ArticlePDF Available

Anti-Adipogenic Effect of Neferine in 3T3-L1 Cells and Primary White Adipocytes

MDPI
Nutrients
Authors:

Abstract and Figures

Neferine, an alkaloid component extracted from lotus seed embryos, is known for its anti-inflammatory, anticancer, and antioxidant properties. However, the anti-adipogenic activity of neferine has not been thoroughly investigated. In this study, neferine was found to inhibit lipid accumulation in a dose-dependent manner during the differentiation of 3T3-L1 cells without inducing cytotoxicity. Real-time polymerase chain reaction and immunoblot analysis revealed the downregulation in the expression of peroxisome proliferator activated receptor gamma (PPARγ), CCAAT/enhancer-binding protein alpha (C/EBPα), sterol regulatory element-binding protein-1c (SREBP-1c), and fatty acid synthase (FAS) and the upregulation in carnitine palmitoyltransferase-1 (CPT-1) and sirtuin 1 (SIRT1) levels following neferine treatment. Furthermore, neferine increased the phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC), which is an important regulator of fatty acid oxidation. Our result indicates that neferine attenuates adipogenesis and promotes lipid metabolism by activating AMPK-mediated signaling. Therefore, neferine may serve as a therapeutic candidate for obesity treatment.
This content is subject to copyright.
nutrients
Article
Anti-Adipogenic Eect of Neferine in 3T3-L1 Cells
and Primary White Adipocytes
Miey Park 1,2, Jinyoung Han 1and Hae-Jeung Lee 1, 2, *
1Department of Food and Nutrition, College of BioNano Technology, Gachon University,
Gyeonggi-do 13120, Korea; mieyp@naver.com (M.P.); hanalice@gc.gachon.ac.kr (J.H.)
2Institute for Aging and Clinical Nutrition Research, Gachon University, Gyeonggi-do 13120, Korea
*Correspondence: skysea@gachon.ac.kr; Tel.: +82-31-750-5968; Fax: +82-31-724-4411
Received: 25 May 2020; Accepted: 17 June 2020; Published: 22 June 2020


Abstract:
Neferine, an alkaloid component extracted from lotus seed embryos, is known for its
anti-inflammatory, anticancer, and antioxidant properties. However, the anti-adipogenic activity
of neferine has not been thoroughly investigated. In this study, neferine was found to inhibit
lipid accumulation in a dose-dependent manner during the dierentiation of 3T3-L1 cells without
inducing cytotoxicity. Real-time polymerase chain reaction and immunoblot analysis revealed the
downregulation in the expression of peroxisome proliferator activated receptor gamma (PPAR
γ
),
CCAAT/enhancer-binding protein alpha (C/EBP
α
), sterol regulatory element-binding protein-1c
(SREBP-1c), and fatty acid synthase (FAS) and the upregulation in carnitine palmitoyltransferase-1
(CPT-1) and sirtuin 1 (SIRT1) levels following neferine treatment. Furthermore, neferine increased
the phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) and acetyl-CoA
carboxylase (ACC), which is an important regulator of fatty acid oxidation. Our result indicates
that neferine attenuates adipogenesis and promotes lipid metabolism by activating AMPK-mediated
signaling. Therefore, neferine may serve as a therapeutic candidate for obesity treatment.
Keywords: neferine; 3T3-L1 preadipocytes; dierentiation; anti-adipogenic activity
1. Introduction
The prevalence of obesity, one of the biggest health problems among all age groups, is increasing
worldwide [
1
,
2
]. In 2014, about 30% of the world’s population was estimated to be overweight or
obese [
3
]. Obesity is characterized by the excessive accumulation of adipocytes, leading to a rise
in body weight. It is a critical predictor of numerous comorbidities such as cardiovascular disease,
insulin resistance-related diabetes, cancer, and depression [
4
6
]. Common weight loss cures in obese
individuals include diets, physical activity, behavioral therapies, and pharmacological treatments [7].
Anti-obesity drugs involved in weight regulation are known to exert harmful side-eects, including
headache and blood pressure abnormalities [
8
,
9
]. Hence, studies have been directed to investigate the
potential role of plants to treat obesity and related metabolic disorders and to elucidate their beneficial
eects on lipid and glucose metabolism [10].
Neferine is a bisbenzylisoquinoline alkaloid isolated from the seed embryo of Nelumbo nucifera,
commonly known as lotus [
11
]. It has been consumed for a long time in India and China [
12
].
Neferine has been found to exhibit therapeutic properties such as antioxidant, anti-inflammatory,
anticancer, and anti-amnesic eects [
13
15
]. Considering these beneficial properties, neferine
may be exploited for the development of curative products with no side-eects [
11
]. For years,
the embryos of N. nucifera seeds have been consumed in China and India to ameliorate various
diseases and its typical bisbenzylisoquinoline alkaloid is neferine [
11
,
12
,
16
]. Previous studies have
demonstrated its antioxidation, anti-inflammation, and anticancer properties [
13
15
]. Neferine can
Nutrients 2020,12, 1858; doi:10.3390/nu12061858 www.mdpi.com/journal/nutrients
Nutrients 2020,12, 1858 2 of 12
be potentially useful to treat cardiovascular diseases such as arrhythmia, thrombosis, and platelet
aggregation [17,18]
. Further, it is known to exert protective eects against Alzheimer’s disease, amnesia,
and
depression [15,19,20]
, which is suggestive of the plausible application of this phytochemical for
curative purposes [18].
The dierentiation of precursor cells into mature adipocytes is controlled by several markers
associated with adipogenesis [
21
]. The transcription factors, peroxisome proliferator-activated
receptor-
γ
(PPAR
γ
), CCAAT/enhancer-binding proteins (CEBPs), and sterol regulatory element-binding
proteins (SREBPs) are key regulators of adipogenesis [
22
24
], and AMP-activated protein kinase (AMPK)
is a chief regulator of the underlying molecular mechanism [
25
]. Mitochondrial beta-oxidation plays
an important role in energy metabolism and is regulated by carnitine palmitoyltransferase-1 (CPT-1)
and acetyl-CoA carboxylase (ACC) [
26
]. AMPK upregulates the activity of CPT-1 and increases the
transport of free fatty acids for beta-oxidation through the inhibition of the phosphorylation of ACC
and decreases in the concentration of malonyl-CoA [
25
,
27
,
28
]. Further, it suppresses the expression of
ACC and fatty acid synthase (FAS), which are critical transcription factors of lipogenesis, by inhibiting
SREBP-1c activity [
29
]. Until now, very few studies have investigated the anti-adipogenic/lipogenic
eect of neferine in 3T3-L1 preadipocytes. In the present study, we evaluate the eects of neferine on
adipogenesis and lipid metabolism of 3T3-L1 preadipocytes.
2. Materials and Methods
2.1. Materials
Neferine (C38H44N2O6) was purchased from Sigma (St. Louis, MO, USA) and solvated in
dimethyl sulfoxide. 3T3-L1 preadipocytes were acquired from the ATCC (
Manassas, VA, USA
).
Cell growth medium (DMEM), bovine calf serum (BCS), trypsin, fetal bovine serum (FBS),
and insulin were supplied by Thermo Fisher (San Jose, CA, USA), while antibiotic-antimycotic
solution, 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), dorsomorphin (Compound C),
3-isobutyl-1-methylxanthine (IBMX), and dexamethasone (DEX) were procured from Sigma (St. Louis,
MO, USA).
2.2. Cell Culture and Dierentiation of Preadipocytes
3T3-L1 preadipocytes were grown in DMEM supplemented with 10% BCS and
antibiotic-antimycotic solution in a 5% CO
2
incubator at 37
C. Dierentiation of preadipocytes was
induced by substituting the medium with DMEM containing 10% FBS and adipocyte dierentiation
cocktail (MDI; 1 µM DEX, 0.5 mM IBMX, and 10 µg/mL insulin) for 3 days.
C57BL/6 mice (Five-week-old males) were used for the isolation of primary adipocytes and stromal
vascular fraction (SVF), as per the protocol described in the journal [
30
]. In brief, lumps of fat tissues
collected from mice were minced with scissors and incubated with phosphate-buered saline (PBS;
Thermo Fisher, San Jose, CA, USA) supplemented with 1.5 U/mL of collagenase D (Sigma, St. Louis,
MO, USA) at 37
C for 30 min to 1 h. The lysate obtained was filtered with 40-mm cell strainers (SPL
Life Science, Pocheon-si, Gyeonggi-do, Korea) and washed with PBS. After centrifugation (1200 rpm,
10 min) the cells were resuspended in DMEM. Adipocytes were cultured in SVF culture medium
and incubated at 37
C and 5% CO
2
atmosphere. The primary white adipose tissue was subjected
to dierentiation using MDI, as previously described. Neferine was prepared at 20 mM and used
to treat 3T3-L1 preadipocytes and SVF cells at 1, 2.5, 5, and 10
µ
M concentrations. The negative
control was undierentiated cells, while the positive control included dierentiated cells without
neferine treatment. 3T3-L1 preadipocytes were treated with an activator or inhibitor of AMPK. AICAR
(10
µ
M) or dorsomorphin (5
µ
M) was added during dierentiation until the cells were harvested.
All experiments were carried out in triplicates, as per the guidelines for the care and use of laboratory
animals of Gachon University (reference number: GIACUC-R2019004).
Nutrients 2020,12, 1858 3 of 12
2.3. Cell Viability Assay
Preadipocytes 3T3-L1 cells were planted in 96-well plates (1
×
10
4
cells/well) and allowed to adhere
and grow for 24 h. Next, the cells were treated with neferine at 1, 2.5, 5, and 10
µ
M concentrations
and incubated at 37
C for 24, 48, and 72 h under 5% CO
2
atmosphere. Cells were subjected to Cell
Counting Kit-8 (Dojindo Molecular Technologies, Rockville, MD, USA) assay, as recommended by
the manufacturer. The absorbance was measured at 450 nm using a microplate reader (BioTek Inc.,
Winooski, VT, USA).
2.4. Lipids Quantification
Experimental control, or neferine-treated 3T3-L1 cells, were rinsed and fixed using 4%
paraformaldehyde for an hour or longer. Cells were gently washed with 60% isopropanol and
allowed to dry. Each well was stained using a filtered Oil Red O working solution in isopropanol:
distilled water for 1 h at room temperature (20–22
C). Images of stained lipid droplets were obtained
under an inverted microscope (Nikon Eclipse, Shinagawa, Tokyo, Japan). The dye was dissolved in
100% isopropanol, and the absorbance was read at 500 nm wavelength.
2.5. Quantification of Gene Expression
Total RNA was isolated by the TaKaRa
®
method according to the instructions of the manufacturer
(TaKaRa Bio, Kusatsu, Shiga, Japan). In total, 2 µg of isolated RNA was used for quantification using
QuantStudio 3 (Thermo Fisher Scientific, San Jose, CA, USA), and 50 ng RNA was reversely transcribed
to complementary DNA using a PCR (TaKaRa Bio, Kusatsu, Shiga, Japan). RT-PCR was performed
using TB Green (TaKaRa Bio, Kusatsu, Shiga, Japan), and all reactions were carried out in triplicates.
The sequences of forward and reverse primer sets are shown in Table 1.
Table 1. Primer sets for real-time quantitative polymerase chain reaction.
Gene Forward (50–30) Reverse (50–30)
PPARγTTTTCAAGGGTGCCAGTTTC AATCCTTGGCCCTCTGAGAT
C/EBPαTTACAACAGGCCAGGTTTCC GGCTGGCGACATACAGTACA
SREBP-1 TGTTGGCATCCTGCTATCTG AGGGAAAGCTTTGGGGTCTA
β-actin CTGTCCCTGTATGCCTCTG ATGTCACGCACGATTTCC
PPAR
γ
: peroxisome proliferator-activated receptor gamma, C/EBP
α
: CCAAT/enhancer-binding protein alpha,
SREBP-1: Sterol regulatory element-binding transcription factor-1.
2.6. Protein Quantification and Immunoblot Analysis
To analyze the expression of proteins such as PPAR
γ
, C/EBP
α
, SREBP-1, FAS, CPT-1, AMPK,
ACC, and
β
-actin, the cells treated with dierent concentrations of neferine were subjected to western
blot analysis. 3T3-L1 adipocytes and primary white adipocytes were extracted with a protein lysis
buer (iNtRON Biotechnology, Seongnam, Korea) containing protease and phosphatase inhibitors
(Thermo Fisher, San Jose, CA, USA). After incubation for 30 min on ice, total protein from each
sample was quantified using a PRO-MEASURE protein measurement solution (iNtRON Biotechnology,
Seongnam, Korea). Samples with same protein amounts were separated on SDS-PAGE gel, and the
separated bands were electro-transferred to a polyvinylidene fluoride (PVDF) membrane. For 1 h,
the membrane was blocked and immunoblotted with primary antibodies for 2 h, followed by probing
with horseradish peroxidase-labeled secondary antibodies for 1 h. The reactive bands of target proteins
were detected by the Quant LAS 500 system (GE Healthcare Bio-Sciences AB, Björkgatan, Uppsala,
Sweden) using an enhanced chemiluminescence (ECL) reagent (Amersham Pharmacia, Little Chalfont,
Buckinghamshire, UK).
Nutrients 2020,12, 1858 4 of 12
2.7. Statistical Analysis
All experiments were independently performed in triplicates and presented as mean
±
standard
deviation (SD). Data were analyzed on GraphPad Prism 5.03 (GraphPad Software Inc., La Jolla, CA,
USA) using the one-way analysis of variance (ANOVA) followed by Tukey’s post-hoc test. Probability
(p) values less than 0.05 (*) were considered statistically significant.
3. Results
3.1. Eect of Neferine on the Viability of 3T3-L1 Cells
A cell viability assay was used to investigate the cytotoxicity of neferine on 3T3-L1 preadipocytes.
At 20
µ
M concentration, neferine significantly reduced the viability of cells after treatment for 24 and
72 h (Figure 1). Therefore, 10 µM neferine concentration was used in subsequent experiments.
Nutrients 2020, 12, x FOR PEER REVIEW 4 of 12
2.7. Statistical Analysis
All experiments were independently performed in triplicates and presented as mean ± standard
deviation (SD). Data were analyzed on GraphPad Prism 5.03 (GraphPad Software Inc., La Jolla, CA,
USA) using the one-way analysis of variance (ANOVA) followed by Tukey’s post-hoc test.
Probability (p) values less than 0.05 (*) were considered statistically significant.
3. Results
3.1. Effect of Neferine on the Viability of 3T3-L1 Cells
A cell viability assay was used to investigate the cytotoxicity of neferine on 3T3-L1
preadipocytes. At 20 μM concentration, neferine significantly reduced the viability of cells after
treatment for 24 and 72 h (Figure 1). Therefore, 10 μM neferine concentration was used in subsequent
experiments.
Figure 1. Effects of neferine on 3T3-L1 preadipocyte viability. Neferine was used at 2.5, 5, 10, and 20
μM concentrations for 24, 48, and 72 h. * p < 0.05 and *** p < 0.001 vs. Con. All data are presented as
mean ± SD, and experiments were performed for at least three times. The positive control (Con) was
differentiated 3T3-L1 cells treated with adipocyte differentiation cocktail.
3.2. Effect of Neferine on Intracellular Lipid Accumulation in 3T3-L1 Adipocytes
After inducing differentiation for 7 days, 3T3-L1 preadipocytes were stained with Oil Red O dye
to observe intracellular lipid accumulation (Figure 2A). In comparison with control cells, those
treated with neferine (1.25, 2.5, 5, and 10 μM) showed a significant decrease in lipid content in a dose-
dependent manner (Figure 2B). These results demonstrated that neferine was involved in the
inhibition of 3T3-L1 cell differentiation and lipid accumulation.
3.3. Effect of Neferine on the Adipogenesis of 3T3-L1 Cells
To investigate the effects of neferine on adipogenesis, we performed RT-PCR. As shown in
Figure 3, neferine significantly decreased the mRNA expression levels of the key adipogenic
transcription factors, PPARγ, C/EBPα, and SREBP-1c (Figure 3A–C).
The relative protein levels of PPARγ, C/EBPα, and SREBP1c in neferine-treated cells reduced in
a dose-dependent manner (Figure 4A–D). Taken together, these data indicated that neferine
downregulated the expression of the key factors associated with adipogenesis.
3.4. Effect of Neferine on Fatty Acid Oxidation in 3T3-L1 Adipocytes
We differentiated 3T3-L1 cells into mature adipocytes and prepared three identical immunoblots
to study the effect of neferine on fatty acid oxidation. Relative CPT-1 protein expression was
Con 2.5 510 20
0
50
100
150
24 h
48 h
72 h
*** *
***
Neferine (μM)
Relative viability of 3T3-L1 cells
(% of control)
Figure 1.
Eects of neferine on 3T3-L1 preadipocyte viability. Neferine was used at 2.5, 5, 10, and 20
µ
M concentrations for 24, 48, and 72 h. * p<0.05 and *** p<0.001 vs. Con. All data are presented as
mean
±
SD, and experiments were performed for at least three times. The positive control (Con) was
dierentiated 3T3-L1 cells treated with adipocyte dierentiation cocktail.
3.2. Eect of Neferine on Intracellular Lipid Accumulation in 3T3-L1 Adipocytes
After inducing dierentiation for 7 days, 3T3-L1 preadipocytes were stained with Oil Red O
dye to observe intracellular lipid accumulation (Figure 2A). In comparison with control cells, those
treated with neferine (1.25, 2.5, 5, and 10
µ
M) showed a significant decrease in lipid content in a
dose-dependent manner (Figure 2B). These results demonstrated that neferine was involved in the
inhibition of 3T3-L1 cell dierentiation and lipid accumulation.
Nutrients 2020,12, 1858 5 of 12
Nutrients 2020, 12, x FOR PEER REVIEW 5 of 12
significantly upregulated following neferine treatment in a dose-dependent manner (Figure 5A).
Neferine increased the expression of sirtuin 1 (SIRT1) at concentrations up to 5 μM (Figure 5B).
(A)
(B)
Figure 2. Effects of neferine on intracellular lipid accumulation. (A) Lipid droplets were measured by
Oil Red O staining. Cell were treated with neferine at concentrations of 1, 2.5, 5, and 10 μM. Scale bar
indicates 100 μm. (B) Relative lipid content is expressed as percentage. ** p < 0.01 and *** p < 0.001 vs.
Con. All data are presented as mean ± SD, and experiments were performed at least thrice. The
positive control (Con) was differentiated 3T3-L1 cells treated with adipocyte differentiation cocktail.
(A) (B) (C)
Figure 3. Effects of neferine on the expression of adipogenic marker genes. PCR was used to assess
mRNA expression levels of (A) PPARγ, (B) C/EBPα, and (C) SREBP1c. * p < 0.05, ** p < 0.01, and
*** p < 0.001 vs. Con. All data are presented as mean ± SD, and experiments were performed at least
thrice. The positive control (Con) was differentiated 3T3-L1 cells treated with adipocyte
differentiation cocktail.
3.5. Effect of Neferine on the AMPK Pathway of 3T3-L1 Adipocytes
We studied the effect of neferine on the AMPK pathway of 3T3-L1 adipocytes using western blot
analysis. The ratio of p-AMPK/AMPK increased following neferine treatment in a dose-dependent
manner, as indicated in Figure 6A. Phosphorylation of ACC also significantly increased following
treatment with neferine at concentrations up to 5 μM (Figure 6B). Thus, neferine activated the
signaling mediated by AMPK.
Con 1.25 2.5 510
0
50
100
150
** ***
***
Neferine (μM)
Lipid a ccumulat ion (% o f control )
PPAR
γ
mRNA
Con 12.5 510
0.0
0.5
1.0
1.5
***
***
*
Neferine (μM)
Relative mRNA expression
C/EBP
α
mRNA
Con 12.5 510
0.0
0.5
1.0
1.5
***
***
***
***
Neferine (μM)
Relative mRNA expression
SREBP-1c mRNA
Con 12.5 510
0.0
0.5
1.0
1.5
**
***
***
**
Neferine (μM)
Relative mRNA expression
Figure 2.
Eects of neferine on intracellular lipid accumulation. (
A
) Lipid droplets were measured by
Oil Red O staining. Cell were treated with neferine at concentrations of 1, 2.5, 5, and 10
µ
M. Scale bar
indicates 100
µ
m. (
B
) Relative lipid content is expressed as percentage. ** p<0.01 and *** p<0.001
vs. Con. All data are presented as mean
±
SD, and experiments were performed at least thrice. The
positive control (Con) was dierentiated 3T3-L1 cells treated with adipocyte dierentiation cocktail.
3.3. Eect of Neferine on the Adipogenesis of 3T3-L1 Cells
To investigate the eects of neferine on adipogenesis, we performed RT-PCR. As shown in Figure 3,
neferine significantly decreased the mRNA expression levels of the key adipogenic transcription factors,
PPARγ, C/EBPα, and SREBP-1c (Figure 3A–C).
Nutrients 2020, 12, x FOR PEER REVIEW 5 of 12
significantly upregulated following neferine treatment in a dose-dependent manner (Figure 5A).
Neferine increased the expression of sirtuin 1 (SIRT1) at concentrations up to 5 μM (Figure 5B).
(A)
(B)
Figure 2. Effects of neferine on intracellular lipid accumulation. (A) Lipid droplets were measured by
Oil Red O staining. Cell were treated with neferine at concentrations of 1, 2.5, 5, and 10 μM. Scale bar
indicates 100 μm. (B) Relative lipid content is expressed as percentage. ** p < 0.01 and *** p < 0.001 vs.
Con. All data are presented as mean ± SD, and experiments were performed at least thrice. The
positive control (Con) was differentiated 3T3-L1 cells treated with adipocyte differentiation cocktail.
(A) (B) (C)
Figure 3. Effects of neferine on the expression of adipogenic marker genes. PCR was used to assess
mRNA expression levels of (A) PPARγ, (B) C/EBPα, and (C) SREBP1c. * p < 0.05, ** p < 0.01, and
*** p < 0.001 vs. Con. All data are presented as mean ± SD, and experiments were performed at least
thrice. The positive control (Con) was differentiated 3T3-L1 cells treated with adipocyte
differentiation cocktail.
3.5. Effect of Neferine on the AMPK Pathway of 3T3-L1 Adipocytes
We studied the effect of neferine on the AMPK pathway of 3T3-L1 adipocytes using western blot
analysis. The ratio of p-AMPK/AMPK increased following neferine treatment in a dose-dependent
manner, as indicated in Figure 6A. Phosphorylation of ACC also significantly increased following
treatment with neferine at concentrations up to 5 μM (Figure 6B). Thus, neferine activated the
signaling mediated by AMPK.
Con 1.25 2.5 510
0
50
100
150
** ***
***
Neferine (μM)
Lipid a ccumulat ion (% o f control )
PPAR
γ
mRNA
Con 12.5 510
0.0
0.5
1.0
1.5
***
***
*
Neferine (μM)
Relative mRNA expression
C/EBP
α
mRNA
Con 12.5 510
0.0
0.5
1.0
1.5
***
***
***
***
Neferine (μM)
Relative mRNA expression
SREBP-1c mRNA
Con 12.5 510
0.0
0.5
1.0
1.5
**
***
***
**
Neferine (μM)
Relative mRNA expression
Figure 3.
Eects of neferine on the expression of adipogenic marker genes. PCR was used to assess
mRNA expression levels of (
A
) PPAR
γ
, (
B
) C/EBP
α
, and (
C
)SREBP1c. * p<0.05, ** p<0.01,
and
*** p<0.001
vs. Con. All data are presented as mean
±
SD, and experiments were performed
at least thrice. The positive control (Con) was dierentiated 3T3-L1 cells treated with adipocyte
dierentiation cocktail.
The relative protein levels of PPAR
γ
, C/EBP
α
, and SREBP1c in neferine-treated cells reduced
in a dose-dependent manner (Figure 4A–D). Taken together, these data indicated that neferine
downregulated the expression of the key factors associated with adipogenesis.
Nutrients 2020,12, 1858 6 of 12
Nutrients 2020, 12, x FOR PEER REVIEW 6 of 12
3.6. Effect of Neferine on the Adipogenesis of Primary White Adipocytes
Primary white adipocytes were isolated from the subcutaneous and epididymal adipose tissues
of C57BL/6 mice to examine the effect of neferine on adipogenic factors. Primary white adipocytes
treated with neferine showed a consdierbale decrease in the relative protein expression of PPARγ,
C/EBPα, and SREBP-1c in a neferine concentration-dependent manner, consistent with the results
observed with 3T3-L1 adipocytes (Figure 7A–D).
(A) (B)
(C) (D)
Figure 4. Effect of neferine on adipogenesis. Protein expression levels of (A) PPARγ, (B) C/EBPα, and
(C) SREBP-1c were analyzed by immunoblotting. (D) Immunoblot results of adipogenic factors in
3T3-L1 cells. These results are expressed following normalization with β-actin level. * p < 0.05, ** p <
0.01, and *** p < 0.001 vs. Con. All data are presented as mean ± SD, and experiments were performed
at least thrice. The control (Con) was positive control that differentiated 3T3-L1 cells treated with
adipocyte differentiation cocktail.
(A) (B)
Figure 5. Effect of neferine on the expression of the protein involved in fatty acid oxidation. Western
blotting was carried out to analyze the protein expression of (A) CPT-1 and (B) SIRT1 following
normalization to β-actin. * p < 0.05, ** p < 0.01, and *** p < 0.001 vs. Con. All data are presented as mean
PPARγ
Con 12.5 510
0.0
0.5
1.0
1.5
***
***
***
Neferine (μM)
Relative expression
C/E B P α
Con 12.5 510
0.0
0.5
1.0
1.5
***
***
**
Neferine (μM)
Relative expr ession
SREBP-1c
Con 12.5 510
0.0
0.5
1.0
1.5
**
** ** **
Neferine (μM)
Relative expression
Figure 4.
Eect of neferine on adipogenesis. Protein expression levels of (
A
) PPAR
γ
, (
B
) C/EBP
α
,
and (
C
) SREBP-1c were analyzed by immunoblotting. (
D
) Immunoblot results of adipogenic factors
in 3T3-L1 cells. These results are expressed following normalization with
β
-actin level. * p<0.05,
** p<0.01
, and *** p<0.001 vs. Con. All data are presented as mean
±
SD, and experiments were
performed at least thrice. The control (Con) was positive control that dierentiated 3T3-L1 cells treated
with adipocyte dierentiation cocktail.
3.4. Eect of Neferine on Fatty Acid Oxidation in 3T3-L1 Adipocytes
We dierentiated 3T3-L1 cells into mature adipocytes and prepared three identical immunoblots to
study the eect of neferine on fatty acid oxidation. Relative CPT-1 protein expression was significantly
upregulated following neferine treatment in a dose-dependent manner (Figure 5A). Neferine increased
the expression of sirtuin 1 (SIRT1) at concentrations up to 5 µM (Figure 5B).
Nutrients 2020, 12, x FOR PEER REVIEW 6 of 12
3.6. Effect of Neferine on the Adipogenesis of Primary White Adipocytes
Primary white adipocytes were isolated from the subcutaneous and epididymal adipose tissues
of C57BL/6 mice to examine the effect of neferine on adipogenic factors. Primary white adipocytes
treated with neferine showed a consdierbale decrease in the relative protein expression of PPARγ,
C/EBPα, and SREBP-1c in a neferine concentration-dependent manner, consistent with the results
observed with 3T3-L1 adipocytes (Figure 7A–D).
(A) (B)
(C) (D)
Figure 4. Effect of neferine on adipogenesis. Protein expression levels of (A) PPARγ, (B) C/EBPα, and
(C) SREBP-1c were analyzed by immunoblotting. (D) Immunoblot results of adipogenic factors in
3T3-L1 cells. These results are expressed following normalization with β-actin level. * p < 0.05, ** p <
0.01, and *** p < 0.001 vs. Con. All data are presented as mean ± SD, and experiments were performed
at least thrice. The control (Con) was positive control that differentiated 3T3-L1 cells treated with
adipocyte differentiation cocktail.
(A) (B)
Figure 5. Effect of neferine on the expression of the protein involved in fatty acid oxidation. Western
blotting was carried out to analyze the protein expression of (A) CPT-1 and (B) SIRT1 following
normalization to β-actin. * p < 0.05, ** p < 0.01, and *** p < 0.001 vs. Con. All data are presented as mean
± SD, and experiments were performed at least thrice. The positive control (Con) was differentiated
3T3-L1 cells treated with adipocyte differentiation cocktail.
PPARγ
Con 12.5 510
0.0
0.5
1.0
1.5
***
***
***
Neferine (μM)
Relative expression
C/E B P α
Con 12.5 510
0.0
0.5
1.0
1.5
***
***
**
Neferine
(
M
)
Relative expr ession
SREBP-1c
Con 12.5 510
0.0
0.5
1.0
1.5
**
** ** **
Neferine (
μ
M)
Relative expression
Figure 5.
Eect of neferine on the expression of the protein involved in fatty acid oxidation. Western
blotting was carried out to analyze the protein expression of (
A
) CPT-1 and (
B
) SIRT1 following
normalization to
β
-actin. * p<0.05, ** p<0.01, and *** p<0.001 vs. Con. All data are presented as mean
±
SD, and experiments were performed at least thrice. The positive control (Con) was dierentiated
3T3-L1 cells treated with adipocyte dierentiation cocktail.
Nutrients 2020,12, 1858 7 of 12
3.5. Eect of Neferine on the AMPK Pathway of 3T3-L1 Adipocytes
We studied the eect of neferine on the AMPK pathway of 3T3-L1 adipocytes using western blot
analysis. The ratio of p-AMPK/AMPK increased following neferine treatment in a dose-dependent
manner, as indicated in Figure 6A. Phosphorylation of ACC also significantly increased following
treatment with neferine at concentrations up to 5
µ
M (Figure 6B). Thus, neferine activated the signaling
mediated by AMPK.
Nutrients 2020, 12, x FOR PEER REVIEW 7 of 12
(A) (B)
Figure 6. Effects of neferine on AMPK and ACC during the differentiation of 3T3-L1 adipocytes.
Ratios of relative expression levels of (A) p-AMPK/AMPK and (B) p-ACC/ACC are presented. * p <
0.05, ** p < 0.01, and *** p < 0.001 vs. Con. All data are presented as mean ± SD, and experiments were
performed at least thrice. The positive control (Con) was differentiated 3T3-L1 cells treated with
adipocyte differentiation cocktail.
(A) (B)
(C) (D)
Figure 7. Effects of neferine on adipogenesis of primary white adipocytes. Protein expression levels
of (A) PPARγ, (B) C/EBPα, and (C) SREBP-1c were investigated. (D) Detected bands of adipogenic
factors. Results are expressed following normalization of values to β-actin level. * p < 0.05, ** p < 0.01,
and *** p < 0.001 vs. Con. All data are presented as mean ± SD, and experiments were performed at
least thrice. The positive control (Con) was differentiated primary white adipocytes treated with
differentiation cocktail.
PPARγ
Con 12.5 510
0.0
0.5
1.0
1.5
*** *** ***
**
Neferine (μM)
Relative expression
C/E B P α
Con 12.5 510
0.0
0.5
1.0
1.5
*** *** ***
Neferine (μM)
Relati ve expression
SREBP-1c
Con 12.5 510
0.0
0.5
1.0
1.5
*** ***
**
Neferine (μM)
Relative expression
Figure 6.
Eects of neferine on AMPK and ACC during the dierentiation of 3T3-L1 adipocytes. Ratios
of relative expression levels of (
A
)p-AMPK/AMPK and (
B
)p-ACC/ACC are presented.
*p<0.05
,
** p<0.01, and *** p<0.001 vs. Con. All data are presented as mean
±
SD, and experiments were
performed at least thrice. The positive control (Con) was dierentiated 3T3-L1 cells treated with
adipocyte dierentiation cocktail.
3.6. Eect of Neferine on the Adipogenesis of Primary White Adipocytes
Primary white adipocytes were isolated from the subcutaneous and epididymal adipose tissues of
C57BL/6 mice to examine the eect of neferine on adipogenic factors. Primary white adipocytes treated
with neferine showed a consdierbale decrease in the relative protein expression of PPAR
γ
, C/EBP
α
,
and SREBP-1c in a neferine concentration-dependent manner, consistent with the results observed
with 3T3-L1 adipocytes (Figure 7A–D).
3.7. Eect of Neferine on the AMPK Pathway of Primary White Adipocytes
To investigate the eect of neferine on the AMPK pathway of primary white adipocytes,
three identical western blots were prepared. AMPK and ACC expression was dose-dependently
upregulated by neferine treatment (Figure 8A,B). Together, these data demonstrated that neferine
activates the AMPK signaling pathway in primary white adipocytes.
3.8. Eect of Neferine on the AMPK Pathway of 3T3-L1 Adipocytes
To confirm whether AMPK activation was involved in mediating the anti-adipogenic eects
of neferine, 3T3-L1 cells were treated with an AMPK inhibitor dorsomorphin (5
µ
M) and AMPK
activator AICAR (10
µ
M). As described in Figure 9, the protein expression level of AMPK increased
following AICAR treatment but reduced after dorsomorphin treatment. Neferine treatment significantly
upregulated AMPK expression as compared to control treatment. These data demonstrate that the
AMPK pathway plays an important role in mediating the anti-adipogenic eects of neferine in
3T3-L1 adipocytes.
Nutrients 2020,12, 1858 8 of 12
Nutrients 2020, 12, x FOR PEER REVIEW 7 of 12
(A) (B)
Figure 6. Effects of neferine on AMPK and ACC during the differentiation of 3T3-L1 adipocytes.
Ratios of relative expression levels of (A) p-AMPK/AMPK and (B) p-ACC/ACC are presented. * p <
0.05, ** p < 0.01, and *** p < 0.001 vs. Con. All data are presented as mean ± SD, and experiments were
performed at least thrice. The positive control (Con) was differentiated 3T3-L1 cells treated with
adipocyte differentiation cocktail.
(A) (B)
(C) (D)
Figure 7. Effects of neferine on adipogenesis of primary white adipocytes. Protein expression levels
of (A) PPARγ, (B) C/EBPα, and (C) SREBP-1c were investigated. (D) Detected bands of adipogenic
factors. Results are expressed following normalization of values to β-actin level. * p < 0.05, ** p < 0.01,
and *** p < 0.001 vs. Con. All data are presented as mean ± SD, and experiments were performed at
least thrice. The positive control (Con) was differentiated primary white adipocytes treated with
differentiation cocktail.
PPARγ
Con 12.5 510
0.0
0.5
1.0
1.5
*** *** ***
**
Neferine (μM)
Relative expression
C/E B P α
Con 12.5 510
0.0
0.5
1.0
1.5
*** *** ***
Neferine (μM)
Relati ve expression
SREBP-1c
Con 12.5 510
0.0
0.5
1.0
1.5
*** ***
**
Neferine (μM)
Relative expression
Figure 7.
Eects of neferine on adipogenesis of primary white adipocytes. Protein expression levels
of (
A
) PPAR
γ
, (
B
) C/EBP
α
, and (
C
) SREBP-1c were investigated. (
D
) Detected bands of adipogenic
factors. Results are expressed following normalization of values to
β
-actin level. * p<0.05, ** p<0.01,
and *** p<0.001 vs. Con. All data are presented as mean
±
SD, and experiments were performed
at least thrice. The positive control (Con) was dierentiated primary white adipocytes treated with
dierentiation cocktail.
Nutrients 2020
,
12
, x FOR PEER REVIEW 8 of 12
3.7. Effect of Neferine on the AMPK Pathway of Primary White Adipocytes
To investigate the effect of neferine on the AMPK pathway of primary white adipocytes, three
identical western blots were prepared. AMPK and ACC expression was dose-dependently
upregulated by neferine treatment (Figure 8A,B). Together, these data demonstrated that neferine
activates the AMPK signaling pathway in primary white adipocytes.
(A) (B)
Figure 8. Effects of neferine on AMPK and ACC in primary white adipocytes. Western blot analysis
was performed to evaluate the ratio of the relative protein expression levels of (A) p-AMPK/AMPK
and (B) p-ACC/ACC. * p < 0.05 and ** p < 0.01 vs. Con. All data are presented as mean ± SD, and
experiments were performed at least thrice. The positive control (Con) was differentiated primary
white adipocytes treated with differentiation cocktail.
3.8. Effect of Neferine on the AMPK Pathway of 3T3-L1 Adipocytes
To confirm whether AMPK activation was involved in mediating the anti-adipogenic effects of
neferine, 3T3-L1 cells were treated with an AMPK inhibitor dorsomorphin (5 μM) and AMPK
activator AICAR (10 μM). As described in Figure 9, the protein expression level of AMPK increased
following AICAR treatment but reduced after dorsomorphin treatment. Neferine treatment
significantly upregulated AMPK expression as compared to control treatment. These data
demonstrate that the AMPK pathway plays an important role in mediating the anti-adipogenic effects
of neferine in 3T3-L1 adipocytes.
Figure 9. Effect of neferine in 3T3-L1 adipocytes treated with an inhibitor (dorsomorphin) and
activator (5-aminoimidazole-4-carboxamide ribonucleotide (AICAR)) of AMPK. ** p < 0.01 and *** p <
0.001 vs. without Neferine. The ratio of p-AMPK/AMPK was analyzed using immunoblotting. All
data are presented as mean ± SD, and experiments were performed at least thrice.
Figure 8.
Eects of neferine on AMPK and ACC in primary white adipocytes. Western blot analysis
was performed to evaluate the ratio of the relative protein expression levels of (
A
)p-AMPK/AMPK and
(
B
)p-ACC/ACC. * p<0.05 and ** p<0.01 vs. Con. All data are presented as mean
±
SD, and experiments
were performed at least thrice. The positive control (Con) was dierentiated primary white adipocytes
treated with dierentiation cocktail.
Nutrients 2020,12, 1858 9 of 12
Nutrients 2020
,
12
, x FOR PEER REVIEW 8 of 12
3.7. Effect of Neferine on the AMPK Pathway of Primary White Adipocytes
To investigate the effect of neferine on the AMPK pathway of primary white adipocytes, three
identical western blots were prepared. AMPK and ACC expression was dose-dependently
upregulated by neferine treatment (Figure 8A,B). Together, these data demonstrated that neferine
activates the AMPK signaling pathway in primary white adipocytes.
(A) (B)
Figure 8. Effects of neferine on AMPK and ACC in primary white adipocytes. Western blot analysis
was performed to evaluate the ratio of the relative protein expression levels of (A) p-AMPK/AMPK
and (B) p-ACC/ACC. * p < 0.05 and ** p < 0.01 vs. Con. All data are presented as mean ± SD, and
experiments were performed at least thrice. The positive control (Con) was differentiated primary
white adipocytes treated with differentiation cocktail.
3.8. Effect of Neferine on the AMPK Pathway of 3T3-L1 Adipocytes
To confirm whether AMPK activation was involved in mediating the anti-adipogenic effects of
neferine, 3T3-L1 cells were treated with an AMPK inhibitor dorsomorphin (5 μM) and AMPK
activator AICAR (10 μM). As described in Figure 9, the protein expression level of AMPK increased
following AICAR treatment but reduced after dorsomorphin treatment. Neferine treatment
significantly upregulated AMPK expression as compared to control treatment. These data
demonstrate that the AMPK pathway plays an important role in mediating the anti-adipogenic effects
of neferine in 3T3-L1 adipocytes.
Figure 9. Effect of neferine in 3T3-L1 adipocytes treated with an inhibitor (dorsomorphin) and
activator (5-aminoimidazole-4-carboxamide ribonucleotide (AICAR)) of AMPK. ** p < 0.01 and *** p <
0.001 vs. without Neferine. The ratio of p-AMPK/AMPK was analyzed using immunoblotting. All
data are presented as mean ± SD, and experiments were performed at least thrice.
Figure 9.
Eect of neferine in 3T3-L1 adipocytes treated with an inhibitor (dorsomorphin) and activator
(5-aminoimidazole-4-carboxamide ribonucleotide (AICAR)) of AMPK. ** p<0.01 and *** p<0.001 vs.
without Neferine. The ratio of p-AMPK/AMPK was analyzed using immunoblotting. All data are
presented as mean ±SD, and experiments were performed at least thrice.
4. Discussion
Obesity, a growing pandemic, is associated with various metabolic disorders. Numerous
researches have been directed to ameliorate obesity and related complications [
31
,
32
]. As most
anti-obesity drugs exert side-eects [
33
], plant-based phytochemicals are gaining attention. In general,
lipid droplet accumulation and preadipocytes dierentiation into mature adipocytes are regarded as the
hallmark events in obesity [
32
,
34
]. The present study suggests that neferine prominently reduces lipid
accumulation and dierentiation of 3T3-L1 adipocytes and primary white adipocytes by regulating
adipogenic transcriptional factors and AMPK pathway.
The dierentiation of 3T3-L1 preadipocytes is mainly mediated by critical nuclear transcription
factors, PPAR
γ
and C/EBP
α
[
35
]. C/EBP
β
and C/EBP
δ
are stimulated in the process of dierentiation,
thereby inducing the expression of PPARγand C/EBPα[36]. Even without hormones, dierentiation
of preadipocytes is induced by the expression of PPAR
γ
. Thus, PPAR
γ
may be a core factor involved
in adipogenesis [
37
]. C/EBP
α
is expressed along with PPAR
γ
after the end of growth during the
adipogenic stage [
38
] and associated with lipid metabolism [
39
]. PPAR
γ
and C/EBP
α
control the
positive feedback loop to mediate adipogenesis [
40
]. SREBP-1c is also a vital regulator involved in
adipocyte dierentiation and lipid metabolism and participates in lipogenesis [
24
,
41
]. In this study,
neferine downregulated the expression of adipogenic/lipogenic mRNAs and proteins, including PPAR
γ
,
C/EBP
α
, and SREBP-1c, in 3T3-L1 adipocytes and primary white adipocytes. In addition, Oil Red O
staining demonstrated the neferine-mediated inhibition of intracellular lipid accumulation.
AMPK plays a key role in mitochondrial energy homeostasis and regulates lipid and fatty acid
metabolism [
42
,
43
]. AMPK is known to exert beneficial eects in many tissues, including the adipose
tissue, and activation of AMPK is known to suppress adipogenesis by reducing the expression of
adipogenic factors [
44
]. ACC, a major regulator of mitochondrial fatty acid oxidation, is phosphorylated
upon AMPK activation [
45
]. In the present study, the cells treated with neferine showed upregulated
AMPK expression and ACC phosphorylation.
SIRT1 is an NAD-dependent protein that separates acetyl groups from various proteins [
46
]. SIRT1,
Like AMPK, is involved in cellular processes such as energy and lipid metabolism and mitochondrial
biogenesis, and controls adipokines in the adipose tissue [
47
,
48
]. CPT-1 is associated with fatty acid
metabolism and imports the acyl group of long-chain fatty acids to mitochondria to generate acyl
carnitines [
49
,
50
]. Further, the activation of AMPK and SIRT1 induces
β
-oxidation by stimulating
CPT-1 expression [
51
]. Here, we found that the protein expression levels of SIRT1 and CPT-1 were
increased in 3T3-L1 adipocytes following neferine treatment.
Nutrients 2020,12, 1858 10 of 12
As previously stated, neferine upregulated p-AMPK/AMPK and p-ACC/ACC ratios. Moreover,
the AMPK activity of neferine-treated 3T3-L1 cells was promoted by AICAR, an AMPK agonist,
and suppressed by the AMPK antagonist dorsomorphin. We showed that the anti-adipogenic eect of
neferine was related to AMPK-mediated regulation.
Taken together, our study demonstrates that neferine prominently inhibits the accumulation of
intracellular lipid and dierentiation of 3T3-L1 and primary white adipocytes into mature adipocytes at
moderate concentrations through the AMPK signaling pathway. Overall, we verify that neferine may
exhibit potential therapeutic properties for obesity management. Further investigation is warranted to
demonstrate the underlying mechanism and substantiate the safety and value of neferine.
Author Contributions:
Investigation, visualization, data arrangement, and writing, M.P.; Investigation and
original draft preparation, M.P., J.H., and H.-J.L.; conceptualization and supervision, H.-J.L. All authors have read
and agreed to the published version of the manuscript.
Funding:
The “Cooperative Research Program of the Center for Companion Animal Research (Project No.
PJ01398402)”, Rural Development Administration, Republic of Korea, supported this work.
Conflicts of Interest: The authors declare no competing financial interests.
References
1.
Dhurandhar, E.J.; Keith, S.W. The aetiology of obesity beyond eating more and exercising less. Best Pr. Res.
Clin. Gastroenterol. 2014,28, 533–544. [CrossRef] [PubMed]
2.
Donohoe, C.L.; O’Farrell, N.J.; Doyle, S.L.; Reynolds, J.V. The role of obesity in gastrointestinal cancer:
Evidence and opinion. Ther. Adv. Gastroenterol. 2013,7, 38–50. [CrossRef] [PubMed]
3.
Tremmel, M.; Gerdtham, U.-G.; Nilsson, P.M.; Saha, S. Economic Burden of Obesity: A Systematic Literature
Review. Int. J. Environ. Res. Public Health 2017,14, 435. [CrossRef] [PubMed]
4.
Gurmaches, J.S.; Guertin, D.A. Adipocyte lineages: Tracing back the origins of fat. Biochim. Biophys.
Acta Bioenerg. 2013,1842, 340–351. [CrossRef]
5. Haslam, D.W.; James, W.P.T. Obesity. Lancet 2005,367, 1197–1209. [CrossRef]
6.
Wajchenberg, B.L. Subcutaneous and Visceral Adipose Tissue: Their Relation to the Metabolic Syndrome.
Endocr. Rev. 2000,21, 697–738. [CrossRef]
7.
Finelli, C.; Padula, M.C.; Martelli, G.; Tarantino, G. Could the improvement of obesity-related co-morbidities
depend on modified gut hormones secretion? World J. Gastroenterol. 2014,20, 16649–16664. [CrossRef]
8.
Karamadoukis, L.; Shivashankar, G.H.; Ludeman, L.; Williams, A. An unusual complication of treatment
with orlistat. Clin. Nephrol. 2009,71, 430–432. [CrossRef]
9.
Slovacek, L.; Pavl
í
k, V.; Slovackova, B. The eect of sibutramine therapy on occurrence of depression
symptoms among obese patients. Nutr. Metab. Cardiovasc. Dis. 2008,18, 18–43. [CrossRef]
10.
Gamboa-G
ó
mez, C.I.; Rocha-Guzm
á
n, N.E.; Gallegos-Infante, J.A.; Moreno-Jim
é
nez, M.R.;
V
á
zquez-Cabral, B.D.; Gonz
á
lez-Laredo, R.F. Plants with potential use on obesity and its complications.
EXCLI J. 2015,14, 809–831.
11.
Asokan, S.M.; Mariappan, R.; Muthusamy, S.; Velmurugan, B.K.; Shibu, M.A.; Ravichandran, M.;
Shanmugavadivu, M. Pharmacological benefits of neferine—A comprehensive review. Life Sci.
2018
,
199, 60–70. [CrossRef] [PubMed]
12.
Sharma, B.R.; Gautam, L.N.S.; Adhikari, D.; Karki, R. A Comprehensive Review on Chemical Profiling
ofNelumbo Nucifera: Potential for Drug Development. Phytother. Res.
2016
,31, 3–26. [CrossRef] [PubMed]
13.
Kadioglu, O.; Law, B.Y.K.; Mok, S.W.F.; Xu, S.-W.; Eerth, T.; Wong, V.K.W. Mode of Action Analyses of
Neferine, a Bisbenzylisoquinoline Alkaloid of Lotus (Nelumbo nucifera) against Multidrug-Resistant Tumor
Cells. Front. Pharmacol. 2017,8, 308. [CrossRef] [PubMed]
14.
Sivalingam, K.S.; Paramasivan, P.; Weng, C.F.; Viswanadha, V.P. Neferine Potentiates the Antitumor Eect
of Cisplatin in Human Lung Adenocarcinoma Cells Via a Mitochondria-Mediated Apoptosis Pathway.
J. Cell. Biochem. 2017,118, 2865–2876. [CrossRef] [PubMed]
15.
Jung, H.A.; Jin, S.E.; Choi, R.J.; Kim, N.H.; Kim, Y.S.; Ryu, J.H.; Kim, N.-W.; Son, Y.K.; Park, J.J.; Choi, J.S.
Anti-amnesic activity of neferine with antioxidant and anti-inflammatory capacities, as well as inhibition of
ChEs and BACE1. Life Sci. 2010,87, 420–430. [CrossRef]
Nutrients 2020,12, 1858 11 of 12
16.
Paudel, K.R.; Panth, N. Phytochemical Profile and Biological Activity of Nelumbo nucifera. Evid. Based
Complement. Altern. Med. 2015,2015, 1–16. [CrossRef]
17.
Miura, D.S.; Wynn, J.; Torres, V.; Laux, B.; Keefe, D.; Somberg, J.C. Antiarrhythmic ecacy of ethmozine in
patients with ventricular tachycardia as determined by programmed electrical stimulation. Am. Heart J.
1986,111, 661–666. [CrossRef]
18.
Zhou, Y.-J.; Xiang, J.-Z.; Yuan, H.; Liu, H.; Tang, Q.; Hao, H.-Z.; Yin, Z.; Wang, J.; Ming, Z. Neferine exerts its
antithrombotic eect by inhibiting platelet aggregation and promoting dissociation of platelet aggregates.
Thromb. Res. 2013,132, 202–210. [CrossRef]
19.
Jung, H.A.; Karki, S.; Kim, J.H.; Choi, J.S. BACE1 and cholinesterase inhibitory activities of Nelumbo nucifera
embryos. Arch. Pharm. Res. 2014,38, 1178–1187. [CrossRef]
20.
Sugimoto, Y.; Nishimura, K.; Itoh, A.; Tanahashi, T.; Nakajima, H.; Oshiro, H.; Sun, S.; Toda, T.; Yamada, J.
Serotonergic mechanisms are involved in antidepressant-like eects of bisbenzylisoquinolines liensinine and
its analogs isolated from the embryo of Nelumbo nucifera Gaertner seeds in mice. J. Pharm. Pharmacol.
2015
,
67, 1716–1722. [CrossRef]
21.
Drolet, R.; Richard, C.; Sniderman, A.D.; Mailloux, J.; Fortier, M.; Huot, C.; Rh
é
aume, C.; Tchernof, A.;
Rh, C. Hypertrophy and hyperplasia of abdominal adipose tissues in women. Int. J. Obes.
2007
,32, 283–291.
[CrossRef] [PubMed]
22.
Gregoire, F.M.; Smas, C.M.; Sul, H.S. Understanding adipocyte dierentiation. Physiol. Rev.
1998
,78, 783–809.
[CrossRef] [PubMed]
23.
Farmer, S.R. Transcriptional control of adipocyte formation. Cell Metab.
2006
,4, 263–273. [CrossRef]
[PubMed]
24.
Eberl
é
, D.; Hegarty, B.; Bossard, P.; Ferre, P.; Foufelle, F. SREBP transcription factors: Master regulators of
lipid homeostasis. Biochimie 2004,86, 839–848. [CrossRef]
25.
Herzig, S.; Shaw, R.J. AMPK: Guardian of metabolism and mitochondrial homeostasis. Nat. Rev. Mol.
Cell Biol. 2017,19, 121–135. [CrossRef]
26.
Schreurs, M.; Kuipers, F.; Van Der Leij, F. Regulatory enzymes of mitochondrial
β
-oxidation as targets for
treatment of the metabolic syndrome. Obes. Rev. 2010,11, 380–388. [CrossRef]
27.
Saggerson, E.D. Malonyl-CoA, a Key Signaling Molecule in Mammalian Cells. Annu. Rev. Nutr.
2008
,28,
253–272. [CrossRef]
28.
Wolfgang, M.J.; Kurama, T.; Dai, Y.; Suwa, A.; Asaumi, M.; Matsumoto, S.-I.; Cha, S.H.; Shimokawa, T.;
Lane, M.D. The brain-specific carnitine palmitoyltransferase-1c regulates energy homeostasis. Proc. Natl.
Acad. Sci. USA 2006,103, 7282–7287. [CrossRef]
29.
Kim, E.; Lee, J.-H.; Ntambi, J.M.; Hyun, C.-K. Inhibition of stearoyl-CoA desaturase1 activates AMPK and
exhibits beneficial lipid metabolic eects in vitro. Eur. J. Pharmacol. 2011,672, 38–44. [CrossRef]
30.
Hausman, R.B.; Park, H.J.; Hausman, G.J. Isolation and Culture of Preadipocytes from Rodent White Adipose
Tissue. In Advanced Structural Safety Studies; Springer Science and Business Media: Berlin, Germany, 2008;
Volume 456, pp. 201–219.
31.
Lavie, C.J.; De Schutter, A.; Parto, P.; Jahangir, E.; Kokkinos, P.; Ortega, F.B.; Arena, R.; Milani, R.V. Obesity and
Prevalence of Cardiovascular Diseases and Prognosis—The Obesity Paradox Updated.
Prog. Cardiovasc. Dis.
2016,58, 537–547. [CrossRef]
32.
Elagizi, A.; Kachur, S.; Lavie, C.J.; Carbone, S.; Pandey, A.; Ortega, F.B.; Milani, R.V. An Overview and Update
on Obesity and the Obesity Paradox in Cardiovascular Diseases. Prog. Cardiovasc. Dis.
2018
,61, 142–150.
[CrossRef]
33.
Onakpoya, I.; Heneghan, C.J.; Aronson, J.K. Post-marketing withdrawal of anti-obesity medicinal products
because of adverse drug reactions: A systematic review. BMC Med. 2016,14, 191. [CrossRef] [PubMed]
34.
Rayalam, S.; Della-Fera, M.A.; Baile, C.A. Phytochemicals and regulation of the adipocyte life cycle.
J. Nutr. Biochem. 2008,19, 717–726. [CrossRef] [PubMed]
35.
Jang, B.-C. Artesunate inhibits adipogeneis in 3T3-L1 preadipocytes by reducing the expression and/or
phosphorylation levels of C/EBP-α, PPAR-γ, FAS, perilipin A, and STAT-3. Biochem. Biophys. Res. Commun.
2016,474, 220–225. [CrossRef] [PubMed]
36.
Wu, Z.; Rosen, E.D.; Brun, R.; Hauser, S.; Adelmant, G.; Troy, E.A.; McKeon, C.; Darlington, G.J.; Spiegelman, B.
Cross-Regulation of C/EBP
α
and PPAR
γ
Controls the Transcriptional Pathway of Adipogenesis and Insulin
Sensitivity. Mol. Cell 1999,3, 151–158. [CrossRef]
Nutrients 2020,12, 1858 12 of 12
37.
Tang, Q.; Lane, M.D. Adipogenesis: From Stem Cell to Adipocyte. Annu. Rev. Biochem.
2012
,81, 715–736.
[CrossRef]
38.
Ji, S.; Doumit, M.E.; Hill, R.A. Regulation of Adipogenesis and Key Adipogenic Gene Expression by 1,
25-Dihydroxyvitamin D in 3T3-L1 Cells. PLoS ONE 2015,10, e0126142. [CrossRef]
39.
Engin, A. Fat Cell and Fatty Acid Turnover in Obesity; Springer Science and Business Media: Berlin, Germany,
2017; Volume 960, pp. 135–160.
40.
Rosen, E.D.; Hsu, C.-H.; Wang, X.; Sakai, S.; Freeman, M.W.; Gonzalez, F.J.; Spiegelman, B.M. C/EBPalpha
induces adipogenesis through PPARgamma: A unified pathway. Genes Dev. 2002,16, 22–26. [CrossRef]
41.
Yahagi, N.; Shimano, H.; Hasty, A.H.; Amemiya-Kudo, M.; Okazaki, H.; Tamura, Y.; Iizuka, Y.; Shionoiri, F.;
Ohashi, K.; Osuga, J.-I.; et al. A crucial role of sterol regulatory element-binding protein-1 in the regulation of
lipogenic gene expression by polyunsaturated fatty acids. J. Biol. Chem.
1999
,274, 35840–35844. [CrossRef]
42.
Viollet, B.; Andreelli, F. AMP-Activated Protein Kinase and Metabolic Control. Handb. Exp. Pharmacol.
2011
,
203, 303–330. [CrossRef]
43.
O’Neill, H.M.; Holloway, G.P.; Steinberg, G.R. AMPK regulation of fatty acid metabolism and mitochondrial
biogenesis: Implications for obesity. Mol. Cell. Endocrinol. 2013,366, 135–151. [CrossRef] [PubMed]
44.
Habinowski, S.A.; Witters, L.A. The Eects of AICAR on Adipocyte Dierentiation of 3T3-L1 Cells.
Biochem. Biophys. Res. Commun. 2001,286, 852–856. [CrossRef] [PubMed]
45.
Angin, Y.; Beauloye, C.; Horman, S.; Bertrand, L. Regulation of Carbohydrate Metabolism, Lipid Metabolism,
and Protein Metabolism by AMPK. Exp. Suppl. 2016,107, 23–43. [CrossRef] [PubMed]
46.
Michan, S.; Sinclair, D.A. Sirtuins in mammals: Insights into their biological function. Biochem. J.
2007
,404,
1–13. [CrossRef]
47.
Fullerton, M.D.; Steinberg, G.R. SIRT1 takes a backseat to AMPK in the regulation of insulin sensitivity by
resveratrol. Diabetes 2010,59, 551–553. [CrossRef]
48.
Banks, A.S.; Kon, N.; Knight, C.; Matsumoto, M.; Gutierrez-Juarez, R.; Rossetti, L.; Gu, W.; Accili, M. SirT1
Gain of Function Increases Energy Eciency and Prevents Diabetes in Mice. Cell Metab.
2008
,8, 333–341.
[CrossRef]
49.
Ducharme, N.A.; Bickel, P.E. Minireview: Lipid Droplets in Lipogenesis and Lipolysis. Endocrinology
2008
,
149, 942–949. [CrossRef]
50.
McGarry, J.D.; Brown, N.F. The Mitochondrial Carnitine Palmitoyltransferase System—From Concept to
Molecular Analysis. Eur. J. Biochem. 1997,244, 1–14. [CrossRef]
51.
Szkudelski, T.; Szkudelska, K. Eects of AMPK activation on lipolysis in primary rat adipocytes: Studies at
dierent glucose concentrations. Arch. Physiol. Biochem. 2016,123, 1–7. [CrossRef]
©
2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
... Adipogenesis leads to the accumulation of excess fat in adipocytes during the adipocyte differentiation process [73]. Several important factors involving the cascade of adipocyte differentiation have been discovered, including the transcription factors CCAAT/enhancer binding protein alpha (C/EBPα) and peroxisome proliferator-activated receptor gamma (PPARG) [74]. ...
Article
Full-text available
Obesity has become a global issue that affects the emergence of various chronic diseases such as diabetes mellitus, dysplasia, heart disorders, and cancer. In this study, an integration method was developed between the metabolite profile of the active compound of Murraya paniculata and the exploration of the targeting mechanism of adipose tissue using network pharmacology, molecular docking, molecular dynamics simulation, and in vitro tests. Network pharmacology results obtained with the skyline query technique using a block-nested loop (BNL) showed that histone acetyltransferase p300 (EP300), peroxisome proliferator-activated receptor gamma (PPARG), and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PPARGC1A) are potential targets for treating obesity. Enrichment analysis of these three proteins revealed their association with obesity, thermogenesis, energy metabolism, adipocytokines, fat cell differentiation, and glucose homeostasis. Metabolite profiling of M. paniculata leaves revealed sixteen active compounds, ten of which were selected for molecular docking based on drug-likeness and ADME results. Molecular docking results between PPARG and EP300 with the ten active compounds showed a binding affinity value of ≤ -5.0 kcal/mol in all dockings, indicating strong binding. The stability of the protein-ligand complex resulting from docking was examined using molecular dynamics simulations, and we observed the best average root mean square deviation (RMSD) of 0.99 Å for PPARG with trans-3-indoleacrylic acid, which was lower than with the native ligand BRL (2.02 Å). Furthermore, the RMSD was 2.70 Å for EP300 and the native ligand 99E, and the lowest RMSD with the ligand (1R,9S)-5-[(E)-2-(4-Chlorophenyl)vinyl]-11-(5-pyrimidinylcarbonyl)-7,11-diazatricyclo[7.3.1.02,7]trideca-2,4-dien-6-one was 3.33 Å. The in vitro tests to validate the potential of M. paniculata in treating obesity showed that there was a significant decrease in PPARG and EP300 gene expressions in 3T3-L1 mature adipocytes treated with M. paniculata ethanolic extract starting at concentrations 62.5 μg/ml and 15.625 μg/ml, respectively. These results indicate that M. paniculata can potentially treat obesity by disrupting adipocyte maturation and influencing intracellular lipid metabolism.
... ATGL is a key regulatory enzyme involved in lipolysis and participates in triglyceride-specific activity. On the other hand, the attenuation of AMPKα and SIRT1 suppressed β-oxidation of fatty acids by downregulating CPT expression [22], which transports long-chain fatty acids into mitochondria for energy production [23]. Although the CPT expression level was not different significantly among groups, we observed a numerical decrease in CPT expression level in the LR group, which indicated that LR diets inhibited fatty acids β-oxidation. ...
Article
Full-text available
Simple Summary Body metabolism and colonic microbiota collectively controlled lipid metabolism in finishing pigs. Diminishing standardized ileal digestible lysine to net energy ratio in high-wheat diets may regulate lipid metabolism via AMP-activated protein kinase α1/sirtuin 1/peroxisome proliferator-activated receptor-γ coactivator-1α pathway and farnesol X receptor/small heterodimer partner pathway, ultimately increased the marbling score of longissimus dorsi muscle. Abstract The present study aimed to investigate the impacts of dietary standardized ileal digestible lysine to net energy (SID Lys:NE) ratio on lipid metabolism in pigs fed high-wheat diets. Thirty-six crossbred growing barrows (65.20 ± 0.38 kg) were blocked into two treatment groups, fed high-wheat diets with either a high SID Lys:NE ratio (HR) or a low SID Lys:NE ratio (LR). Each treatment group consisted of three replicates, with six pigs per pen in each replicate. The diminishing dietary SID Lys:NE ratio exhibited no adverse impacts on the carcass trait (p > 0.05) but increased the marbling score of the longissimus dorsi muscle (p < 0.05). Meanwhile, LR diets tended to increase the serum triglyceride concentration (p < 0.1). LR diets upregulated fatty acid transport protein 4 and acetyl-coA carboxylase α expression levels and downregulated the expression level of adipose triglyceride lipase (p < 0.05). LR diets improved energy metabolism via decreasing the expression levels of AMP-activated protein kinase (AMPK) α1, sirtuin 1 (SIRT1), and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) (p < 0.05). Additionally, LR diets stimulated hepatic bile acid synthesis via upregulating the expression levels of cytochrome P450 family 7 subfamily A member 1 and cytochrome P450 family 27 subfamily A member 1, and downregulating farnesol X receptor (FXR) and small heterodimer partner (SHP) expression levels (p < 0.05). A lowered SID Lys:NE ratio affected the colonic microbial composition, characterized by increased relative abundances of YRC22, Parabacteroides, Sphaerochaeta, and Bacteroides, alongside a decreased in the proportion of Roseburia, f_Lachnospiraceae_g_Clostridium, Enterococcus, Shuttleworthia, Exiguobacterium, Corynebacterium, Subdoligranulum, Sulfurospirillum, and Marinobacter (p < 0.05). The alterations in microbial composition were accompanied by a decrease in colonic butyrate concentration (p < 0.1). The metabolomic analysis revealed that LR diets affected primary bile acid synthesis and AMPK signaling pathway (p < 0.05). And the mantel analysis indicated that Parabacteroides, Sphaerochaeta, f_Lachnospiraceae_g_Clostridium, Shuttleworthia, and Marinobacter contributed to the alterations in body metabolism. A reduced dietary SID Lys:NE ratio improves energy metabolism, stimulates lipogenesis, and inhibits lipolysis in finishing pigs by regulating the AMPKα/SIRT1/PGC-1α pathway and the FXR/SHP pathway. Parabacteroides and Sphaerochaeta benefited bile acids synthesis, whereas f_Lachnospiraceae_g_Clostridium, Shuttleworthia, and Marinobacter may contribute to the activation of the AMPK signaling pathway. Overall, body metabolism and colonic microbiota collectively controlled the lipid metabolism in finishing pigs.
... The liver is the primary site for maintaining lipid metabolic homeostasis in poultry (Cui et al., 2018). Many researchers have demonstrated the crucial role of Sirtuin 1 (SIRT1) and AMP-activated protein kinase (AMPK) in regulating lipid metabolism (Park et al., 2020, Poornima et al., 2022. Furthermore, the interrelationship between AMPK and SIRT1 is intricate. ...
Article
Full-text available
Dietary supplementation with bioactive substances that can regulate lipid metabolism is an effective approach for reducing excessive fat deposition in chickens. Genistein (GEN) has the potential to alleviate fat deposition; however, the underlying mechanism of GEN's fat-reduction action in chickens remains unclear. Therefore, the present study aimed to explore the underlying mechanism of GEN on the reduction of fat deposition from a novel perspective: intercellular transmission of adipokine between adipocytes and hepatocytes. The findings showed that GEN enhanced the secretion of adiponectin (APN) in chicken adipocytes, and the enhancement effect of GEN was completely blocked when the cells were pretreated with inhibitors targeting estrogen receptor β (ERβ) or proliferator-activated receptor γ (PPARγ) signals, respectively. Furthermore, the results demonstrated that both co-treatment with GEN and APN or treatment with the medium supernatant (Med SUP) derived from chicken adipocytes treated with GEN significantly decreased the content of triglyceride and increased the protein levels of ERβ, Sirtuin 1 (SIRT1) and phosphor-AMP-activated protein kinase (p-AMPK) in chicken hepatocytes compared to the cells treated with GEN or APN alone. Moreover, the increase in the protein levels of SIRT1 and p-AMPK induced by GEN and APN co-treatment or Med SUP treatment were blocked in chicken hepatocytes pretreated with the inhibitor of ERβ signals. Importantly, the up-regulatory effect of GEN and APN co-treatment or Med SUP treatment on the protein level of p-AMPK was also blocked in chicken hepatocytes pretreated with a SIRT1 inhibitor; however, the increase in the protein level of SIRT1 induced by GEN and APN co-treatment or Med SUP treatment was not reversed when the hepatocytes were pretreated with an AMPK inhibitor. In conclusion, the present study demonstrated that GEN enhanced APN secretion by activating the ERβ-Erk-PPARγ signaling pathway in chicken adipocytes. Subsequently, adipocyte-derived APN synergized with GEN to activate the ERβ-mediated SIRT1-AMPK signaling pathway in chicken hepatocytes, ultimately reducing fat deposition. These findings provide substantial evidence from a novel perspective, supporting the potential use of GEN as a dietary supplement to prevent excessive fat deposition in poultry.
... There are plenty of bitter contributors from citrus fruit, lotus seeds and tartary buckwheat exhibiting the effects through the regulation of adipocyte differentiation, especially the conversion of white and brown adipose tissue. In adipocytes, liensinine could inhibit the beige cells to white ones [94]; neferine could alleviate adipogenesis and promote lipid metabolism [95]; quercetin could reduce the ROS levels and remodel white cells into brown-like ones, which prevented obesity [96,97]. Some studies also found that quercetin could decrease total body fat, the fat in the arms, the body mass index (BMI), and waist circumference [98,99].In addition, nomilin showed anti-obesity and anti-hyperglycemic functions via activating G protein-coupled receptor TGR5, which promoted weight loss and insulin sensitivity [100,101]. ...
... This process generates a great amount of adipocytes, which become the predominant cells and thus form adipose tissue [23]. The transcriptional regulation of critical genes including peroxisome-proliferator-activated receptors (PPARs), 5' AMP-activated protein kinase (AMPK), and the induction of lipogenic genes such as sterol regulatory-element-binding protein (SREBP), acetyl-CoA carboxylase (ACC) and fatty acid synthase (FAS) are essential in adipogenesis [24]. An important role could also be played by ATP citrate lyase (ACLY), an enzyme at the crossroads between lipid and carbohydrate metabolism and related to the inflammatory response in macrophages [25]. ...
Article
Full-text available
Recently, microalgae are arousing considerable interest as a source of countless molecules with potential impacts in the nutraceutical and pharmaceutical fields. Haematococcus pluvialis, also named Haematococcus lacustris, is the largest producer of astaxanthin, a carotenoid exhibiting powerful health effects, including anti-lipogenic and anti-diabetic activities. This study was carried out to investigate the properties of two selected strains of H. pluvialis (FBR1 and FBR2) on lipid metabolism, lipolysis and adipogenesis using an in vitro obesity model. FBR1 and FBR2 showed no antiproliferative effect at the lowest concentration in 3T3-L1 adipocytes. Treatment with FBR2 extract reduced lipid deposition, detected via Oil Red O staining and the immunocontent of the adipogenic proteins PPARγ, ACLY and AMPK was revealed using Western blot analysis. Extracts from both strains induced lipolysis in vitro and reduced the secretion of interleukin-6 and tumor necrosis factor-α. Moreover, the FBR1 and FBR2 extracts improved mitochondrial function, reducing the levels of mitochondrial superoxide anion radical and increasing mitochondrial mass compared to untreated adipocytes. These findings suggest that FBR2 extract, more so than FBR1, may represent a promising strategy in overweight and obesity prevention and treatment.
... Liensinine promotes the accumulation of CTSB, CTSD, and CTSL to inhibit lysosomal protease activity, thereby inhibiting mitochondrial autophagy flux, ultimately retaining the molecular characteristics of beige adipocytes, to reduce obesity (Xie et al. 2019). Neferine significantly inhibits intracellular lipid accumulation and inhibits the differentiation of 3T3-L1 and primary white adipocytes into mature adipocytes via AMP-activated protein kinase (AMPK) signaling pathway at moderate concentrations (Park et al. 2020a). ...
Article
Full-text available
Cepharanthine (CEP) is a bisbenzylisoquinoline alkaloid compound found in plants of the Stephania genus, which has biological functions such as regulating autophagy, inhibiting inflammation, oxidative stress, and apoptosis. It is often used for the treatment of inflammatory diseases, viral infections, cancer, and immune disorders and has great clinical translational value. However, there is no detailed research on its specific mechanism and dosage and administration methods, especially clinical research is limited. In recent years, CEP has shown significant effects in the prevention and treatment of COVID-19, suggesting its potential medicinal value waiting to be discovered. In this article, we comprehensively introduce the molecular structure of CEP and its derivatives, describe in detail the pharmacological mechanisms of CEP in various diseases, and discuss how to chemically modify and design CEP to improve its bioavailability. In summary, this work will provide a reference for further research and clinical application of CEP. Graphical Abstract
Article
Objective: Osteoporosis, a skeletal ailment marked by bone metabolism imbalance and disruption of bone microarchitecture, Neferine, a bisbenzylisoquinoline alkaloid with diverse pharmacological activities, has received limited attention in the context of osteoporosis treatment. Methods: We employed a bilateral ovariectomy (OVX) rat model to induce osteoporosis and subsequently administered Neferine treatment for four weeks following successful model establishment. Throughout the modeling and treatment phases, we closely monitored rat body weights. We assessed alterations in bone tissue microstructure through micro-CT, HE staining, and safranin O-fast green staining. Levels of bone formation and resorption markers in serum were evaluated using ELISA assay. Western blot analysis was employed to determine the expression levels of p38MAPK, p-p38MAPK, and bone formation-related genes in bone tissue. We isolated and cultured OVX rat BMSCs (OVX-BMSCs) and induced osteogenic differentiation while simultaneously introducing Neferine and the p38MAPK inhibitor SB203580 for intervention. Results: Neferine treatment effectively curbed the rapid weight gain in OVX rats, ameliorated bone loss, and decreased serum levels of TRAP, CTX-I, PINP, and BALP. Most notably, Neferine promoted the expression of bone formation-related factors in bone tissue of OVX rats, while concurrently activating the p38MAPK signaling pathway. In in vitro experiments, Neferine facilitated the expression of bone formation-related factors in OVX-BMSCs, increased the osteogenic differentiation potential of OVX-BMSCs, and activated the p38MAPK signaling pathway. Nevertheless, SB203580 partially reversed Neferine's promotive effect. Conclusion: Neferine can boost the osteoblastic differentiation of BMSCs and alleviate OVX-induced osteoporosis in rats by activating the p38MAPK signaling pathway.
Article
Full-text available
Six undescribed and six known bisbenzylisoquinoline alkaloids were isolated from the embryo of Nelumbo nucifera seeds. Their structures were fully characterized by a combination of ¹H, ¹³C NMR, 2D NMR, and HRESIMS analyses, as well as ECD computational calculations. The antiadipogenic activity of 11 alkaloids was observed in a dose-responsive manner, leading to the suppression of lipid accumulation in 3T3-L1 cells. Luciferase assay and Western blot analysis showed that the active alkaloids downregulated peroxisome proliferator-activated receptor gamma (PPARγ, a key antiadipogenic receptor) expression in 3T3-L1 cells. Analysis of the structure–activity relationship unveiled that a 1R,1′S configuration in bisbenzylisoquinoline alkaloids led to a notable enhancement in antiadipogenic activity. The resistance level against lipid accumulation highlighted a consistent pattern with the suppressive effect on the PPARγ expression. These activity results indicate that alkaloids from the embryo of N. nucifera seeds have a potential of antiobesity effects through PPARγ downregulation.
Article
Background: Nonalcoholic fatty liver disease (NAFLD), peculiarly nonalcoholic steatohepatitis (NASH), has become the main cause of liver transplantation and liver-related death. However, the US Food and Drug Administration has not approved a specific medication for treating NASH. Neferine (NEF), a natural bisbenzylisoquinoline alkaloid separated from the traditional Chinese medicine Nelumbinis plumula, has a variety of pharmacological properties, especially on metabolic diseases. Nevertheless, the anti-NASH effect and mechanisms of NEF remain unclear. Purpose: This study aimed to investigate the amelioration of NEF on NASH and the potential mechanisms. Study design: HepG2 cells, hepatic stellate cells (HSCs) and high-fat diet (HFD)+carbon tetrachloride (CCl4) induced C57BL/6 mice were used to observe the effect of NEF against NASH and investigate the engaged mechanism. Methods: HSCs and HepG2 cells stimulated by oleic acid (OA) were treated with NEF. C57BL/6 mice were fed with HFD+CCl4 to induce NASH mouse model and treated with or without NEF (5 mg/kg or 10 mg/kg, once daily, i.p) for 4 weeks. Results: NEF significantly attenuated the accumulation of lipid droplets, intracellular triglyceride (TG) levels and hepatocytes apoptosis in OA-exposed HepG2 cells. NEF not only enhanced the AMPK and ACC phosphorylation in OA-stimulated HepG2 cells, but also reduced inflammatory response and fibrosis in lipopolysaccharide (LPS)-stimulated HepG2 and in LX-2, respectively. In HFD+CCl4-induced NASH mice, pathological staining confirmed NEF treatment mitigated hepatic lipid deposition, inflammatory cell infiltration as well as hepatic fibrosis. Furthermore, the liver weight, serum and hepatic TG and total cholesterol (TC) and aspartate aminotransferase (AST) and alanine aminotransferase (ALT) were decreased compared with the model group. HFD+CCl4 also induced the upregulation of specific proteins and genes associated to inflammation (ILs, TNF-α, NLRP3, ASC, CCL2 and CXCL10) and hepatic fibrosis (collagens, α-SMA, TGF-β and TIPM1), which were also suppressed by NEF treatment. Conclusion: Our results demonstrated that NEF played a protective role in hepatic steatosis via the regulation of AMPK pathways, which may serve as an attractive candidate for a potential novel strategy on prevention and treatment of NASH.
Article
Full-text available
Neferine, a bisbenzylisoquinoline alkaloid isolated from the green seed embryos of Lotus (Nelumbo nucifera Gaertn), has been previously shown to have various anti-cancer effects. In the present study, we evaluated the effect of neferine in terms of P-glycoprotein (P-gp) inhibition via in vitro cytotoxicity assays, R123 uptake assays in drug-resistant cancer cells, in silico molecular docking analysis on human P-gp and in silico absorption, distribution, metabolism, and excretion (ADME), quantitative structure activity relationships (QSAR) and toxicity analyses. Lipinski rule of five were mainly considered for the ADME evaluation and the preset descriptors including number of hydrogen bond donor, acceptor, hERG IC50, logp, logD were considered for the QSAR analyses. Neferine revealed higher toxicity toward paclitaxel- and doxorubicin-resistant breast, lung or colon cancer cells, implying collateral sensitivity of these cells toward neferine. Increased R123 uptake was observed in a comparable manner to the control P-gp inhibitor, verapamil. Molecular docking analyses revealed that neferine still interacts with P-gp, even if R123 was pre-bound. Bioinformatical ADME and toxicity analyses revealed that neferine possesses the druggability parameters with no predicted toxicity. In conclusion, neferine may allocate the P-gp drug-binding pocket and prevent R123 binding in agreement with P-gp inhibition experiments, where neferine increased R123 uptake.
Article
Full-text available
Background: The rising prevalence of obesity represents an important public health issue. An assessment of its costs may be useful in providing recommendations for policy and decision makers. This systematic review aimed to assess the economic burden of obesity and to identify, measure and describe the different obesity-related diseases included in the selected studies. Methods: A systematic literature search of studies in the English language was carried out in Medline (PubMed) and Web of Science databases to select cost-of-illness studies calculating the cost of obesity in a study population aged ≥18 years with obesity, as defined by a body mass index of ≥30 kg/m², for the whole selected country. The time frame for the analysis was January 2011 to September 2016. Results: The included twenty three studies reported a substantial economic burden of obesity in both developed and developing countries. There was considerable heterogeneity in methodological approaches, target populations, study time frames, and perspectives. This prevents an informative comparison between most of the studies. Specifically, there was great variety in the included obesity-related diseases and complications among the studies. Conclusions: There is an urgent need for public health measures to prevent obesity in order to save societal resources. Moreover, international consensus is required on standardized methods to calculate the cost of obesity to improve homogeneity and comparability. This aspect should also be considered when including obesity-related diseases.
Article
Full-text available
Background We identified anti-obesity medications withdrawn since 1950 because of adverse drug reactions after regulatory approval, and examined the evidence used to support such withdrawals, investigated the mechanisms of the adverse reactions, and explored the trends over time. Methods We conducted searches in PubMed, the World Health Organization database of drugs, the websites of drug regulatory authorities, and selected full texts, and we hand searched references in retrieved documents. We included anti-obesity medications that were withdrawn between 1950 and December 2015 and assessed the levels of evidence used for making withdrawal decisions using the Oxford Centre for Evidence-Based Medicine criteria. Results We identified 25 anti-obesity medications withdrawn between 1964 and 2009; 23 of these were centrally acting, via monoamine neurotransmitters. Case reports were cited as evidence for withdrawal in 80% of instances. Psychiatric disturbances, cardiotoxicity (mainly attributable to re-uptake inhibitors), and drug abuse or dependence (mainly attributable to neurotransmitter releasing agents) together accounted for 83% of withdrawals. Deaths were reportedly associated with seven products (28%). In almost half of the cases, the withdrawals occurred within 2 years of the first report of an adverse reaction. Conclusions Most of the drugs that affect monoamine neurotransmitters licensed for the treatment of obesity over the past 65 years have been withdrawn because of adverse reactions. The reasons for withdrawal raise concerns about the wisdom of using pharmacological agents that target monoamine neurotransmitters in managing obesity. Greater transparency in the assessment of harms from anti-obesity medications is therefore warranted. Electronic supplementary material The online version of this article (doi:10.1186/s12916-016-0735-y) contains supplementary material, which is available to authorized users.
Article
Obesity increases a number of cardiovascular disease (CVD) risk factors, but patients with many types of CVD may have a better prognosis if classified as overweight or obese, a phenomenon known as the "obesity paradox". This paradoxical benefit of a medically unfavorable phenotype is particularly strong in the overweight and class I obesity, and less pronounced in the more severe or morbidly obese populations (class II-III and greater). Rather than an obesity paradox, it is possible that this phenomenon may represent a "lean paradox", in which individuals classified as normal weight or underweight may have a poorer prognosis with respect to CVD, as a result of a progressive catabolic state and lean mass loss. Cardiorespiratory fitness (CRF) is a fundamental part of this discussion. A greater CRF is associated with lower CVD risk, regardless of body mass index (BMI). Also, the assessment of body composition compartments (i.e., fat mass, fat-free mass, lean mass) and the presence of metabolic derangements may be better indicators of CVD risk than BMI alone. The focus of this review is to summarize the current evidence of the obesity paradox. Moreover, we discuss the utility and limitations of BMI for cardiometabolic risk stratification, in addition to concepts such as "metabolically healthy obesity" (MHO) and the "fat but fit" phenomenon, which describe patients who are diagnosed with obesity using BMI, but without major metabolic derangements and with greater CRF, respectively. Finally, we propose that obese patients presenting with an excess body fat, yet without metabolic abnormalities, should still be viewed as an "at risk" population, and as such should receive advice to change their lifestyle to improve their CRF and to prevent the development of impaired fasting glucose, diabetes mellitus and other CVD risk factors as a form of primary prevention.
Article
This article recapitulates the existing in vitro and in vivo studies focusing on the effects of neferine-an alkaloid derivative of lotus plant, in various disease models and its effects on key signaling molecules. The review also compiles a large number of research studies that demonstrate methods for isolation and extraction, biosynthetic pathway, pharmacological activity and mode of action of neferine and their underlying mechanisms at cellular level. Neferine is a unique bis-benzylisoquinoline alkaloid that possesses a number of therapeutic effects such as anti-cancer, anti-diabetic, anti-aging, anti-microbial, anti-thrombotic, anti-arrhythmic, anti-inflammatory and even anti-HIV. It also enhances the anti-cancer properties of other anti-cancer drugs like cisplatin, adriamycin, taxol, etc. It is also reported to reverse chemo-resistance and enhance sensitivity of the cancer cells towards anti-cancer drugs. The underlying mechanisms for its activities mainly include apoptosis, autophagy and G1 arrest. Neferine protects them against the effect of drugs like cisplatin. The therapeutic properties of neferine is widely diverse, while it shows toxicity to cancer it also shows cyto-protective effects against cardio-vascular diseases, pulmonary disease, and is also effective against Alzheimer's disease and elicits anti-oxidative effect in many cellular systems. This article thus is the first ever attempt to review the therapeutic activities of neferine established in in vitro and in vivo models and to compile all the fragmented data available on the omnipotent activities of neferine.
Article
Gregoire, Francine M., Cynthia M. Smas, and Hei Sook Sul. Understanding Adipocyte Differentiation. Physiol. Rev. 78: 783–809, 1998. — The adipocyte plays a critical role in energy balance. Adipose tissue growth involves an increase in adipocyte size and the formation of new adipocytes from precursor cells. For the last 20 years, the cellular and molecular mechanisms of adipocyte differentiation have been extensively studied using preadipocyte culture systems. Committed preadipocytes undergo growth arrest and subsequent terminal differentiation into adipocytes. This is accompanied by a dramatic increase in expression of adipocyte genes including adipocyte fatty acid binding protein and lipid-metabolizing enzymes. Characterization of regulatory regions of adipose-specific genes has led to the identification of the transcription factors peroxisome proliferator-activated receptor-γ (PPAR-γ) and CCAAT/enhancer binding protein (C/EBP), which play a key role in the complex transcriptional cascade during adipocyte differentiation. Growth and differentiation of preadipocytes is controlled by communication between individual cells or between cells and the extracellular environment. Various hormones and growth factors that affect adipocyte differentiation in a positive or negative manner have been identified. In addition, components involved in cell-cell or cell-matrix interactions such as preadipocyte factor-1 and extracellular matrix proteins are also pivotal in regulating the differentiation process. Identification of these molecules has yielded clues to the biochemical pathways that ultimately result in transcriptional activation via PPAR-γ and C/EBP. Studies on the regulation of the these transcription factors and the mode of action of various agents that influence adipocyte differentiation will reveal the physiological and pathophysiological mechanisms underlying adipose tissue development.