You are on page 1of 9

European Journal of Medicinal Chemistry 143 (2018) 114e122

Contents lists available at ScienceDirect

European Journal of Medicinal Chemistry


journal homepage: http://www.elsevier.com/locate/ejmech

Research paper

Design, syntheses and lipid accumulation inhibitory activities of novel


resveratrol mimics
Chanjuan Li 1, Bao Cheng 1, Sai Fang, Huihao Zhou, Qiong Gu**, Jun Xu*
School of Pharmaceutical Sciences, Sun Yat-Sen University, 132 East Circle Road at University City, Guangzhou 510006, China

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

Article history: Hispidine was initially discovered from Ficus Hispida for cardiovascular protection. In this paper, hispi-
Received 1 September 2017 dine derivatives, which contain a novel resveratrol-like scaffold, have been designed, synthesized, and
Received in revised form assayed as agents against lipid accumulations in 3T3-L1 pre-adipocytes. Six hispidine derivatives have
20 October 2017
the activity of reducing TG in 3T3-L1 adipocytes in dosage-dependent manner. The most active com-
Accepted 5 November 2017
Available online 7 November 2017
pound can reduce the lipid accumulation up to 78.4% at 10 mM qPCR and Western blotting results
demonstrate that the two most active compounds inhibit both lipodenesis and adipogenesis in 3T3-L1
cells through (1) increasing the phosphorylations of AMPK and ACC, promoting SIRT1 expression. These
Keywords:
Lipid accumulation
three proteins are key regulators for lipogenesis and energy metabolism. (2) Decreasing the expressions
AMPK pathway of PPARg, sREBP-1c, and FABP4, which are pivotal regulators for adipogenesis. Overall, this work proves
PPARg pathway that hispidine derivatives diminish the lipid accumulation in 3T3-L1 cell line by downregulating lipo-
SIRT1 genic and adipogenic pathways.
Adipocyte © 2017 Elsevier Masson SAS. All rights reserved.

1. Introduction resveratrol and hispidine-like scaffold, were designed and syn-


thesized in pursuing to find lipid accumulation inhibitory agents.
Hispidine, which is extracted from Ficus Hispida, and resveratrol 3T3-L1 cell line, the in vitro model recapitulating both pre-
are structurally mimics (Fig. 1). Local people make Ficus Hispida adipocyte differentiation (adipogenesis) and lipid accumulation
juice as a substitute for milk. The stem and leaf of the plant are also (lipogenesis) of adipocyte differentiation, was adopted in this study
used as natural therapeutic agents against hypoglycemia, diar- for screening [12]. The differentiation of 3T3-L1 pre-adipocytes is
rhoeal, and cardiovascular risk [1e4]. The mechanisms of actions of mainly regulated by peroxisome proliferator-activated receptor g
hispidine derivatives are not well understood. (PPARg) pathway [13]. Increased PPARg expression can stimulate
Resveratrol, the naturally occurring polyphenol found in red sterol regulatory element-binding protein-1c (sREBP-1c) and fatty
wine and grape skin, was initially reported as a potent anti-tumor acid-binding protein 4 (FABP4) expressions, which induce adipo-
agent [5]. From then on, resveratrol was further reported for cyte differentiation in the early stage [14e16]. The lipogenesis and
many pharmacological activities such as, cardio-protection, anti- energy metabolism are mainly orchestrated by AMPK pathway [17].
aging, anti-oxidation, anti-cancer, anti-inflammation [6e8], anti- Activated AMPK down-regulates ACC activity through phosphory-
obesity and, anti-diabetes [9]. The mechanism of actions of lating ACC, resulting in inhibited lipogenesis and increased energy
resveratrol are reported to be associated with Silent information metabolism in SIRT1 related manner [18].
regulator 1 (SIRT1) activation [10], Adenosine 5-monophosphate- To understand the mechanisms of actions of these hispidine
activated protein kinase (AMPK) phosphorylation and, Acetyl CoA derivatives on 3T3-L1 pre-adipocytes differentiation, experiments
carboxylase (ACC) phosphorylation [11]. were further designed to study how hispidine derivatives diminish
Considering the structures and physiological benefits of both lipid accumulation and, through which pathway.
resveratrol and hispidine, derivatives, which contain a novel
2. Chemistry
* Corresponding author.
** Corresponding author.
E-mail addresses: guqiong@mail.sysu.edu.cn (Q. Gu), junxu@biochemomes.com
The synthetic route for hispidine derivatives are shown in
(J. Xu). Scheme 1 and Scheme 2. Commercially available 4-hydroxy-6-
1
Authors contribute equally to this article. methyl-2H-pyran-2-one was first reacted with K2CO3 and

https://doi.org/10.1016/j.ejmech.2017.11.017
0223-5234/© 2017 Elsevier Masson SAS. All rights reserved.
C. Li et al. / European Journal of Medicinal Chemistry 143 (2018) 114e122 115

3.1.2. General procedure for the preparation of compounds 3-14


Compound 2 (2.40 mmol) and appropriate aldehydes
(2.40 mmol) in the presence of Mg(OCH3)2 (10 mL) was refluxed
under N2 overnight. After the reaction was completed as indicated
by TLC, the mixture was concentrated under reduced pressure. The
residue was purified with silica gel chromatography to obtain
target compounds 3e14.

Fig. 1. Structures of hispidine and resveratrol. 3.1.2.1. (E)-4-methoxy-6-styryl-2H-pyran-2-one (3). Yield 56%; m.p.
147.2e149.3  C; white solid; 1H NMR (400 MHz, CDCl3) d 7.58 (d,

Scheme 1. Syntheses of 1e14. Reagents and conditions: (a) K2CO3, acetone, (CH3)2SO4, r.t., overnight, yield: 85%. (b) aldehydes Mg(OCH3)2, reflux, N2, overnight, yield: 32%e59%.

J ¼ 7.1 Hz, 2H), 7.44 (s, 1H), 7.37 (d, J ¼ 7.6 Hz, 1H), 7.31 (s, 2H), 6.59
(dd, J ¼ 16.0, 4.1 Hz, 1H), 5.95 (s, 1H), 5.50 (s, 1H), 3.82 (s, 3H).

3.1.2.2. (E)-4-methoxy-6-(2-methoxystyryl)-2H-pyran-2-one (4).


Yield: 57%; m.p. 134.0e136.1  C; white solid; 1H NMR (400 MHz,
CDCl3) d 7.79 (d, J ¼ 16.1 Hz, 1H), 7.49 (d, J ¼ 7.0 Hz, 1H), 7.30 (dd,
J ¼ 15.5, 8.3 Hz, 2H), 7.11e6.85 (m, 3H), 6.69 (t, J ¼ 14.5 Hz, 1H), 5.91
(s, 1H), 5.46 (s, 1H), 3.90 (s, 3H), 3.83 (s, 3H). 13C NMR (100 MHz,
Scheme 2. Syntheses of 15e23. Reagents and conditions: (c) excessive liquid NH3, CDCl3) d 171.2, 164.2, 159.4, 158.0, 131.4, 130.5, 128.2, 124.3, 120.7,
EtOH, 120  C, in the sealed tube, 36 h, yield: 19%e51%. 119.4, 111.1, 100.9, 88.6, 55.8, 55.4. LCMS (ESI) m/z: 258.1 [M þ H]þ.

3.1.2.3. (E)-4-methoxy-6-(3-methoxystyryl)-2H-pyran-2-one (5).


(CH3)2SO4 at room temperature in acetone overnight to give com-
Yield: 54%; white solid; 1H NMR (400 MHz, CDCl3) d 7.46e7.33 (m,
pound 2. Compound 2 was then reacted with different aldehydes in
1H), 7.19 (dd, J ¼ 11.1, 8.5 Hz, 1H), 7.03 (d, J ¼ 7.6 Hz, 1H), 6.93 (d,
the presence of Mg(OCH3)2 and was refluxed under N2 to give
J ¼ 10.9 Hz, 1H), 6.82 (dd, J ¼ 8.2, 2.2 Hz, 1H), 6.49 (d, J ¼ 15.9 Hz,
compounds 3e14. Compounds 3e14 were reacted with excessive
1H), 5.87 (d, J ¼ 1.8 Hz, 1H), 5.42 (d, J ¼ 1.8 Hz, 1H), 3.76 (s, 3H), 3.75
liquid NH3 (28%e30%) in ethanol at 120  C in sealed tube to provide
(s, 3H). 13C NMR (100 MHz, CDCl3) d 171.0, 163.9, 159.9, 158.5, 136.6,
target compounds 15e23.
135.7, 129.9, 120.1, 118.9, 115.2, 112.5, 101.4, 88.9, 55.9, 55.3. LCMS
(ESI) m/z: 258.1 [M þ H]þ.
3. Experiments
3.1.2.4. (E)-4-methoxy-6-(4-methoxystyryl)-2H-pyran-2-one (6).
3.1. Chemistry Yield 59%; m.p. 152.5e155.3  C; white solid; 1H NMR (400 MHz,
CDCl3) d 7.37 (d, J ¼ 5.9 Hz, 1H), 7.32 (s, 2H), 6.82 (d, J ¼ 8.2 Hz, 2H),
All chemicals and solvents were purchased from commercial 6.37 (d, J ¼ 15.9 Hz, 1H), 5.81 (s, 1H), 5.39 (s, 1H), 3.75 (s, 3H), 3.74 (s,
sources and were used without further purification. Column 3H). 13C NMR (100 MHz, CDCl3) d 171.2, 164.1, 160.7, 159.1, 135.4,
chromatography (CC) was carried out on silica gel (200e300 mesh, 135.4, 128.9, 128.0, 116.4, 114.4, 114.4, 100.4, 88.3, 55.8, 55.3. LCMS
Qingdao Ocean Chemical Company, China), and thin-layer chro- (ESI) m/z: 258.1 [M þ H]þ.
matography (TLC) analyses were carried out on silica gel GF254
glass plates. 1H NMR and 13C NMR were recorded in CDCl3, Acetone- 3.1.2.5. (E)-6-(2-chlorostyryl)-4-methoxy-2H-pyran-2-one (7).
d6, or Methanol-d4 at room temperature on the Bruker spectrom- Yield 43%; m.p. 115.6e116.8  C; light yellow solid; 1H NMR
eter. Electrospray ionizationemass spectrometry (ESI-MS) spectra (400 MHz, CDCl3) d 7.76 (d, J ¼ 16.0 Hz, 1H), 7.51 (dd, J ¼ 6.1, 3.4 Hz,
were recorded on an Agilent G1946D mass spectrometer. 1H), 7.33 (dd, J ¼ 5.8, 3.5 Hz, 1H), 7.20 (d, J ¼ 2.7 Hz, 1H), 7.18 (d,
J ¼ 2.9 Hz, 1H), 6.51 (d, J ¼ 16.0 Hz, 1H), 5.92 (d, J ¼ 2.0 Hz,1H), 5.44
3.1.1. 4-methoxy-6-methyl-2H-pyran-2-one (2) (d, J ¼ 2.0 Hz, 1H) 3.78e3.54 (m, 3H). 13C NMR (100 MHz, CDCl3)
A mixture of 4-hydroxy-6-methyl-2H-pyran-2-one (2.60 mmol) d 169.8, 162.7, 157.2, 133.5, 132.5, 130.6, 129.1, 126.0, 120.4, 101.0,
and K2CO3 (2.60 mmol) and (CH3)2SO4 (2.60 mmol) in acetone 88.3, 54.9. LCMS (ESI) m/z: 263.1 [M þ H]þ.
(15 mL) was stirred at room temperature overnight. The mixture
was filtered. The filtrate was concentrated in vacuum. The residue 3.1.2.6. (E)-6-(4-chlorostyryl)-4-methoxy-2H-pyran-2-one (8).
was purified with silica gel chromatography using chloroform as Yield 47%; m.p. 139.0e141.2  C; light yellow solid; 1H NMR
eluent to obtain target compound 2 as white solid. Yield: 86%; 1H (400 MHz, CDCl3) d 7.37 (d, J ¼ 8.9 Hz, 1H), 7.34 (s, 2H), 7.27 (d,
NMR (400 MHz, CDCl3) d 5.71 (s, 1H), 5.34 (s, 1H), 3.73 (s, 3H), 2.14 J ¼ 8.4 Hz, 2H), 6.47 (d, J ¼ 16.0 Hz, 1H), 5.87 (d, J ¼ 1.8 Hz, 1H), 5.42
(s, 3H). (d, J ¼ 1.8 Hz, 1H), 3.82e3.63 (m, 3H). 13C NMR (100 MHz, CDCl3)
116 C. Li et al. / European Journal of Medicinal Chemistry 143 (2018) 114e122

d 170.9, 163.7, 158.2, 135.2, 134.3, 133.7, 129.1, 129.1, 128.5, 128.5, 3.1.3.2. (E)-4-hydroxy-6-(2-methoxystyryl)pyridin-2(1H)-one (16).
119.1, 101.6, 89.0, 55.9. LCMS (ESI) m/z: 263.1 [M þ H]þ. Yield: 27%; m.p. 248.0e249.7  C; light yellow solid; 1H NMR
(400 MHz, Methanol-d4) d 7.57 (d, J ¼ 7.3 Hz, 2H), 7.31 (t, J ¼ 7.4 Hz,
3.1.2.7. (E)-6-(4-(dimethylamino)styryl)-4-methoxy-2H-pyran-2-one 1H), 7.01 (d, J ¼ 8.3 Hz, 1H), 6.96 (m, 1H), 6.86 (d, J ¼ 16.6 Hz, 1H),
(9). Yield 32%; m.p. 145.0e146.7  C; yellow solid; 1H NMR (400 MHz, 6.10 (s, 1H), 5.48 (s, 1H), 3.90 (s, 3H). 13C NMR (100 MHz, Methanol-
CDCl3) d 7.37 (d, J ¼ 2.9 Hz, 1H), 7.31 (s, 2H), 6.62 (d, J ¼ 4.4 Hz, 2H), d4) d 165.4, 164.2, 159.2, 157.2, 143.9, 129.6, 127.9, 126.6, 124.0, 120.3,
6.31 (dd, J ¼ 15.8, 4.2 Hz, 1H), 5.77 (s, 1H), 5.36 (s, 1H), 3.75 (d, 120.0, 110.5, 97.7, 54.2. LCMS (ESI) m/z: 244.2 [M þ H]þ.
J ¼ 4.4 Hz, 3H), 3.15e2.79 (m, 6H). 13C NMR (100 MHz, CDCl3) d 171.4,
164.4, 159.9, 151.2, 136.3, 128.9, 128.9, 123.2, 113.6, 112.0, 112.0, 99.2, 3.1.3.3. (E)-4-hydroxy-6-(3-methoxystyryl)pyridin-2(1H)-one (17).
87.6, 55.7, 40.1, 40.1. LCMS (ESI) m/z: 272.1 [M þ H]þ. Yield: 23%; m.p. 168.4e171.1  C; light yellow solid; 1H NMR
(400 MHz, Acetone-d6) d 7.33 (s, 1H), 7.30 (d, J ¼ 7.3 Hz, 1H), 7.21 (d,
3.1.2.8. (E)-6-(2-cyclohexylvinyl)-4-methoxy-2H-pyran-2-one (10). J ¼ 6.7 Hz, 2H), 6.91 (d, J ¼ 7.4 Hz, 1H), 6.86 (d, J ¼ 16.0 Hz, 1H), 6.28
Yield 57%; white solid; 1H NMR (400 MHz, CDCl3) d 6.57 (dd, (s, 1H), 6.08 (s, 1H), 5.08 (s, 1H), 3.84 (s, 3H). 13C NMR (100 MHz,
J ¼ 15.7, 7.0 Hz, 1H), 5.82 (d, J ¼ 15.7 Hz, 1H), 5.70 (d, J ¼ 2.0 Hz, 1H), Acetone-d6) d 163.4, 161.1, 160.0, 159.3, 138.1, 134.4, 130.6, 121.4,
5.35 (d, J ¼ 2.0 Hz, 1H), 3.74 (s, 3H), 2.04 (dd, J ¼ 10.9, 7.0 Hz, 1H), 120.7, 115.7, 113.1, 101.6, 84.4, 55.5. LCMS (ESI) m/z: 244.1 [M þ
1.67 (d, J ¼ 10.8 Hz, 4H), 1.59 (d, J ¼ 12.7 Hz, 1H), 1.22 (m, 3H), 1.07 H]þ.
(m, 3H). 13C NMR (100 MHz, CDCl3) d 171.2, 164.0, 158.9, 145.0, 118.9,
99.5, 88.0, 55.7, 40.7, 32.0, 25.7. LCMS (ESI) m/z: 235.1 [M þ H]þ. 3.1.3.4. (E)-4-hydroxy-6-(4-methoxystyryl)pyridin-2(1H)-one (18).
Yield: 31%; m.p. 144.6e146.5  C; light yellow solid; 1H NMR
3.1.2.9. 4-methoxy-6-((1E,3E)-4-phenylbuta-1,3-dien-1-yl)-2H-py- (400 MHz, Acetone-d6) d 7.58 (d, J ¼ 7.9 Hz, 2H), 7.29 (d, J ¼ 15.9 Hz,
ran-2-one (11). Yield: 43%; light yellow solid; 1H NMR (400 MHz, 1H), 6.96 (d, J ¼ 7.7 Hz, 2H), 6.69 (d, J ¼ 15.9 Hz, 1H), 6.24 (s, 2H),
CDCl3) d 7.38 (d, J ¼ 7.2 Hz, 2H), 7.27 (d, J ¼ 7.7 Hz, 3H), 7.25e7.15 (m, 6.02 (s, 1H), 5.05 (s, 1H), 3.83 (s, 3H). 13C NMR (100 MHz, Acetone-
3H), 6.86e6.71 (m, 2H), 6.08 (d, J ¼ 15.1 Hz, 1H), 5.80 (s, 1H), 5.40 (s, d6) d 163.5, 161.5, 160.2, 159.7, 134.1, 129.7, 129.7, 129.3, 118.8, 115.1,
1H), 3.74 (d, J ¼ 2.7 Hz, 3H). 13C NMR (100 MHz, CDCl3) d 170.0, 115.1, 100.6, 84.0, 55.6. LCMS (ESI) m/z: 244.1 [M þ H]þ.
163.0, 157.6, 137.2, 135.4, 135.2, 127.7, 127.7, 127.6, 126.0, 125.9, 125.9,
121.0, 99.9, 87.7, 54.8. LCMS (ESI) m/z: 255.1 [M þ H]þ. 3.1.3.5. (E)-6-(2-chlorostyryl)-4-hydroxypyridin-2(1H)-one (19).
Yield: 19%; m.p. 213.1e215.4  C; light yellow solid; 1H NMR
3.1.2.10. (E)-4-methoxy-6-(2-(5-methylfuran-2-yl)vinyl)-2H-pyran- (400 MHz, Acetone-d6) d 7.84 (s, 1H), 7.70 (d, J ¼ 5.9 Hz, 1H), 7.48 (d,
2-one (12). Yield 43%; m.p. 148.5e150.4  C; light yellow solid; 1H J ¼ 2.9 Hz, 1H), 7.37 (d, J ¼ 2.6 Hz, 2H), 6.91 (d, J ¼ 7.7 Hz, 1H), 6.33 (s,
NMR (400 MHz, CDCl3) d 7.11 (d, J ¼ 15.6 Hz, 1H), 6.33 (dd, J ¼ 9.2, 2H), 6.16 (s, 1H), 5.11 (d, J ¼ 5.4 Hz, 1H). 13C NMR (100 MHz,
6.2 Hz, 2H), 5.99 (d, J ¼ 2.7 Hz, 1H), 5.80 (s, 1H), 5.38 (d, J ¼ 1.9 Hz, Acetone-d6) d 163.2, 159.9, 158.7, 134.5, 131.0, 130.8, 129.4, 128.3,
1H), 3.73 (d, J ¼ 10.1 Hz, 3H), 2.27 (s, 3H). 13C NMR (100 MHz, CDCl3) 128.0, 124.0, 102.6, 84.7. LCMS (ESI) m/z: 248.1 [M þ H]þ.
d 171.1, 164.0, 158.9, 154.6, 150.2, 122.7, 114.9, 108.8, 100.5, 88.2, 55.8,
13.8. LCMS (ESI) m/z: 233.2 [M þ H]þ. 3.1.3.6. (E)-6-(2-cyclohexylvinyl)-4-hydroxypyridin-2(1H)-one (20).
Yield: 51%; m.p. 173.2e175.3  C; light yellow solid; 1H NMR
3.1.2.11. (E)-6-(2-(furan-2-yl)vinyl)-4-methoxy-2H-pyran-2-one (400 MHz, CDCl3) d 6.60 (dd, J ¼ 15.7, 7.0 Hz, 1H), 5.83 (d, J ¼ 15.7 Hz,
(13). Yield 41%; m.p. 143.0e144.7  C; light yellow solid; 1 HNMR 1H), 5.61 (s, 1H), 5.11 (s, 1H), 4.94 (s, 2H), 2.08 (d, J ¼ 7.3 Hz, 1H), 1.71
(400 MHz, CDCl3) d 7.39 (s, 1H), 7.21 (t, J ¼ 4.7 Hz, 1H), 7.17 (d, (d, J ¼ 10.2 Hz, 4H), 1.23 (t, J ¼ 16.8 Hz, 3H), 1.11 (t, 3H). 13C NMR
J ¼ 4.8 Hz, 1H), 6.50 (d, J ¼ 15.7 Hz, 2H), 6.40 (d, J ¼ 3.1 Hz, 1H), 5.86 (100 MHz, CDCl3) d 164.7, 159.4, 158.7, 144.8, 119.3, 98.4, 84.9, 40.8,
(s, 1H), 5.41 (d, J ¼ 2.9 Hz, 1H), 3.76 (s, 3H). 13C NMR (100 MHz, 32.1, 32.1, 25.9, 25.7, 25.7. LCMS (ESI) m/z: 220.1 [M þ H]þ.
CDCl3) d 170.0, 162.9, 157.4, 150.5, 142.9, 121.5, 115.5, 112.3, 111.3,
100.2, 87.6, 54.9. LCMS (ESI) m/z: 219.1 [M þ H]þ. 3.1.3.7. (E)-4-hydroxy-6-(2-(5-methylfuran-2-yl)vinyl)pyridin-
2(1H)-one (21). Yield: 34%; m.p. 181.7e183.5  C; yellow solid; 1H
3.1.2.12. (E)-4-methoxy-6-(2-(thiophen-2-yl)vinyl)-2H-pyran-2-one NMR (400 MHz, Acetone-d6) d 6.78 (d, J ¼ 15.7 Hz, 1H), 6.30 (d,
(14). Yield 37%; m.p. 166.1e168.2  C; light yellow solid; 1H NMR J ¼ 2.7 Hz, 1H), 6.18 (d, J ¼ 15.7 Hz, 1H), 5.99 (s, 2H), 5.88 (s, 1H), 5.76
(400 MHz, CDCl3) d 7.54 (d, J ¼ 15.6 Hz, 1H), 7.24 (d, J ¼ 4.9 Hz, 1H), (s, 1H), 4.77 (s, 1H), 2.61 (s, 3H). 13C NMR (100 MHz, Acetone-d6)
7.11 (d, J ¼ 3.0 Hz, 1H), 6.96 (t, J ¼ 3.8 Hz, 1H), 6.30 (d, J ¼ 15.6 Hz, d 163.4, 160.2, 159.3, 155.0, 151.4, 121.8, 117.0, 115.0, 109.5, 100.8,
1H), 5.83 (s, 1H), 5.40 (s, 1H), 3.74 (s, 3H). 13C NMR (100 MHz, CDCl3) 84.0, 13.7. LCMS (ESI) m/z: 218.1 [M þ H]þ.
d 171.0, 163.8, 158.3, 140.6, 129.5, 128.6, 128.1, 127.1, 117.7, 100.9, 88.6,
55.9. LCMS (ESI) m/z: 235.2 [M þ H]þ. 3.1.3.8. (E)-6-(2-(furan-2-yl)vinyl)-4-hydroxypyridin-2(1H)-one
(22). Yield: 36%; m.p. 174.5e176.9  C; yellow solid; 1H NMR
3.1.3. General procedure for the preparation of compounds 15-23 (400 MHz, Acetone-d6) d 7.62 (s, 1H), 7.12 (d, J ¼ 15.8 Hz, 1H), 6.72 (t,
Compounds 3e14 (1.2 mmol) were reacted with excessive liquid J ¼ 11.6 Hz, 1H), 6.56 (d, J ¼ 15.7 Hz, 2H), 6.28 (s, 2H), 6.07 (d,
NH3 (28%), stirred in the ethanol at 120  C, in sealed tubes, for 36 h. J ¼ 1.2 Hz, 1H), 5.06 (d, J ¼ 1.7 Hz, 1H). 13C NMR (100 MHz, Acetone-
Each mixture was concentrated in vacuum. The residue was puri- d6) d 163.4, 160.1, 158.9, 152.8, 144.9, 121.7, 118.7, 113.4, 113.1, 101.4,
fied with silica gel chromatography using chloroform/methanol (V: 84.3. LCMS (ESI) m/z: 204.1 [M þ H]þ.
V ¼ 20:1) as eluent to obtain target compounds 15e23.
3.1.3.9. (E)-4-hydroxy-6-(2-(thiophen-2-yl)vinyl)pyridin-2(1H)-one
3.1.3.1. (E)-4-hydroxy-6-styrylpyridin-2(1H)-one (15). Yield: 28%; (23). Yield: 32%; m.p. 201.7e203.8  C; yellow solid; 1H NMR
light yellow solid; 1H NMR (400 MHz, Methanol-d4) d 7.57 (d, (400 MHz, Methanol-d4) d 7.52 (d, J ¼ 15.7 Hz, 1H), 7.44 (d,
J ¼ 7.1 Hz, 2H), 7.42 (s, 1H), 7.38 (dd, J ¼ 9.6, 4.8 Hz, 3H), 7.33 (dd, J ¼ 5.0 Hz, 1H), 7.27 (d, J ¼ 3.2 Hz, 1H), 7.06 (dd, J ¼ 5.0, 3.7 Hz, 1H),
J ¼ 5.2, 1.8 Hz, 1H), 6.79 (d, J ¼ 16.0 Hz, 1H), 6.09 (d, J ¼ 1.9 Hz, 1H), 6.53 (d, J ¼ 15.7 Hz, 1H), 6.04 (d, J ¼ 1.7 Hz, 1H), 5.11 (d, J ¼ 1.9 Hz,
5.14 (d, J ¼ 1.9 Hz, 1H). 13C NMR (100 MHz, Methanol-d4) d 167.8, 1H). 13C NMR (100 MHz, Methanol-d4) d 167.8, 162.5, 159.7, 142.0,
162.6, 160.0, 136.9, 136.0, 130.3, 129.9, 129.9, 128.5, 128.5, 120.5, 130.5, 129.1, 128.9, 128.3, 119.4, 101.6, 83.4. LCMS (ESI) m/z: 220.1
101.9, 83.5. LCMS (ESI) m/z: 214.1 [M þ H]þ. [M þ H]þ.
C. Li et al. / European Journal of Medicinal Chemistry 143 (2018) 114e122 117

3.2. Cell culture and adipocyte differentiation were presented as the relative TG content compared to the positive
control cells.
Cells (HEK293T, 3T3-L1) were cultured in Dulbecco's modified
Eagle's medium (DMEM) supplemented with 10% fetal bovine serum 3.4. Oil red O (ORO) staining
(FBS), 100 U/mL penicillin and 100 mg/mL streptomycin at 37  C in 5%
CO2. Adipocyte differentiation was carried out on 3T3-L1 in accor- Cells were fixed with 10% (v/v) formaldehyde in PBS for 1 h at
dance with Ji-Ming Ye [19]. Briefly, 2 days after full confluence (day room temperate and stained with freshly prepared ORO working
0), the medium was changed to differentiation medium (DMEM solution for 30 min followed by three washes with water. Plates
supplemented with 10% FBS, 100 U/mL penicillin, 100 mg/mL strep- were pictured using a microscope (Nikon, Tokyo, Japan) equipped
tomycin, 2 mg/mL insulin, 100 ng/mL dexamethasone, 0.5 mM 3- with a CCD digital camera (Nikon, Tokyo, Japan). Constant
isobutyl-1-methylxanthine (IBMX) and 10 ng/mL biotin) for 3 days. condenser and light intensity settings were used throughout the
Then (day 3), differentiation medium was switched to post- imaging process.
differentiation medium (DMEM supplemented with 10% FBS, 100
U/mL penicillin, 100 mg/mL streptomycin, 2 mg/mL insulin) for 3.5. Cytotoxicity assay
additional 3 days. At the end (day 6), cells were used for further
analysis. Tested compounds were dissolved in dimethyl sulfoxide HEK293T cells were seeded at the density of 5000 per well into
(DMSO) and supplemented at indicated concentrations throughout 96-well plates to grow overnight. Then the cells were treated with
the course of differentiation. Berberine (BBR) was used as positive various concentrations of compounds for 24 h. 0.5% DMSO as vehicle
control. 0.1% DMSO was used as vehicle control. control. MTT (0.05 mg) was added to each well and incubated for 4 h,
after which 100 mL DMSO was added to each well. The absorbance of
3.3. Triglycerides (TGs) assay wells was recorded at 492 nm after 20 min of shaking. The inhibition
percentage was calculated as (Aexperiment eAblank)/(Acontrol e Ablank).
3T3-L1 cells were collected at the end of differentiation and
washed twice with ice-cold PBS buffer (0.2 M NaCl, 10 mM 3.6. Real-time quantitative PCR
Na2HPO4, 3 mM KCl, 2 mM KH2PO4, pH 7.4). Then the cells were
lysed, the levels of intracellular triglycerides were determined us- Total RNA was isolated using RNAiso plus (TaKaRa, Dalian, China)
ing commercial TG assay kit (Jiancheng Bioengineering Institution, according to the manufacturer's protocol. Total RNA (1 mg) was
Nanjing, China) according to the manufacture's protocol. Results converted to cDNA using ReverTra Ace qPCR RT Master Mix (Toyobo,

Fig. 2. (A) TG assay results for 11 active hispidine derivatives. 3T3-L1 pre-adipocytes were treated with 10 mM of 21 compounds on days 0 and 3, for 3 days during the differ-
entiation. The levels of intracellular TG were determined using commercial TG assay kit. Data are represented as mean ± SD of three independent experiments (*p < 0.05 vs vehicle,
**p < 0.01 vs vehicle). (B) The structures of compounds with best TG-lowing effect.
118 C. Li et al. / European Journal of Medicinal Chemistry 143 (2018) 114e122

Osaka, Japan) according to the manufacturer's protocol. cDNA and quantified using Pierce Rapid Gold BCA Protein Assay Kit (Ther-
SYBR Green was used to amplify specific target genes by Real-time moFisher scientific, Waltham, USA). Protein samples were dena-
PCR Master Mix (Toyobo, Osaka, Japan). The following PCR primer ture in SDS sample buffer and then subjected to SDS-PAGE and
sequences were used: b-actin, sense 50 -TGGAATCCTGTGGCATC- blotted onto polyvinylidene difluoride (Millipore, Darmstadt,
CATGAAA-30 and antisense 50 -TAAAACGCAGCTCAGTAACAGTCC-3’; Germany) membranes. The membranes were then blocked with
AMPK, sense 50 -AAGCCGACCCAATGACATCA-30 and antisense 50 - 5% (w/v) skim milk in TBST (Tris-buffered saline containing 0.1% (v/
CTTCCTTCGTACACGCAAAT-3’; ACC, sense 50 -AGGATTTGCTGTTTCT- v) Tween 20) for 2 h and then incubated with primary antibodies
CAGAGCTT-30 and antisense 50 -CAGGATCTACCCAGGCCACAT-3’; SIR overnight at 4  C. After three times washing in TBST, the mem-
T1, sense 50 -TTGTGAAGCTGTTCGTGGAG-30 and antisense 50 -GCGT branes were then incubated with anti-mouse or anti-rabbit IgG
GGAGGTTTTTCAGTA-3’; PPARg, sense 50 -TGCTGTTATGGGTGAAACT secondary antibodies (conjugated to Horseradish Peroxidase) at
CTG-30 antisense 50 -GAAATCAACTGTGGTAAAGGGC-3’; sREBP-1c, room temperature for 2 h. Immunoreactive signals were detected
sense 50 -CAGCTCAGAGCCGTGGTGA-3'and antisense 50 -TGTGTGCAC using enhanced chemiluminescence reagents with Chem-
TTCGTAGGGTC-3’; FABP4, sense 50 -GTCACCATCCGGTCAGAGAGTAC- iluminescence imager (Tanon 5200, Shanghai, China). Densitom-
3'and antisense50 -TCGTCTGCGGTGATTTCATC-3’. etry analysis was performed using Quantity One Software (Bio-Rad
Laboratories, USA) and quantified to the loading control a-Tublin.
3.7. Western blotting Antibodies used including phospho (Thr172)- and total-AMPKa
(Cell Signaling, Danvers, USA), phospho (Ser79)- and total- ACC
Cell lysates were prepared in lysis buffer containing 1% (v/v) (Cell Signaling, Danvers, USA), SIRT1 (Cell Signaling, Danvers,
NP-40, 0.25% (w/v) deoxycholate, 0.15% (w/v) SDS, protease in- USA), PPARg (Sigma, St. Louis, USA), sREBP-1c (Santa Cruz,
hibitor (Beyotime, Shanghai, China), and phosphatase inhibitor Dallas, USA), FABP4 (Abcam, Cambridge, USA), a-Tublin (Sigma,
cocktail (Beyotime, Shanghai, China). Protein concentrations were St. Louis, USA).

Fig. 3. (A) Hispidine derivatives dose-dependently reduce lipid accumulation in 3T3-L1 cells. (B) Compounds 20 and 21 are superior to BBR and resveratrol in terms of reducing TG
accumulation. Pictures were taken on day-6 with 10 magnification. UN: undifferentiated cells. Vehicle: differentiated cells without compounds. 293T cells received treatment of
compounds for 24 h to test their cytotoxicity. Data are represented as mean ± SD of three independent experiments (*p < 0.05 vs vehicle, **p < 0.01 vs vehicle).
C. Li et al. / European Journal of Medicinal Chemistry 143 (2018) 114e122 119

4. Results there is almost no lipid (red spots) can be observed in the cells
treated with BBR, resveratrol, compound 20 or 21 at 10 mM. There is
4.1. Identifying hispidine derivatives as lipid-lowering agents significantly reduced lipid in the cells treated with those agents at
through 3T3-L1 cell-based assays 1 mM. Previous cytotoxicity tests demonstrated that 1 or 10 mM are
far away below the CC50 of these agents. Specifically, compound 21
The 3T3-L1 cells were treated with differentiation cocktail is not toxic to 3T3-L1 cells at up to 50 mM (Fig. 3B).
together with 21 compounds at the concentration of 10 mM. 11
compounds exhibited the ability of the down-regulating TG accu- 4.3. Hispidine derivatives suppressed adipogenic transcript factors
mulations in 3T3-L1 cells (Fig. 2A). Among the 11 active com- expressions
pounds, 6 compounds significantly exhibited greater activities than
BBR (a well-known positive control agent for 3T3-L1 assay). Com- PPARg and sREBP-1c are the main transcription factors
pound 20 was the best TG accumulation reducer, which reduced involved in adipocyte differentiation. FABP4 is a marker of
78.4% TG accumulation. The chemical structures of the 6 most adipocyte differentiation. To elucidate the mechanism of actions
active compounds are depicted in Fig. 2B. of compounds 20 and 21 in the differentiation of 3T3-L1 pre-
adipocyte, we investigated compounds 20 and 21 effects on the
4.2. Measuring the inhibitory efficacies of lipid accumulation mRNA and protein expressions of adipogenic genes PPARg,
in 3T3-L1 cells sREBP-1c, and downstream transcriptional factor FABP4 through
qPCR and Western blotting analyses. The experiments demon-
To further evaluate the inhibitory efficacies of these compounds strate that when treated with differentiation cocktail, PPARg,
on lipid accumulation, the 3T3-L1 pre-adipocytes were treated sREBP-1c and FABP4 mRNA were significantly up-regulated. After
with differentiation cocktail together with 6 active compounds treated with compound 20 or 21 for 3 days, the mRNA expres-
using different concentrations (10, 3.3, 1.1, and 0.37 mM) for 6 days. sions of PPARg, sREBP-1c and FABP4 were significantly sup-
All 6 active compounds significantly reduced the intracellular lipid pressed comparing with that of vehicle (Fig. 4CeE). Consistently,
accumulation in a dosage-dependent manner (Fig. 3A). Compounds PPARg, sREBP-1c and FABP4 protein expression levels were
20 and 21 are superior to others in terms of reducing TG accumu- increased when treated with differentiation cocktail. After treated
lation. ORO staining was applied for visual inspecting the quantity with compound 21, PPARg, sREBP-1c and FABP4 protein expres-
of lipid, which was stained in red. The agents tested in 3T3-L1 as- sion levels were significantly attenuated (Fig. 4A and B). These
says were BBR, resveratrol, and compounds 20 and 21. In Fig. 3B, results indicate that compounds 20 and 21 can suppress the

Fig. 4. Hispidine derivatives blocked PPARg-FABP4 pathway. 3T3-L1 pre-adipocytes were treated with differentiation cocktail with or without compound 20 or 21 at day 0 for 3
days. Then the cells were harvested for qPCR and Western blotting analyses. Un: undifferentiated cells. Dif. cocktail: differentiation cocktail. (A) PPARg, sREBP-1c, and FABP4 protein
levels. (B) Quantitative PPARg, sREBP-1c, and FABP4 protein levels. (C)e(E) mRNA levels for PPARg (C), sREBP-1c (D), and FABP4 (E). Data are represented as mean ± SD of 3 in-
dependent experiments (*p < 0.05 vs vehicle, **p < 0.01 vs vehicle).
120 C. Li et al. / European Journal of Medicinal Chemistry 143 (2018) 114e122

Fig. 5. Hispidine derivatives suppress lipogenesis by up-regulating AMPK pathway. 3T3-L1 pre-adipocytes were treated with differentiation cocktail with or without compounds 20
or 21 at day 0 for 3 days. Then the cells were harvested for qPCR and Western blotting analysis. Un: undifferentiated cells. Dif. cocktail: differentiation cocktail. (A) Protein expression
levels of the phosphorylated AMPKa, total AMPKa, phosphorylated ACC, total ACC, and SIRT1. (B) Quantitative AMPKa, total AMPKa, phosphorylated ACC, total ACC, and SIRT1
protein levels. (C) The mRNA levels for AMPK, ACC, and SIRT1. Data are represented as mean ± SD of 3 independent experiments (*p < 0.05 vs vehicle, **p < 0.01 vs vehicle).

differentiation of 3T3-L1 pre-adipocytes by inhibiting the mRNA substrate of AMPK) phosphorylation was increased. Specifically,
and protein expressions of adipogenic genes. compounds 20 and 21 increased the AMPKa phosphorylation by
3.45 and 3.10 fold, and that of ACC by 2.98 and 4.12 fold. In short, the
4.4. Hispidine derivatives suppress lipogenesis by up-regulating mRNA expression was not increased, the phosphorylation was
AMPK pathway increased instead.
AMPKa activation has many therapeutic benefits because of its
AMPK activation can inhibit lipogenesis and induce energy enhancing oxidative metabolism and mitochondrial biogenesis.
metabolism. To prove compounds 20 and 21 up-regulating AMPK This process relies on SIRT1 [10], which is required for AMPKa
pathway, qPCR and Western blotting analyses were performed on activation during mitochondrial biogenesis as well as energy
3T3-L1 cells treated with compound 20 or 21. In Fig. 5, the mRNA metabolism [17]. Here, the mRNA and SIRT1 protein expressions
and protein levels of t-AMPK were up-regulated when treated with were also studied. Results indicate that SIRT1 mRNA and protein
differentiation cocktail. Treatment with compound 20 or 21 caused expression levels in differentiated 3T3-L1 cells are significantly
increased AMPKa phosphorylation. Consequently, ACC (the higher than pre-adipocytes. After the 3T3-L1 cells were treated

Table 1
Correlations between molecular properties and activity.

Descriptor code Description R2 Significance

ASA Water accessible surface area of hydrophobic atoms 0.444 0.001


ASA_H Total hydrophobic surface area 0.444 0.001
vdw_area Van der Waals (VDW) surface area 0.380 0.003
a_acc Number of H-bond acceptor atoms 0.379 0.003
vsa_acc VDW acceptor surface area 0.379 0.003
vdw_vol VDW volume 0.361 0.004
vsa_hyd VDW hydrophobic surface area 0.341 0.005
a_hyd Number of hydrophobic atoms 0.219 0.033
E Potential energy 0.204 0.400
logP Log octanol/water partition coefficient 0.128
logS Log solubility in water 0.120
C. Li et al. / European Journal of Medicinal Chemistry 143 (2018) 114e122 121

of lipogenic proteins like ACC. FABP4 is another downstream


factor of PPARg and a biomarker of adipocyte differentiation [21].
Decreased PPARg, sREBP-1c, and FABP4 expressions result in
inhibited adipogenesis in 3T3-L1 pre-adipocyte differentiation
process.
In conclusion, the hispidine derivatives can block both adipo-
genesis and lipogenesis through increasing the phosphorylation of
AMPK and ACC, up-regulating SIRT1, down-regulating PPARg,
sREBP-1c, and FABP4. Hispidine derivatives can be promising leads
for the therapeutic solutions against obesity and type 2 diabetes.

Author's contributions

J.X. and Q.G. designed research. CJ.L., B.C., and S.F. performed
research, all authors analyzed data, J.X. CJ.L., and B.C. wrote the
paper, all authors approved the final version.

Competing interests

The authors declare no competing financial interest.

Acknowledgements

Fig. 6. Pathway of compounds 20 and 21 blocking adipogenesis and lipogenesis. This work was funded by the Science and Technology Planning
Project of Guangdong Province (No. 2016A020217002), the Medical
Scientific Research Foundation of Guangdong Province (A2016133),
with compounds 20 or 21, SIRT1 mRNA and protein expression the National Natural Science Foundation of China (No. 81603030),
levels are even higher. This means that SIRT1 involves in AMPK the National Natural Science Foundation of China (No. 81473138),
activation during pre-adipocytes differentiation (Fig. 5A, B). the National Natural Science Foundation of China (No. 81603026),
and the 111 Project (No. B16047).

5. Discussion Appendix A. Supplementary data

In this study, hispidine derivatives, which contain a novel Supplementary data related to this article can be found at
resveratrol-like scaffold, have been designed, synthesized, and https://doi.org/10.1016/j.ejmech.2017.11.017.
assayed as agents against lipid accumulations in 3T3-L1 pre-
adipocytes. The structure-activity relationship can be drawn References
through 3D and 2D molecular property descriptors calculation using
Molecular Operating Environment (MOE). According to R2 and sta- [1] R. Ghosh, K. Sharatchandra, S. Rita, I. Thokchom, Hypoglycemic activity of
tistical significance, 8 molecular property descriptors (descriptors Ficus hispida (bark) in normal and diabetic albino rats, Indian J. Pharmacol. 36
(2004) 222.
concerning surface area, conformation dependent charge, pharma-
[2] S.C. Mandal, C.K.A. Kumar, Studies on anti-diarrhoeal activity of Ficus hispida.
cophore feature, and physical properties) are found to be signifi- Leaf extract in rats, Fitoterapia 73 (2002) 663e667.
cantly related to their TG inhibitory activities. Potential energy, logP, [3] S.C. Mandal, B. Saraswathi, C.K.A. Kumar, S.M. Lakshmi, B.C. Maiti, Protective
effect of leaf extract of Ficus hispida Linn. against paracetamol-induced hep-
and logS are also considered related (Table 1). Increased hydropho-
atotoxicity in rats, Phytotherapy Res. 14 (2000) 457e459.
bic surface, H-bond acceptor, potential energy, logP, and decreased [4] S. Shahreen, J. Banik, A. Hafiz, S. Rahman, A.T. Zaman, M. Abu Shoyeb,
logS reduces TG inhibitory activity (Figs. S44AeS44K, right), indi- M.H. Chowdhury, M. Rahmatullah, Antihyperglycemic activities of leaves of
cating that these hispidine derivatives should keep a balance in three edible fruit plants (averrhoa carambola, Ficus hispida and syzygium
samarangense) of Bangladesh, Afr. J. Tradit. Complem 9 (2012) 287e291.
hydrophilicity and lipophilicity (logP ~2). Compared to hydroxyl [5] M. Jang, J.M. Pezzuto, Cancer chemopreventive activity of resveratrol, Drugs
group, hispidine derivatives with a methoxyl group at C-4 position under Exp. Clin. Res. 25 (1999) 65e77.
can significantly increase hydrophobic surface, H-bond acceptor, [6] S. Dore, Unique properties of polyphenol stilbenes in the brain: more than
direct antioxidant actions; Gene/protein regulatory activity, Neurosignals 14
VDW acceptor surface, and logP, and decrease logS (Fig. S44, left). (2005) 61e70.
Consequently, this reduced TG inhibitory activity. [7] C.A. de la Lastra, I. Villegas, Resveratrol as an anti-inflammatory and anti-aging
The differentiation of pre-adipocytes into mature adipocytes agent: mechanisms and clinical implications, Mol. Nutr. food Res. 49 (2005)
405e430.
consists of adipogenesis and lipogenesis [12,20], and compounds [8] Z.Y. Zhang, S.F. Lowry, L. Guarente, B. Haimovich, Roles of SIRT1 in the acute
20 and 21 can block both processes. Based on the results, we can and restorative phases following induction of inflammation, J. Biol. Chem. 285
construct the pathway of compounds 20 and 21 blocking adi- (2010) 41391e41401.
[9] J.A. Baur, K.J. Pearson, N.L. Price, H.A. Jamieson, C. Lerin, A. Kalra, V.V. Prabhu,
pogenesis and lipogenesis. As shown in Fig. 6, we conclude (1) J.S. Allard, G. Lopez-Lluch, K. Lewis, P.J. Pistell, S. Poosala, K.G. Becker, O. Boss,
the hispidine derivatives (compounds 20 and 21) increase AMPK D. Gwinn, M.Y. Wang, S. Ramaswamy, K.W. Fishbein, R.G. Spencer,
phosphorylation and SIRT1 expression, down-regulate ACC ac- E.G. Lakatta, D. Le Couteur, R.J. Shaw, P. Navas, P. Puigserver, D.K. Ingram, R. de
Cabo, D.A. Sinclair, Resveratrol improves health and survival of mice on a
tivity through phosphorylating ACC. Consequently, the com-
high-calorie diet, Nature 444 (2006) 337e342.
pounds inhibit lipogenesis and increase energy metabolism in [10] N.L. Price, A.P. Gomes, A.J.Y. Ling, F.V. Duarte, A. Martin-Montalvo, B.J. North,
3T3-L1 pre-adipocytes differentiation process. (2) The hispidine B. Agarwal, L. Ye, G. Ramadori, J.S. Teodoro, B.P. Hubbard, A.T. Varela,
derivatives inhibit PPARg, sREBP-1c, and FABP4 expressions. The J.G. Davis, B. Varamini, A. Hafner, R. Moaddel, A.P. Rolo, R. Coppari,
C.M. Palmeira, R. de Cabo, J.A. Baur, D.A. Sinclair, SIRT1 is required for AMPK
adipogenesis is mainly regulated by PPARg pathway [13]. sREBP- activation and the beneficial effects of resveratrol on mitochondrial function,
1c is a downstream factor of PPARg, and regulates the expression Cell metab. 15 (2012) 675e690.
122 C. Li et al. / European Journal of Medicinal Chemistry 143 (2018) 114e122

[11] D.G. Hardie, D.A. Pan, Regulation of fatty acid synthesis and oxidation by the [17] C. Canto, Z. Gerhart-Hines, J.N. Feige, M. Lagouge, L. Noriega, J.C. Milne,
AMP-activated protein kinase, Biochem. Soc. Trans. 30 (2002) 1064e1070. P.J. Elliott, P. Puigserver, J. Auwerx, AMPK regulates energy expenditure by
[12] Q.Q. Tang, M.D. Lane, Adipogenesis: from stem cell to adipocyte, Annu. Rev. modulating NAD(þ) metabolism and SIRT1 activity, Nature 458 (2009)
Biochem. 81 (81) (2012) 715e736. 1056eU1140.
[13] Y.H. Lee, S.H. Kim, E.S. Kang, B.W. Lee, B.S. Cha, J.W. Kim, H.C. Lee, Tran- [18] S.H. Park, S.R. Gammon, J.D. Knippers, S.R. Paulsen, D.S. Rubink, W.W. Winder,
scription factor snail is a novel regulator of adipocyte differentiation via Phosphorylation-activity relationships of AMPK and acetyl-CoA carboxylase in
inhibiting the expression of peroxisome proliferator-activated receptor muscle, J. Appl. Physiology 92 (2002) 2475e2482.
gamma, Diabetes 62 (2013). A533eA533. [19] X.Y. Zeng, X. Zhou, J. Xu, S.M.H. Chan, C.L. Xue, J.C. Molero, J.M. Ye, Screening
[14] E.D. Rosen, O.A. MacDougald, Adipocyte differentiation from the inside out, for the efficacy on lipid accumulation in 3T3-L1 cells is an effective tool for the
Nature reviews, Mol. Cell Biol. 7 (2006) 885. identification of new anti-diabetic compounds, Biochem. Pharmacol. 84
[15] F.M. Gregoire, C.M. Smas, H.S. Sul, Understanding adipocyte differentiation, (2012) 830e837.
Physiol. Rev. 78 (1998) 783e809. [20] H.S. Camp, D.L. Ren, T. Leff, Adipogenesis and fat-cell function in obesity and
[16] S.W. Qian, X. Li, Y.Y. Zhang, H.Y. Huang, Y.A. Liu, X. Sun, Q.Q. Tang, Charac- diabetes, Trends Mol. Med. 8 (2002) 442e447.
terization of adipocyte differentiation from human mesenchymal stem cells in [21] E.D. Rosen, O.A. MacDougald, Adipocyte differentiation from the inside out,
bone marrow, Bmc Dev. Biol. 10 (2010). Nat. Rev. Mol. Cell Bio 7 (2006) 885e896.

You might also like