studies focusing on PI3K/AKT activation have shown a significant decrease in fibrosis markers and a significant improvement in liver function. The review emphasizes how this pathway may prevent ECM synthesis and hepatic stellate cell (HSC) activation, ultimately reducing the fibrotic response. The specific mechanisms and downstream effectors of the PI3K/AKT pathway in liver fibrosis constitute a rapidly developing field of study. In conclusion, the PI3K/AKT signaling pathway plays a significant role in attenuating liver fibrosis. Its complex role in regulating HSC activation and ECM production, demonstrated both in vitro and in vivo, underscores its potential as a effective therapeutic approach for managing liver fibrosis and slowing disease progression. A comprehensive review of this field provides valuable insights into its future developments and implications for clinical applications.
Article
Background: Almost all chronic liver diseases cause fibrosis, which can lead to cirrhosis and eventually liver cancer. Liver fibrosis is now considered to be a reversible pathophysiological process and suppression of fibrosis is necessary to prevent liver cancer. At present, no specific drugs have been found that have hepatic anti-fibrotic activity. Objective: The research progress of anti-hepatic fibrosis compounds in recent ten years was reviewed to provide a reference for the design and development of anti-hepatic fibrosis drugs. Methods: According to the structure of the compounds, they are divided into monocyclic compounds, fused-heterocyclic compounds, and acyclic compounds. Results: In this article, the natural products and synthetic compounds with anti-fibrotic activity in recent ten years were reviewed, with emphasis on their pharmacological activity and structure-activity relationship (SAR). Conclusion: Most of these compounds are natural active products and their derivatives, and there are few researches on synthetic compounds and SAR studies on natural product.
Article
Full-text available
Nonalcoholic fatty liver disease (NAFLD)/nonalcoholic steatohepatitis (NASH) is considered to be a hepatic manifestation of metabolic syndrome, and its incidence is rapidly increasing worldwide. It is currently the most common chronic liver disease. NASH can progress to liver cirrhosis and hepatocellular carcinoma, and may result in liver-related death. Currently, the principal treatment for NAFLD/NASH is lifestyle modification by diet and exercise. However, pharmacological therapy is indispensable because obese patients with NAFLD often have difficulty maintaining improved lifestyles. The pathogenesis of NAFLD/NASH has not been completely elucidated. However, insulin resistance, inflammatory cytokines, and oxidative stress are thought to be important in the development and/or progression of the disease. Currently, insulin sensitizers (thiazolidinediones) and antioxidants (vitamin E) seem to be the most promising therapeutic agents for NAFLD/NASH, and lipid-lowering drugs, pentoxifylline, angiotensin receptor blockers, and n-3 polyunsaturated fatty acids also have promise. However, there is a lack of consensus regarding the most effective and appropriate pharmacotherapy for NAFLD/NASH. Animal experiments suggest that herbal medicines and natural products may be promising therapeutic agents for NAFLD/NASH, but their efficacy and safety are yet to be investigated in human studies. In this paper, we review the existing and potential pharmacological therapies for NAFLD/NASH.
Article
Full-text available
#### Summary points Non-alcoholic fatty liver disease (NAFLD) is now more common than alcoholic liver disease owing to the rapid rise in the prevalence of obesity,1 and NAFLD is the most common cause of abnormal liver function tests.2 Its prevalence worldwide is thought to be approximately 20% in the general population and up to 70% in patients with type 2 diabetes mellitus.3 The first recognisable stage of NAFLD is hepatic steatosis, when fat content exceeds 5% of liver volume. Simple steatosis is usually benign in terms of risk of progression to …
Article
Full-text available
Re-epithelialization is a complex process that involves migration and proliferation of keratinocytes, in addition to the production of cytokines and growth factors that affect other cells. The induction of transcription factors during these processes is crucial for successful wound healing. The transcription factor forkhead boxO-1 (FOXO1) has recently been found to be an important regulator of wound healing. In particular, FOXO1 has significant effects through regulation of transforming growth factor-beta (TGF-β) expression and protecting keratinocytes from oxidative stress. In the absence of FOXO1, there is increased oxidative damage, reduced TGF-β1 expression, reduced migration and proliferation of keratinocytes and increased keratinocytes apoptosis leading to impaired re-epithelialization of wounds.
Article
The increase in Non-alcoholic Fatty Liver Disease (NAFLD) and the imminent disappearance of chronic viral hepatitis thanks to new and effective therapies is motivating hepatologists to change their clinical approach to chronic liver disease. NAFLD-cirrhosis or NAFLD-Hepatocellular Carcinoma (HCC) are now the second cause of liver transplantation in the USA. This short-review is focused to the epidemiology of NAFLD/Non-alchoholic Steatohepatitis (NASH), including the definition of this disease which should be revised as well discussing the prevalence, risk factors for progression, natural history and mortality. NAFLD is considered to be the hepatic manifestation of the metabolic syndrome (MS). It affects 25-30% of the general population and the risk factors are almost identical to those of MS. The natural history involves either the development of cardiovascular diseases or cirrhosis and HCC. HCC can also develop in NASH in the absence of cirrhosis (45% of cases). We conclude that an international consensus conference on the definition, natural history, policies of surveillance and new pharmacological treatments of NAFLD and NASH is urgently needed. © 2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Article
This study investigated the effects and mechanism of esculetin (6,7-dihydroxycoumarin) on non-alcoholic fatty liver in diabetic mice fed high-fat diet (HFD). The diabetic mice model was induced by injection of streptozotocin, after which they were fed HFD diet with or without esculetin for 11 weeks. Non-diabetic mice were provided a normal diet. Diabetes induced hepatic hypertrophy, lipid accumulation and droplets; however, esculetin reversed these changes. Esculetin treatment in diabetic mice fed HFD significantly down-regulated expression of lipid synthesis genes (Fasn, Dgat2 and Pap) and inflammation genes (Tlr4, Myd88, Nfkb, Tnfα and Il6). Moreover, the activities of hepatic lipid synthesis enzymes (fatty acid synthase and phosphatidate phosphohydrolase) and gluconeogenesis enzyme (glucose-6-phosphatase) in the esculetin group were decreased compared with the diabetic group. In addition, esculetin significantly reduced blood HbA1c, serum cytokines (TNF-α and IL-6) and chemokine (MCP-1) levels compared with the diabetic group without changing the insulin content in serum and the pancreas. Hepatic SOD activity was lower and lipid peroxidation level was higher in the diabetic group than in the normal group; however, esculetin attenuates these differences. Overall, these results demonstrated that esculetin supplementation could protect against development of non-alcoholic fatty liver in diabetes via regulation of lipids, glucose and inflammation.
Article
Angiotensin II (Ang II) acts through Angiotensin Converting Enzyme (ACE)/Ang II type 1 receptor (AT1R) axis to promote renal failure whereas the Ang II type 2 receptor (AT2R)/Angiotensin Converting Enzyme 2 (ACE2)/Ang1-7/Mas axis constitutes the protective arm of Renin Angiotensin System (RAS). Though Ang II has been known to activate the Nuclear Factor-κB (NF-κB) signalling pathway through different receptor subtype(s) in different tissues under various diseases, the subtype orchestrating this stimulation in type 2 diabetic kidney remains elusive. ACE2, a protective monocarboxypeptidase, responsible for conversion of Ang II to Ang1-7, opposes the deleterious effects of RAS pathway but how its expression is altered with blockade of AT1R and AT2R is not yet known. Hence, the present study was conceived to understand the regulation of NF-κB and ACE2 by using specific AT1 and AT2 receptor antagonists in non-genetic model of type 2 diabetic nephropathy. Our results show that the AT1R and AT2R antagonists lead to the activation and repression of NF-κB signalling pathway, respectively which suggests the role of AT1R in NF-κB activation. The blockade of AT2R led to an increase in ACE2 expression, which may be a compensatory response to the drastically increased inflammatory mediators and oxidative stress in the diabetic kidney. To the best of our knowledge, this is the first study showing the differential regulation of NF-κB and ACE2 by Ang II receptor subtypes and thus this study improves our understanding regarding regulation of inflammatory cascade and ACE2 by AT1R and AT2R in type 2 diabetic kidney, which may help in designing novel strategies to combat the disease in future. Copyright © 2015. Published by Elsevier B.V.
Article
FoxO1, one of the most widely expressed sub-families of the winged helix forkhead factors, is biologically 'omni-functional' owing to its far-flung roles in metabolism, cell cycle, tissue differentiation and development and oxidative stress response. The knowledge of involvement of FoxO1 in metabolic disorders has long been there, but the potential target remained underutilized due to unavailability of specific and potent inhibitors. The review provides an insight into the role of FoxO1 in orchestrating metabolic diseases' pathogenesis (including diabetes, its secondary complications and obesity) and compiles the literature on FoxO1 inhibitors. The emergence of various natural molecules and synthesized small molecules like AS1842856 as FoxO1 inhibitors urges us to think further and decide the future course of drug development for the management of metabolic disorders.
Article
Post translational histone modifications (PTHMs) play a pivotal role in pathogenesis of diabetic complications. Esculetin is reported to prevent glomerulosclerosis by reversing PTHMs in diabetic rats, but until now its cardioprotective role is unexplored. Hence, the present study aimed to investigate the effect of esculetin on diabetic cardiomyopathy (DCM) and its associated PTHMs. Insulin resistance (IR) and type 2 diabetic rats' heart had augmented permissive PTHMs which contributed to DCM. Besides this, for the first time we have demonstrated increased histone H2AK119Ub and H2BK120Ub levels in DCM. Esculetin treatment reduced metabolic alterations, hypertension, cardiomyocytes hypertrophy, and fibrosis in the diabetic heart. In addition, esculetin attenuated alteration in the renin–angiotensin system, oxidative stress (Keap1) and cell proliferation (Ki67); thus preventing DCM. Remarkably, esculetin treatment restored normal level of permissive PTHMs and H2A/H2B ubiquitination in IR and diabetic heart which might be the basic mechanism behind its cardioprotective role.
Article
Esculetin (6, 7- dihydroxycoumarin) was found to be protective against hepatic and renal damage associated with Streptozotocin (STZ) induced type 1 diabetes, because of its radical scavenging property. However, there are no reports regarding its effect on vascular dysfunction under hyperinsulinemic and hyperglycemic conditions. Hence, the present study aimed to investigate the effect of esculetin on vascular dysfunction under these conditions. Non-genetic model of hyperinsulinemia and hyperglycemia were developed by high fat diet (HFD) feeding and HFD+ Streptozotocin (STZ, 35mg/kg, I.P) treatment in Wistar rats, respectively. Esculetin was administered at 50 and 100 mg/kg/day (P.O, 2 weeks) doses and biochemical, vascular reactivity and immunohistochemical experiments were performed to assess the effect of esculetin on vascular dysfunctions. Esculetin treatment significantly attenuates metabolic perturbations, alleviates insulin levels in hyperinsulinemic condition. Thoracic aorta of hyperinsulinemic and hyperglycemic rats showed hyper-responsiveness to Ang II mediated contraction and impaired acetylcholine mediated relaxation, and esculetin attenuates alterations in vascular reactivity to Ang II and acetylcholine challenges. In addition, immunohistochemical evaluations revealed that esculetin prevents increase in AT1R, AT2R, Keap1, TGF-β, and decrease in ACE2 expression in aorta of hyperinsulinemic and hyperglycemic rats. Copyright © 2015. Published by Elsevier Inc.
Article
The effects of esculetin on hepatosteatosis and insulin resistance in high-fat diet (HFD)-induced obese mice were investigated. Esculetin (0.02%, w/w) provided to mice fed a HFD (40% kcal from fat) for 12 weeks reduced body weight gain, visceral adiposity, hepatosteatosis, hyperlipidemia and hyperglycemia, whereas it increased plasma adiponectin level and the protein and mRNA expression of hepatic AdipoR2, which led to the activation of AMPK. The protein and mRNA expression of hepatic PPARα were up-regulated by esculetin and those of SREBP-1c were down-regulated. Esculetin elevated expression of hepatic fatty acid oxidation genes (PGC-1α, PPARα, Acsl1 and CPT), while suppressing expression of fatty acid synthesis gene. Anti-insulin resistant effects of esculetin were mediated by increased hepatic GLUT2 and glucokinase mRNA levels and decreased glucose-6-phosphatase mRNA level. These findings suggest that esculetin supplementation has beneficial effects on hepatosteatosis and insulin resistance in HFD-induced obese mice, partly via activation of the AdiopR2–AMPK pathway.