Sunteți pe pagina 1din 6

Fitoterapia 83 (2012) 104–109

Contents lists available at SciVerse ScienceDirect

Fitoterapia
journal homepage: www.elsevier.com/locate/fitote

Bioactive phenolics from the fruits of Livistona chinensis


Xiaobin Zeng a, Yihai Wang a, Qian Qiu b, Chenguang Jiang c, Yuntiao Jing c,
Guofu Qiu a, Xiangjiu He a,⁎
a
School of Pharmaceutical Sciences, Wuhan University, and Key Laboratory of combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Educa-
tion, Wuhan 430071, China
b
Department of Pharmacy Office, Renmin Hospital of Wuhan University, Wuhan 430060, China
c
Guangdong Dexin Pharmaceutical Company, Jiangmen 529100, China

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

Article history: This study investigated the antioxidant and cytotoxic activity of the phenolics isolated from the
Received 6 July 2011 fruits of Livistona chinensis. Four new compounds, 1-{ω-isoferul[6- (4-hydroxybutyl)pentadecanoic
Received in revised form 19 September 2011 acid]}-glycerol (1), E-[6′-(5″-hydroxypentyl)tricosyl]-4-hydroxy-3-methoxycinnamate (2), 2-(3′-
Accepted 29 September 2011 hydroxy-5′-methoxyphenyl)-3-hydroxylmethyl-7-methoxy-2,3-dihydrobenzofuran-5- carboxylic
Available online 8 October 2011
acid (3), 7-hydroxy-5,4′-dimethoxy-2-arylbenzofuran (4), together with eleven known phenolics
(5–15), were isolated and identified. Among these compounds, 1–4, 5-O-caffeoylshikimic acid
Keywords: (5), caffeic acid (7), and 3-O-caffeoylshikimic acid (8) showed potent antioxidant activity. 1–5,
Livistona chinensis and 8 showed potent antiproliferative activities with IC50 values among 5–150 μM against HepG2
1-(ω-isoferuloylalkanoyl)-glycerol
human liver cancer, HL-60 human myeloid leukemia, K562 human myeloid leukemia, and CNE-1
Long-chain trans-ferulate
human nasopharyngeal carcinoma cell lines. On the basis of these findings, it could be proposed
Benzofuran
Antioxidant activity that the fruits of L. chinensis may serve as attractive mines of powerful anticancer and antioxidant
Antiproliferative activity agents for various purposes.
© 2011 Elsevier B.V. All rights reserved.

1. Introduction antiproliferative effects against human myeloid leukemia


cells (L1210, P388, HL-60), gastric cancer cells (SGC7901), cer-
As a symbol of tropical landscapes, the genus Livistona is vical cancer cells (HeLa), human liver cancer cells (HepG2,
widely distributed in several ecosystems, including upland Hele7404), melanoma cells (B16), colon cancer cells (HT-29),
hardwoods, flatwoods, and tropical hammocks over the trop- and bladder cancer cells (T24) as well [6–11]. Earlier studies
ical zone. Its central bud has been served as a delicious food on the chemical composition of L. chinensis fruits reported the
eaten raw or cooked for swamp cabbage or heart of palm presence of some flavonoids, steroids, phenolics, fatty acids,
salad. There are three species of this genus growing in amino acids, and vitamins [12–17].
South China [1], and their fruits have traditionally been In our continuing search for natural bioactive agents from
used for analgesic and hemostatic purposes, and to treat na- high plants, the fruits of L. chinensis were fractionated and
sopharyngeal carcinoma, choriocarcinoma, esophageal can- purified. Fifteen phenolics, including four new compounds,
cer, and leukemia [2–3]. It has been reported that the fruits 1-{ω-isoferul[6-(4-hydroxybutyl)pentadecanoic acid]}-glycerol
of Livistona chinensis showed anticancer, antibacterial and (1), E - [6′- (5″ - hydroxypentyl)tricosyl] - 4 - hydroxy - 3 -
anti-HIV-1 activities [4–5]. The fruits showed significantly methoxycinnamate (2), 2-(3′-hydroxy-5′-methoxyphenyl)-3 -
hydroxylmethyl - 7 - methoxy - 2,3 - dihydrobenzofuran - 5 -
carboxylic acid (3), 7-hydroxy-5,4′-dimethoxy-2-arylbenzofuran
⁎ Corresponding author at: School of Pharmaceutical Sciences, Wuhan
University, Wuhan 430071 China. Tel.: + 86 27 6875 9923; fax: + 86 27
(4), and eleven known (5–15), were isolated and identified
6875 9850. from the chloroform soluble fraction of a 70% EtOH extract.
E-mail address: hexiangjiu@whu.edu.cn (X. He). Antiproliferative activities against four human tumor cell lines

0367-326X/$ – see front matter © 2011 Elsevier B.V. All rights reserved.
doi:10.1016/j.fitote.2011.09.020
X. Zeng et al. / Fitoterapia 83 (2012) 104–109 105

(HL-60, K562, HepG2, and CNE-1), and antioxidant assays for combined with preparative HPLC, yielding compounds 5
scavenging ability of DPPH and superoxide anion free radicals (12 mg), 10 (20 mg), 11 (25 mg), and 12 (15 mg), respec-
(O2−), were evaluated. tively. The CHCl3–MeOH (5:1) elution was subjected to an
octadecylsilanized silica gel (ODS) column, followed by a
2. Experimental preparative HPLC with 25% methanol (containing 0.1% CF3-
COOH, pH 3.0), yielding compound 3 (10 mg). The CHCl3–
2.1. General MeOH (3:1) elution was subjected to an ODS column,
followed by a preparative Rp-HPLC with 22% methanol
Optical rotations were measured using a JASCO P-1030 (containing 0.1% CF3COOH, pH 3.0), yielding compound
automatic digital polarimeter (Tokyo, Japan). NMR spectra 13 (10 mg). The structures of compounds 1–15 are shown
were recorded on a Bruker DPX-400 spectrometer using stan- in Fig. 1.
dard Bruker pulse programs. Chemical shifts were shown as 1-{ω-isoferul[6-(4-hydroxybutyl)pentadecanoic acid]}-glycerol
δ-values with reference to tetramethylsilane (TMS) as an inter- (1) was an amorphous powder; [α]25D +6.1 (c 0.20, CHCl3);
13
nal standard. GC–MS analysis was done using a Shimadzu GC- C NMR (CDCl3, 100 MHz) and 1H NMR (CDCl3, 400 MHz)
14A unit coupled with a GCMS-QP 2000 instrument (Tokyo, data, see Table 1; HRESIMS (positive ion mode) m/z [M+H]+
Japan). ESI-MS data were obtained on an Agilent 1200 579.3939 (calcd. 579.3931).
HPLC/6410B TripleQuad mass spectrometer (Santa Clara, CA), E - [6′ - (5″ - hydroxypentyl)tricosyl] - 4 - hydroxy - 3 -
and HR-ESIMS were measured on a Bruker APEX II mass spec- methoxycinnamate (2) was an amorphous powder; [α]25D-2.1
trometer (Bremen, Germany). Sephadex LH-20 (Pharmacia, (c 0.20, CHCl3); 13C NMR (CDCl3, 100 MHz) and 1H NMR
Sweden), silica gel (Qingdao Ocean Chemical Co., Ltd, Qingdao, (CDCl3, 400 MHz) data, see Table 1; HRESIMS (positive ion
China), and ODS (40–63 μm, Merck, Darmstadt, Germany) mode) m/z [M+ H]+ 603.4996 (calcd. 603.4991).
were used for column chromatography. TLC was carried out 2-(3′-hydroxy-5′-methoxyphenyl)-3-hydroxylmethyl-7-
on preparative silica gel 60 F254 and RP-18 F254 plates (Merck, methoxy-2,3-dihydrobenzofuran-5-carboxylic acid (3) was an
Darmstadt, Germany), and spots were visualized by spraying amorphous powder; [α] 25D + 8.2 (c 0.20, MeOH); 13C NMR
the plates with 15% H2SO4, and heating them at 105 °C. Prepar- (DMSO-d6, 100 MHz) and 1H NMR (DMSO-d6, 400 MHz) data,
ative HPLC was performed using an ODS column (XTerra®, see Table 2. HRESIMS (positive ion mode) m/z [M+ Na]+
19× 250 mm, 10 μm, Waters, Milford, MA). 369.0940 (calcd. 369.0945).
7-Hydroxy-5,4′-dimethoxy-2-arylbenzofuran (4) was yel-
2.2. Plant material low needles; 13C NMR (CDCl3, 100 MHz) and 1H NMR (CDCl3,

The fresh fruits of L. chinensis (Jacq.) R.Br. were collected in


Jiangmen, Guangdong Province, China, in September 2008, and
were identified by Prof. Xiangjiu He of School of Pharmaceutical
Sciences at Wuhan University. A voucher specimen (no.
20090920) is available at School of Pharmaceutical Sciences,
Wuhan University in Wuhan (430071), China.

2.3. Extraction and isolation

The air-dried fruits of L. chinensis (20.0 kg) were refluxed


with 70% EtOH (150 L × 3). Evaporation of the organic solvent
under a vacuum at 55 °C yielded a crude extract (3.0 kg). The
concentrated brown syrup was resuspended in water and parti-
tioned with petroleum ether (3 L ×3), chloroform (3 L × 3),
ethyl acetate (3 L ×3) and water-saturated n-butanol (3 L ×3)
gradually to afford 90.21 g, 91.72 g, 55.05 g and 554.35 g of
dried organic extracts, respectively.
The CHCl3 fraction was fractionated over a silica gel (200–300
mesh) column by eluting gradually with MeOH in CHCl3. This
process yielded 19 fractions. The CHCl3–MeOH (50:1) elution
was separated on a silica gel column, using cyclohexane/ethyl
acetate (100:1→1:1) to yield compounds 8 (35 mg), 6
(15 mg), and 14 (20 mg), respectively. The CHCl3–MeOH
(30:1) elution was further purified by a silica gel column and
eluted with cyclohexane/ethyl acetate (100:1→1:1) to obtain
compounds 1 (50 mg) and 2 (65 mg), respectively. The CHCl3–
MeOH (20:1) elution was subjected to a silica gel column and
eluted with cyclohexane/ethyl acetate (100:1→1:1), yielding
compounds 4 (250 mg), 7 (350 mg), 9 (50 mg), and 15
(30 mg), respectively. The CHCl3–MeOH (10:1) elution was fur-
ther purified with silica gel column and Sephadex LH-20 column, Fig. 1. Chemical structures of compounds 1–15.
106 X. Zeng et al. / Fitoterapia 83 (2012) 104–109

Table 1 Table 3
1
H NMR and 13C NMR data of compounds 1 and 2 in CDCl3 (δ in ppm, J in Hz). 1
H NMR, 13C NMR, 1H–1H COSY and HMBC data of compound 4 in CDCl3 (δ in
ppm, J in Hz).
Position Proton Carbon
1 13 1
Position H-NMR C- H–1H HMBC
1 2 1 2
NMR COSY
1 126.9 127.0
2 156.9
2 7.04 (s, 1H) 7.04 (d, 2, 1H) 109.1 109.3
3 6.79 (s, 1H) 100.4 C(2), C(3a), C(4),
3 147.7 147.9
C(5), C(7a), C(1′)
4 146.5 146.8
4 6.60 (d, 2.4, 1H) 94.8 H(6) C(3), C(5), C(6),
5 6.92 (d, 8.0, 1H) 6.92 (d, 7.6, 1H) 115.5 115.7
C(7), C(7a)
6 7.08 (br d, 8.6, 1H) 7.08 (dd, 7.6, 2, 1H) 122.9 123.0
5 145.5
7 7.62 (d, 16.0, 1H) 7.61 (d, 16.4, 1H) 144.5 144.6
6 6.43 (d, 2.0, 1H) 97.1 H(4) C(4), C(5), C(7), C(7a)
8 6.30 (d, 16.4, 1H) 6.29 (d, 16.4, 1H) 114.5 114.6
7 157.0
9 167.3 167.4
3a 131.0
\OCH3 3.93 (s, 3H) 3.93 (s, 3H) 55.8 55.9
7a 139.4
1′ 4.18 (m, 2H) 4.19 (t, 6.8, 2H) 64.9 65.0
1′ 123.6
\CH 2.32 (m, 1H) 2.34 (m) 34.0 34.1
2′ 7.73(d, 8.4, 1H) 126.9 H(3′) C(3′), C(4′), C(5′), C(6′)
\CH2b)c) 2.32 (m, 2H) 2.34 (m) 32.8 32.8
d) 3′ 6.89(d, 8.4, 1H) 115.9 H(2′) C(1′), C(4′), C(5′)
4′ 156.3
\Ca) 3.65 (t, 6.8, 2H) 173.7 63.1
5′ 6.89(d, 8.4, 1H) 115.9 H(6′) C(1′), C(3′), C(4′)
\CH2 1.25–1.64 (m, 2H) 1.25–1.69 (m) 28.8– 28.8–
6′ 7.73(d, 8.4, 1H) 126.9 H(5′) C(2′), C(3′), C(4′), C(5′)
31.8 31.9
5-OCH3 4.00 (s, 3H) 55.3 C(5)
\CH3 0.88 (t, 6.4, 3H) 0.88 (t, 6.4, 3H) 14.0 14.1
4′-OCH3 3.85 (s, 3H) 55.1 C(4′)
1″ 4.18 (m, 2H) 65.0
2″ 4.18 (m, 1H) 68.4
3″ 3.64 (t, 6.8, 1H) 63.1 CHCl3 was to give a white solid. GC–MS was performed to elu-
4.05 (t, 6.8, 1H) cidate the structure of the white solid. Also, the H2O fraction
a–d) Carbon groups in the structure of compounds 1 and 2 (see Fig. 1). was neutralized with dilute HCl and extracted with Et2O. The
Et2O extract was identified as ferulic acid by NMR analysis.

400 MHz) data, see Table 3. HRESIMS (positive ion mode) m/z 2.5. DPPH radical scavenging assay
[M+ H]+ 271.0967 (calcd. 271.0965).
The scavenging effects of the phenolics on DPPH radicals
2.4. Hydrolysis of compounds 1 and 2 were determined according to a previously reported method
[19]. DPPH (50 mg/L) was dissolved in MeOH. The samples
The hydrolysis reaction was determined according to the were dissolved in DMSO. The DPPH solution (995 μL) was
reported protocol [18]. Compound 1 or 2 (10 mg) was treated mixed with 5 μL of each of the samples. The mixture was shak-
with 5 mL 5% NaOH in EtOH. The mixture was refluxed for en and allowed to stand at room temperature in the dark for
10 h, and the solvent was removed by evaporation. The residue 20 min. The absorbance of the resulting solution was measured
was suspended in H2O and extracted with CHCl3. The dried spectrophotometrically at 517 nm.

2.6. Superoxide anion free radical (O2−) scavenging assay


Table 2
1
H NMR, 13C NMR, 1H–1H COSY and HMBC data of compound 3 in DMSO-d6 Superoxide radical scavenging activity was assayed by the
(δ in ppm, J in Hz).
NBT reduction method according to a previously reported
Position 1
H-NMR 13
C- 1
H–1H HMBC protocol [20] with some modification. The 1000 μL reaction
NMR COSY mixture used for the O2− scavenging activity assay contained
2 5.56 (d, 6.6, 1H) 8.81 H(3) C(1′), C(4′), C(4), Tris–HCl (pH 8.1, 50 mM, 445 μL), NADH (0.15 mM, 250 μL),
C(7a), C(9) PMS (0.03 mM, 50 μL), NBT (0.10 mM, 250 μL) and sample
3 3.55 (brs, 1H) 52.2 H(2), H(9) C(1′) solution (5 μL). All samples were dissolved in Tris–HCl
4 7.55 (s, 1H) 119.1 H(6) C(3), C(6), C(7),
50 mM, pH 8.1. The reaction was conducted at 37 °C for 5 min,
C(7a), C(8)
5 123.7 and initiated by the addition of PMS. The absorbance of the
6 7.44 (brs, 1H) 113.2 H(4) C(4), C(5), C(7), resulting solution was measured spectrophotometrically at
C(7a), C(8) 560 nm.
7 143.3
8 167.1
9 3.70 (brs, 1H) 62.6 H(3)
2.7. Antiproliferative assay
3a 129.6
7a 151.6 The antiproliferative assay was performed on four human
1′ 131.6 tumor cell lines, namely human myeloid leukemia HL-60,
2′ 6.78 (s, 1H) 115.4 H(4) C(1′), C(3′), C(4′)
human myeloid leukemia K562, human liver cancer HepG2
3′ 147.6
4′ 6.93 (s, 1H) 110.5 H(2), H(6) C(2), C(5′), C(6′) and human nasopharyngeal carcinoma CNE-1, performed
5′ 146.6 according to a reported protocol [21]. All the cells were cultured
6′ 6.78 (s, 1H) 119.0 H(4) in RPMI-1640 medium (Hyclone, Logan, UT), and supplemented
3′-OCH3 3.83 (s, 3H) 55.6 C(3′) with 10% fetal bovine serum (Hyclone) and antibiotic
7-OCH3 3.75 (s, 3H) 55.6 C(7)
(100 units/mL penicillin and 100 μg/mL streptomycin) in 5%
X. Zeng et al. / Fitoterapia 83 (2012) 104–109 107

CO2 at 37 °C. The cytotoxicity assay was performed according to be E-[6′-(5″-hydroxypentyl)tricosyl]-4-hydroxy-3-methoxy-


the MTT method in 96-well microplates. Briefly, 200 μL of ad- cinnamate, which was a new compound.
herent cells was seeded into each well of 96-well cell culture Compound 3 was obtained as amorphous powder. Posi-
plates and allowed to adhere for 12 h before drug addition, tive electrospray ionization mass spectrometry (ESI-MS) pro-
while suspended cells were seeded just before drug addition duced the ion [M + Na] + at m/z 369, indicating a molecular
with an initial density of 5 ×104 cells/mL. Each tumor cell line mass of 346, which is compatible with the molecular formula
was exposed to the test compound at concentrations of 50, 25, C18H18O7. Its molecular formula was confirmed by HR-ESIMS,
12.5, 6.25, 3.125, and 1.56 μg/mL in quadruples for 48 h. which showed the ion [M + Na] + at m/z 369.0940 (calcd.
369.0950). The 1H NMR, 13C NMR, DEPT and HMQC data for
3 (Table 2) indicated the presence of a 1,3,5-trisubstituted
3. Results and discussion benzene moiety [δH 6.78 (2H, s, H-2′, 6′), 6.93 (1H, s, H-4′)],
a 1,2,3,5-tetrasubstituted benzene ring [δH 7.55 (1H, s, H-2),
3.1. Structure elucidation of new compound 7.44 (1H, brs, H-6)], two methoxy groups [δH 3.83 (3H, s,
3′-OCH3), 3.75 (3H, s, 7-OCH3)], one oxymethine [δH 5.56
Compound 1 was obtained as white amorphous powder. (1H, d, J = 6.6 Hz, H-2)], one oxymethylene [δH 3.70 (1H,
The positive ESI-MS spectrum showed a molecular ion [M brs, H-9)] and one methene [δH 3.55 (1H, brs, H-3)]. From
+ H] + at m/z 579 and fragment ions m/z [glycerol + H] + the 1H– 1H COSY spectrum, a partial structure (\OCHCHCH2-
and [ferulate + H] + at m/z 93 and 195, respectively. In the OH) was feasibly deduced. From above, it was deduced that 3
1
H NMR spectrum (Table 1), three signals at δH 6.92 (1H, d, contains a benzofuran fragment. In the HMBC spectrum, the
J = 8.0 Hz), 7.04 (1H, d, J = 2.4 Hz), and 7.08 (1H, dd, J = 8.0, correlations of H-2 (δH 5.56) to C-9 (δC 62.6), C-4′ (δC
2.4 Hz) indicated the presence of a 1,3,4-trisubstituted ben- 110.5), C-2′ (δC 115.4), C-1′ (δC 131.6) and C-7a (δC 151.6),
zene. The signals at δH 6.30 (1H, d, J = 15.6 Hz) and 7.61 H-3 (δH 3.55) to C-3a (δC 129.6), 3′-OCH3 (δH 3.83) to C-3′
(1H, d, J = 15.6 Hz) showed a trans-double bond in the mole- (δC 147.6), 7-OCH3 (δH 3.75) to C-7 (δC 143.3), and \COOH
cule. The 1H NMR spectrum revealed the presence of a series (δC 167.1) to H-4 (δH 7.55) and H-6 (δH 7.44) suggested 3
of methylene groups, assigned to a fatty acid (δH 0.88–2.37), could be 2-(3′-hydroxy-5′-methoxyphenyl)-3-hydroxylmethyl-
and seven strongly deshielded protons at 3.64 (1H, t, 7-methoxy-2,3-dihydrobenzofuran-5-carboxylic acid. Since
J = 6.4 Hz), 4.05 (1H, t, J = 6.4 Hz) and 4.18 (5H, m). The 13C the coupling constant of H-2 was 6.6 Hz, the relative config-
NMR spectrum of 1 (Table 1) showed two carbonyls, eight ar- uration of C-2 and C-3 was determined to be trans [23]. This
omatic carbons, one \OCH3 group, three \CH2OH groups, configuration was opposite to that of the compound isolated
one \CHOH group and a series of signals at δC 14.1–34.1. A from Tarenna attenuate [24]. The optical rotation value of 3
comparison of the 1H and 13C NMR data of 1 with the ([α] 20D + 8.2 (c 0.20, MeOH)), which was different from
reported data [22] indicated that the structure of 1 is similar that of balanophonin [23], suggested the absolute configura-
to that of 1-(ω-isoferuloylalkanoyl)-glycerol. To deduce the tions of C-2 and C-3 to be 2S and 3R by comparing with the
exact structure of the fatty acid, 1 was hydrolyzed in 5% literature [25], respectively. Therefore, the structure of com-
NaOH. After the usual workup, the nonpolar organic extract pound 3 was established as 2-(3′-hydroxy-5′-methoxyphenyl)-
was analyzed with GC/MS and NMR spectroscopy, and 3 - hydroxylmethyl - 7 - methoxy - 2,3 -dihydrobenzofuran-5-
the structure of the fatty acid was identified to be 6-(4- carboxylic acid, which was a new compound.
hydroxybutyl)pentadecanoic acid. Therefore, compound 1 Compound 4 was obtained as yellow needles, and the ESI-
was elucidated to be 1-{ω-isoferul[6-(4-hydroxybutyl) MS spectrum showed an ion [M + H] + at m/z 271, consisted
pentadecanoic acid]}-glycerol, which was a new compound. with the molecular formula C16H14O4. The 1H and 1H- 1H
Compound 2 was obtained as a white amorphous powder. COSY spectrum (Table 3) indicated the presence of one AA′
The negative ESI-MS spectrum showed a molecular ion [M– BB′ spin system comprised of protons resonating at δ 6.89
H]− at m/z 601, and a fragment ion [ferulate]− at m/z 193. In (2H, d, J = 8.4 Hz) and δ 7.73 (2H, d, J = 8.4 Hz). Additionally,
the 1H NMR spectrum (Table 1), three signals at δH 6.92 (1H, the 1H NMR and 1H– 1H COSY spectrum of 4 displayed a
d, J = 8.0 Hz ), 7.04 ( 1H, d, J = 2.4 Hz), and 7.08 (1H, dd, 1,2,3,5-tetrasubstituted benzene unit [δH 6.60 (1H, d,
J = 7.6, 2.0 Hz), two signals at δH 6.30 (1H, d, J = 15.6 Hz) and J = 2.0 Hz), and δH 6.43 (1H, d, J = 2.0 Hz)]. The 13C NMR
7.61 (1H, d, J = 15.6 Hz) and one signal at δH 3.93 (3H, s) indi- spectrum (Table 3) included 14 nonequivalent carbon sig-
cated the presence of a ferulic acid moiety. The 1H NMR also nals, implying the presence of a 6-2-6 system indicative of
revealed a series of methylene groups assigned to a long an arylbenzofuran skeleton. This was further confirmed by
chain hydrocarbon (δH 0.88–1.69). Furthermore, four strongly the 1H NMR spectrum, which showed a doublet at δ 6.79
deshielded protons at 3.64 (2H, t, J = 6.8 Hz) and 4.19 (2H, t, (1H, s) assignable to H-3. These data suggested that 4 was
J = 6.8 Hz) revealed two methylene groups attached to oxygen. an isomeride of livistone C, a compound isolated previously
The 13C NMR spectrum of 2 (Table 1) showed the presence of a from the fruits of L. chinensis [15]. The NMR spectra data of
carbonyl carbon, eight aromatic carbons, a \OCH3 group, two 4 and livistone C were very similar, with the exception of
\CH2OH groups and a series of signals at δC 14.1–34.1. In one methoxy signal. On the basis of HMBC correlations
order to determine the lengths of the long chain hydrocarbon (Table 3), the singlet signals at δ 3.85 (3H, s) and δ 4.00
and the position of the methyne, 2 was hydrolyzed in 5% (3H, s), which typically represent methoxy protons, showed
NaOH. A series of long chain hydrocarbons and ferulic acid correlations with C-4′ (δ 156.3) and C-5 (δ 145.5), thus con-
was obtained from the hydrolysis. The long chain hydrocarbons firming the assignment of the methoxy groups at C-4′ and
were identified as 6-octadecylundecane-1,11-diol by means of C-5, respectively. From the data above, compound 4 was
GC/MS and NMR analysis. Thus, compound 2 was elucidated to identified as 7-hydroxy-5,4′-dimethoxy-2-arylbenzofuran.
108 X. Zeng et al. / Fitoterapia 83 (2012) 104–109

The known compounds were identified, by comparing their Table 5


spectroscopic data with the data previously reported, as 5-O- Antiproliferative activities of the phenolics (1–9) against HL-60, Mata,
HepG2 and CNE-1 cell lines.
caffeoylshikimic acid (5) [26], (E)-3-(4-hydroxyphenyl)acrylic
acid (6) [27], caffeic acid (7) [28], 3-O-caffeoylshikimic Compound IC50 (μM)
acid (8) [29], 6,7-dihydroxy-2H-chromen-2-one (9) [30],
HL-60 Mata HepG2 CNE-1
4-hydroxybenzaldehyde (10), 3-hydroxy-4-methoxybenzoic
1 26.06 ± 2.09 33.68 ± 3.22 45.96 ± 4.18 37.73 ± 2.35
acid (11), 4 - hydroxybenzoic acid (12), 4 - hydroxy - 3 -
2 28.44 ± 1.96 40.58 ± 3.72 43.16 ± 3.23 40.53 ± 3.35
methoxybenzoic acid (13), 3-hydroxy-4-methoxybenzaldehyde 3 73.20 ± 4.19 82.68 ± 3.12 62.76 ± 6.13 79.73 ± 6.25
(14) and 3,4-dihydroxybenzoic acid (15), respectively. 4 7.63 ± 0.49 10.68 ± 0.82 9.76 ± 0.73 7.73 ± 0.65
5 121.91 ± 133.96 ± 109.54 ± 92.3 ± 7.93
3.2. Antioxidant and antiproliferative activity of the phenolics 10.07 12.16 2.04
6 N150 N 150 N 150 N150
7 N150 N 150 N 150 N150
The DPPH scavenging and superoxide anion free radical 8 126.24 ± 135.79 ± N 150 N150
(O2−) scavenging abilities of the phenolics (1–9) isolated 11.55 13.34
from L. chinensis were shown in Table 4. 1 showed the high- 9 N150 N 150 N 150 113.73
± 9.78
est antioxidant activity based on the results of these two
Cisplatin 12.60 ± 0.89 15.70 ± 0.63 15.50 ± 1.21 14.54 ± 1.06
antioxidant screening models, followed by 5. Table 4 demon-
strates that 1, 2, 5 and 8 had higher antioxidant capacity than
that of caffeic acid (7) or (E)-3-(4-hydroxyphenyl)acrylic 2 and HL-60, which showed an antiproliferative efficacy of
acid (6) on an equivalent molar basis. Furthermore, the ester- 67.3% and 71.5% at the highest concentration of 100 μg/mL, re-
ification that occurred at position 6 of shikimic acid was more spectively. From the dose–response curve of antiproliferative
effective in boosting antioxidant activity than the esterifica- activity against HL-60 cells among 1, 2 and 4 (Fig. 2), 1 and 2
tion that occurred at position 4 among two caffeoylshikimic showed higher dose–response efficacy than 4. Maybe, 1 and 2,
acids. whose structures have both hydrophilic and hydrophobic
The antiproliferative activities of the phenolics against four groups, are beneficial to permeate that the complex cell system
human tumor cell lines were shown in Table 5. 4 exhibited sig- of the molecules.
nificant inhibition of proliferation against the four cell lines, and
showed stronger inhibition of the proliferation of these selected 4. Conclusions
cell lines than cisplatin. It should be noted that the analogs of 3
and 4 have been reported to exhibit significant cytotoxic activ- In current research, the phytochemical analysis of the 70%
ity against p-388, HB16, A549 and HT-29 cells [31–32]. 1 and 2 EtOH extract of the fruits of L. chinensis led to the isolation of
exhibited significant effects on these cancer cell lines. This find- fifteen phenolics, four of them were new compounds, and ten
ing was in accordance with the former research that ferulate were isolated for the first time from the genus Livistona. Sev-
and caffeic alkyl esters demonstrate antitumor proliferation ac- eral constituents of L. chinensis fruits have been shown to
tivity with colon breast, lung, and gastric human cell lines [33]. possess strong antioxidant and antiproliferative activities.
Moreover, the results showed that inhibitory activity increased Depending on the method used, 1, 2 and 4 proved to be effec-
greatly when caffeic acid was esterified with shikimic acid. 4 tive radical scavengers and cancer preventing agents in vitro.
seemed to cause complete inhibition of the viability four target Free radical induced oxidative stress has been hypothesized
cell lines, resulting in an antiproliferative efficacy of about 90% to be a major factor in the development of several degenerative
at the concentration 12.5 μg/mL. Also, 1 showed complete anti- chronic diseases. Oxidative stress can cause oxidative damage
proliferation of HL-60 at the highest concentration tested with
the antiproliferative efficacy of 75.2%. Good reducing capacity
for the tetrazolium salt was also observed for 2 against HepG
100
Cell Proliferation Inhibition (%)

Table 4 80
Antioxidant activities of the isolated compounds (1–9).

Compound IC50 (μM)


60
DPPH radical Superoxide anion
free radical (O2−)
Compound 1
1 2.26 ± 0.21 1.91 ± 0.13 Compound 2
2 2.72 ± 0.22 2.50 ± 0.23 40 Compound 4
3 4.90 ± 0.32 4.50 ± 0.31
4 4.30 ± 0.42 3.50 ± 0.33
5 2.60 ± 0.17 2.40 ± 0.09
6 7.54 ± 0.67 6.53 ± 0.49 20
0 10 20 30 40 50 60
7 3.76 ± 0.23 3.51 ± 0.25
Concentration (µg/ml )
8 2.87 ± 0.16 2.54 ± 0.11
9 6.55 ± 0.45 5.32 ± 0.33
Fig. 2. Dose–response curve of antiproliferative activity against HL-60 of
Quercetin 5.40 ± 0.30 4.60 ± 0.20
compounds 1, 2 and 4 (mean ± SD, n = 3).
X. Zeng et al. / Fitoterapia 83 (2012) 104–109 109

to large biomolecules, such as lipids, proteins, and DNA, result- [3] Zhao GP, Dai S, Chen ES. Dictionary of Traditional Chinese Medicine.
Shanghai: Shanghai Science and Technology Press; 2001. p. 2459–60.
ing in an increased risk for inflammatory diseases, cardiovascu- [4] Kaur G, Singh RP. Food Chem Toxicol 2008;46:2429–34.
lar disease, cancer, diabetes, Alzheimer's disease, cataracts, and [5] Li CY, Zeng YB, Peng F, Dai HF, Zheng YT. Chin Trad Herb Drugs 2008;39:
age-related functional decline [34–35]. The DPPH radical assay 1833–8.
[6] Wang H, Li A, Dong XP, Xu XY. J Chin Med Mat 2008;31:718–22.
was known to give reliable information concerning the antiox- [7] Chen Y, Lin XH, Li SG, Weng SM, Yao H. J Fujian Med Univ 2008;42:
idant ability of the tested compounds and extracts. At the same 93–5.
time, the superoxide anion free radical assay has been widely [8] Zhu YL, Chen SL, Peng LL, Tang L. Chem Bioeng 2007;24:35–7.
[9] Sartippour MR, Liu CH, Shao ZM, Go VL, Herber D, Nguyen M. Oncol Rep
used to evaluate the free radical scavenging ability. And this 2001;8:1355–7.
radical was generated by a chemical system composed of [10] Cheung S, Tai J. Oncol Rep 2005;14:1331–6.
PMS, NADH and oxygen. This assay was known to give reliable [11] Huang WC, Hsu RM, Chi LM, Leu YL, Chang YS, Yu JS. Cancer Lett
2007;248:137–46.
information concerning the antioxidant ability of the tested
[12] Chen P, Yang JS. Chin Trad Herb Drugs 2007;8:665–7.
compounds. The phenolics isolated from the fruits of cabbage [13] Chen P, Yang JS. Chin Pharm J 2008;43:1669–70.
palm showed potential free radical and superoxide anion free [14] Liu ZP, Cui JG, Liu HX. Cereal Oil Proc 2009;2:43–4.
radical (O2−) scavenging activities. [15] Tao Y, Yang SP, Zhang HY, Liao SG, Wei W, Yan W, et al. J Asian Nat Prod
Res 2009;11:243–9.
These results indicated the fruits of L. chinensis could be [16] Mala V, Dahot MU. Sci Int (Lahore) 1994;6:231–5.
served as cancer preventing agents, which were in accordance [17] Zeng XB, Qiu Q, Jiang CG, Jing YT, Qiu GF, He XJ. Fitoterpia 2011;82:
with their traditional uses. It can serve as potent anticancer and 609–14.
[18] Boonyaratavej S, Tantayanontha S, Kitchanachai P, Chaichantipyuth C,
antioxidant agents. The mechanisms of those compounds in Chittawong V, Miles DH. J Nat Prod 1992;55:1761–3.
cancer prevention are worthy of further investigation. [19] Yen GC, Chen HY. J Agric Food Chem 1995;43:27–32.
[20] Valentao P, Fernandes E, Carvalho F, Andrade PB, Seabra RM, Bastos
MD. Biol Pharm Bull 2002;25:1324–7.
Acknowledgments [21] He XJ, Liu RH. J Agric Food Chem 2007;55:4366–70.
[22] Kawanishi K, Hashimoto Y. Phytochemistry 1987;26:749–52.
This research was partly supported by the Start Fund of [23] Sy LK, Brown GD. Phytochemistry 1999;50:781–5.
[24] Yang XW, Zhao PJ, Ma YL, Xiao HT, Zuo YQ, He HP, et al. J Nat Prod
Wuhan University. The HR-ESIMS and optical rotations were 2007;70:521–5.
measured by Dr. Jinshan Tang, Institute of Traditional Chinese [25] Zhao XH, Chen DH, Si JY, Pan RL, Shen LG. Acta Pharmacol Sin 2002;37:
Medicine & Natural Products, Jinan University, China. The ESI- 535–8.
[26] Veit M, Weidner C, Strack D, Wray V, Witte L, Czygan FC. Phytochemistry
MS were kindly measured by Professor Naili Wang, Shenzhen 1992;31:3483–5.
Research Center of Traditional Chinese medicine & Natural [27] Chiang YM, Liu HK, Lo JM, Chien SC, Chan YF, Lee TH, et al. J Chin Chem
Products. Special thanks were given to Miss Lillian Nordahl, Soc 2003;50:161–6.
[28] Deyama T, Ikawa T, Kitagawa S, Nishibe S. Chem Pharm Bull 1987;35:
Department of Pharmacology at University of Cambridge, for 1785–9.
her kind help in manuscript preparation. [29] Ozawa T, Imagawa H. Agric Biol Chem 1988;52:595–7.
[30] Cussans NJ, Huckerby TN. Tetrahedron 1975;31:2719–26.
[31] Tsai IL, Hsieh CF, Duh CY, Chen IS. Phytochemistry 1996;43:1261–3.
References [32] Sandro M, Lauro ESB, Young GS, Patrick FJ, Chai HB, Eun JP, et al.
Phytochemistry 2002;59:635–41.
[1] Pei SJ, Cheng SY, Tong SJ. Flora of China. Beijing: Science Press; 1991. [33] Jayaprakasam B, Vanisree M, Zhang Y, Dewitt DL, Nair MG. J Agric Food
p. 25. Chem 2006;54:5375–81.
[2] Healthy Ministry of Guangzhou Force Logistics. Common Chinese Herbal [34] Ames BN, Gold LS. Mutat Res 1991;250:3–16.
Medicine Handbook. Beijing: People's Health Publishing House; 1969. [35] Ames BN, Shigenaga MK, Gold LS. Environ Health Perspect 1993;101:
p. 772–3. 35–44.

S-ar putea să vă placă și