a new megastigmane glycoside from akebia quinata

7
RESEARCH ARTICLE A new megastigmane glycoside from Akebia quinata Hong-Guang Jin A Ryun Kim Hae Ju Ko Eun-Rhan Woo Received: 20 December 2013 / Accepted: 9 February 2014 Ó The Pharmaceutical Society of Korea 2014 Abstract A new megastigmane glycoside, 8S*,9R*- megastigman-3-one-4,6-diene-8,9-diol-9-O-b-D-glucopy- ranoside, named akequintoside D (1), as well as six known compounds, roseoside II (2), 3-O-caffeoylquinic acid (3), methyl-3-O-caffeoylquinate (4), 3,4,5-trimethoxyphenyl-b- D-glucopyranoside (5), cuneataside D (6), 3,4-dimethoxy- phenyl-6-O-(a-L-rhamnopyranosyl)-b-D-glucopyranoside (7) were isolated from the stem of Akebia quinata. The structures of compounds (1-7) were identified based on 1D and 2D NMR, including 1 H– 1 H COSY, HSQC, HMBC and NOESY spectroscopic analyses. The inhibitory activity of these isolated compounds against interleukin-6 (IL-6) production in TNF-a stimulated MG-63 cells was also examined. Keywords Megastigmane glycoside IL-6 inhibitory effect Lardizabalaceae Akebia quinata Introduction Akebia quinata D ECAISENE (Lardizabalaceae) is a creeping woody vine which is widely distributed in East Asia, including Korea, China, and Japan (Lee 2003). Its dried stem is used as a diuretic agent for the treatment of the painful urinary dribbling, edema and ascites (Ahn 1998; Bensky et al. 2004). Previous phytochemical investigations resulted in the isolation of triterpenes, triterpene glyco- sides, and phenylethanoid glycosides (Gao and Wang 2006; Mimaki et al. 2003; 2007). Regarding the biological activity of A. quinata, only the cytotoxic effect of oleanane disaccharides has been reported so far (Jung et al. 2004). Moreover, the anti-inflammatory activity of this plant has not been explored in detail. In an ongoing investigation into anti-inflammatory compounds from this plant, the methanol extract of A. quinata was investigated. By means of repeated column chromatography using silica gel, MCI gel, Sephadex LH- 20, and LiChroprep RP-18, a new megastigmane glycoside, akequintoside D (1), along with six known compounds were isolated. The structures of the known compounds were identified as roseoside II (2), 3-O-caffeoylquinic acid (3), methyl-3-O-caffeoylquinate (4), 3,4,5-trimethoxyphenyl- b-D-glucopyranoside (5), cuneataside D (6), and 3,4-dime- thoxyphenyl-6-O-(a-L-rhamnopyranosyl)-b-D-glucopyranoside (7), by comparing their spectroscopic data with those reported in the literature (Fig. 1). Furthermore, these six known compounds were isolated from this plant for the first time. The inhibitory activity of these isolated compounds against IL-6 production in TNF-a stimulated MG-63 cells was examined. This paper reports the isolation and structural charac- terization of these compounds and their inhibitory activities against IL-6 production. Electronic supplementary material The online version of this article (doi:10.1007/s12272-014-0357-x) contains supplementary material, which is available to authorized users. H.-G. Jin A. R. Kim H. J. Ko E.-R. Woo (&) College of Pharmacy, Chosun University, 375 Seosuk-dong, Dong-gu, Gwangju 501-759, Republic of Korea e-mail: [email protected] H.-G. Jin College of Pharmacy, Jilin Medical College, Jilin 132013, China 123 Arch. Pharm. Res. DOI 10.1007/s12272-014-0357-x

Upload: eun-rhan

Post on 24-Jan-2017

213 views

Category:

Documents


0 download

TRANSCRIPT

RESEARCH ARTICLE

A new megastigmane glycoside from Akebia quinata

Hong-Guang Jin • A Ryun Kim • Hae Ju Ko •

Eun-Rhan Woo

Received: 20 December 2013 / Accepted: 9 February 2014

� The Pharmaceutical Society of Korea 2014

Abstract A new megastigmane glycoside, 8S*,9R*-

megastigman-3-one-4,6-diene-8,9-diol-9-O-b-D-glucopy-

ranoside, named akequintoside D (1), as well as six known

compounds, roseoside II (2), 3-O-caffeoylquinic acid (3),

methyl-3-O-caffeoylquinate (4), 3,4,5-trimethoxyphenyl-b-

D-glucopyranoside (5), cuneataside D (6), 3,4-dimethoxy-

phenyl-6-O-(a-L-rhamnopyranosyl)-b-D-glucopyranoside

(7) were isolated from the stem of Akebia quinata. The

structures of compounds (1-7) were identified based on 1D

and 2D NMR, including 1H–1H COSY, HSQC, HMBC and

NOESY spectroscopic analyses. The inhibitory activity of

these isolated compounds against interleukin-6 (IL-6)

production in TNF-a stimulated MG-63 cells was also

examined.

Keywords Megastigmane glycoside � IL-6 inhibitory

effect � Lardizabalaceae � Akebia quinata

Introduction

Akebia quinata DECAISENE (Lardizabalaceae) is a creeping

woody vine which is widely distributed in East Asia,

including Korea, China, and Japan (Lee 2003). Its dried

stem is used as a diuretic agent for the treatment of the

painful urinary dribbling, edema and ascites (Ahn 1998;

Bensky et al. 2004). Previous phytochemical investigations

resulted in the isolation of triterpenes, triterpene glyco-

sides, and phenylethanoid glycosides (Gao and Wang

2006; Mimaki et al. 2003; 2007). Regarding the biological

activity of A. quinata, only the cytotoxic effect of oleanane

disaccharides has been reported so far (Jung et al. 2004).

Moreover, the anti-inflammatory activity of this plant has

not been explored in detail.

In an ongoing investigation into anti-inflammatory

compounds from this plant, the methanol extract of A.

quinata was investigated. By means of repeated column

chromatography using silica gel, MCI gel, Sephadex LH-

20, and LiChroprep RP-18, a new megastigmane glycoside,

akequintoside D (1), along with six known compounds were

isolated. The structures of the known compounds were

identified as roseoside II (2), 3-O-caffeoylquinic acid (3),

methyl-3-O-caffeoylquinate (4), 3,4,5-trimethoxyphenyl-

b-D-glucopyranoside (5), cuneataside D (6), and 3,4-dime-

thoxyphenyl-6-O-(a-L-rhamnopyranosyl)-b-D-glucopyranoside

(7), by comparing their spectroscopic data with those

reported in the literature (Fig. 1). Furthermore, these six

known compounds were isolated from this plant for the first

time. The inhibitory activity of these isolated compounds

against IL-6 production in TNF-a stimulated MG-63 cells

was examined.

This paper reports the isolation and structural charac-

terization of these compounds and their inhibitory activities

against IL-6 production.

Electronic supplementary material The online version of thisarticle (doi:10.1007/s12272-014-0357-x) contains supplementarymaterial, which is available to authorized users.

H.-G. Jin � A. R. Kim � H. J. Ko � E.-R. Woo (&)

College of Pharmacy, Chosun University, 375 Seosuk-dong,

Dong-gu, Gwangju 501-759, Republic of Korea

e-mail: [email protected]

H.-G. Jin

College of Pharmacy, Jilin Medical College, Jilin 132013, China

123

Arch. Pharm. Res.

DOI 10.1007/s12272-014-0357-x

Materials and methods

General experimental procedure

Optical rotations were measured using an Autopol-IV

polarimeter. IR spectra were recorded on an IMS 85

(Bruker). CD spectra were recorded on a JASCO J-810

spectropolarimeter. HR-ESI–MS spectra were obtained on

a Q-TOF (Synapt HDMS system, Waters, USA) mass

spectrometer. NMR spectra, including NOESY, COSY,

heteronuclear multiple quantum coherence (HMQC) and

HMBC experiments, were recorded on a Varian UNITY

INOVA 500 NMR spectrometer (KBSI-Gwangju center)

operating at 500 MHz (1H) and 125 MHz (13C), respec-

tively, with chemical shifts given in ppm (d). TLC was

carried out on precoated Kieselgel 60 F254 (art. 5715,

Merck) and RP-18 F254s (art. 15389, Merck) plates. Col-

umn chromatography was performed on silica gel 60

(40–63 and 63–200 lm, Merck), MCI gel CHP20P

(75–150 lm, Mitsubishi Chemical Co.), and Sephadex LH-

20 (25–100 lm, Sigma). Silver carbonate (Ag2CO3,

Aldrich Co.) and (?)-D-glucose (C6H12O6, Sigma) were

used as neutralization reagent and standard sugar on acid

hydrolysis experiment, respectively. Low pressure liquid

chromatography was carried out over a Merck Lichroprep

Lobar�-A RP-18 (240 9 10 mm) column with a FMI

QSY-0 pump (ISCO).

Plant materials

The stem of A. quinata were collected in Gyeongju, Gy-

eongbuk province, Korea, in August 2011 and identified by

Dr. J. H. Lee, Professor of the department of Korean

Medicine, Dongguk University. A voucher specimen

(CSU-877-17) was deposited in the Herbarium of the

College of Pharmacy, Chosun University.

Extraction and isolation

The air-dried stem of A. quinata (11 kg) were cut and

extracted with MeOH three times for 4 h at 80 �C. The

resultant MeOH extract (480 g) was suspended in water

(1.5 L 9 3) and then partitioned sequentially with equal

volumes of dichloromethane, ethyl acetate, and n-butanol.

Each fraction was evaporated in vaccuo to yield the resi-

dues of CH2Cl2 (45.2 g), EtOAc (11.0 g), n-BuOH

(57.0 g), and water (150.3 g) extracts. The n-BuOH soluble

fraction (57.0 g) was subjected to column chromatography

(CC) over a diaion HP 20 column and eluted with H2O/

MeOH (100:0 ? 0:100) gradient system. The fractions

were combined based on their TLC pattern to yield sub-

fractions designated B1–B6. Fraction B2 (3.47 g) was

purified by MCI gel CC (MeOH/H2O, 1:9 ? 2:8) to yield

four subfractions (B21–B24). Subfraction B23 (0.54 g),

containing 3, 5, and 7 was purified by Lichroprep RP 18

OOH O

OHOH

HOO

OH

1

3 5

6

13

8

1'

6'

121110

O OOH

OHHO

O

OH

OH

1 2

OHHO

COORHO

O

OOH

OH

R2

R3R1

OOH

OHHO

O

OR4

3 R = H 5 R1 = OCH3 R2 = OCH3 R3 = OCH3 R4 = H4 R = CH3 6 R1 = OCH3 R2 = OH R3 = OH R4 = Rha

7 R1 = OCH3 R2 = OCH3 R3 = H R4 = Rha

Fig. 1 Structures of compounds 1–7

H.-G. Jin et al.

123

CC (MeOH/H2O, 1:20), and finally by silica gel CC

(CHCl3/MeOH/H2O, 4:1:0.2 ? 2:1:0.2) to yield 3

(121.0 mg), 5 (26.5 mg), and 7 (17.0 mg). In addition,

subfraction B21 (2.04 g) was purified by Sephadex LH 20

CC (MeOH/H2O, 1:20), and finally by silica gel CC

(CHCl3/MeOH/H2O, 4:1:0.2 ? 2:1:0.2) to yield 6

(15.0 mg). Fraction B3 (5.6 g) was subjected to silica gel

CC eluting with a CHCl3/MeOH/H2O (6:1:0.1 ? 2:1:0.1)

in a gradient system to yield six subfractions (B31–B36).

Subfraction B31 (0.79 g) was then purified by repeated

Lichroprep RP 18 CC (MeOH/H2O, 1:3) to yield 1

(4.4 mg), 2 (3.2 mg), and 4 (3.0 mg).

Akeqintoside D (1)

Colorless gum; [a]D20 -19.6� (MeOH; c 0.37); HR-ESI–MS

(positive mode) m/z: 409.1841 [M ? Na]? (calcd for C19-

H30O8Na, 409.1838); IR mmax (film) cm-1: 3,410, 2,935,

1,655, 1,650, 1,076, 1,035; 1H, and 13C NMR: see Table 1.

Roseoside II (2)

Colorless gum; [a]D20 ?82.9� (MeOH; c 0.16); HR-ESI–MS

m/z: 385.1862 [M-H]- (calcd for C19H29O8, 385.1861);

CD (MeOH, c 8.5 9 10-6 M): 241 (?41.37), 318

(-54.25) nm; IR mmax (film) cm-1: 3,400, 2,940, 1,653,

1,076, 1,035; 1H , and 13C NMR: see Table 1.

3-O-Caffeoylquinic acid (3)

Brown powder; [a]D20 -134.6� (MeOH; c 0.44); ESI–MS

m/z: 353 [M-H]?; IR mmax (film) cm-l: 3,412, 2,928,

1,689, 1,082, 853; 1H NMR (500 MHz, CD3OD) 7.57 (1H,

d, J = 16.0 Hz, H-70), 7.05 (1H, d, J = 2.0 Hz, H-20), 6.94

(1H, dd, J = 2.0, 8.0 Hz, H-60), 6.77 (1H, d, J = 8.0 Hz,

H-50), 6.29 (1H, d, J = 16.0 Hz, H-80), 5.38 (1H, dd,

J = 5.0, 10.0 Hz, H-3), 4.13 (1H, dd, J = 3.0, 6.0 Hz,

H-5), 3.68 (1H, dd, J = 3.0, 10.0 Hz, H-4), 2.15 (1H, dd,

J = 3.0, 15.0 Hz, H-6), 2.10 (1H, m, H-2), 2.00 (1H, m,

H-2), 1.94 (1H, dd, J = 3.0, 15.0 Hz, H-6); 13C NMR

(125 MHz, CD3OD) d: 181.1 (C-7), 169.3 (C-90), 149.7 (C-

40), 147.0 (C-70), 146.9 (C-30), 127.9 (C-10), 123.1 (C-60),116.6 (C-50), 115.7 (C-20), 115.2 (C-80), 77.9 (C-1), 75.2

(C-4), 73.2 (C-5), 72.8 (C-3), 40.8 (C-2), 39.2 (C-6).

Methyl-3-O-caffeoylquinate (4)

Pale brownish powder; [a]D20 -30.7� (MeOH; c 0.15); ESI–

MS m/z: 367 [M-H]?; IR mmax (film) cm-l: 3,408, 2,930,

1,672, 1,082, 872; 1H NMR (500 MHz, CD3OD) d: 7.53

(1H, d, J = 16.0 Hz, H-70), 7.04 (1H, d, J = 2.0 Hz, H-20),6.94 (1H, dd, J = 2.0, 8.0 Hz, H-60), 6.77 (1H, d,

J = 8.0 Hz, H-50), 6.22 (1H, d, J = 16.0 Hz, H-80), 5.27

(1H, dd, J = 5.0, 7.5 Hz, H-3), 4.13 (1H, dt, J = 3.0,

7.5 Hz, H-5), 3.73 (1H, dt, J = 3.0, 7.5 Hz, H-4), 3.69

Table 1 1H,13C NMR Data of

1 and 2

500 MHz, CD3OD for 1H and

125 MHz for 13C NMR;

chemical shifts in ppm relative

to TMS; coupling constants

(J) in Hz

No. 1 2

dH dC dH dC

1 40.1 42.6

2 2.37 (2H, s) 54.6 2.15 (1H, d, J = 17.0 Hz) 50.8

2.53 (1H, d, J = 17.0 Hz)

3 202.1 201.3

4 5.96 (1H, s) 127.2 5.86 (1H, s) 127.3

5 159.2 167.4

6 145.1 80.1

7 6.10 (1H, d, J = 9.5 Hz) 135.5 5.87 (1H, d, J = 1.5 Hz) 131.7

8 4.93 (1H, dd, J = 4.0, 9.5 Hz) 71.4 5.87 (1H, d, J = 3.0 Hz) 135.4

9 3.96 (1H, dd, J = 4.0, 6.5 Hz) 79.6 4.42 (1H, qdd, J = 1.5, 3.0, 6.5 Hz) 77.4

10 1.24 (3H, d, J = 6.5 Hz) 15.6 1.29 (3H, d, J = 6.5 Hz) 21.3

11 1.34 (3H, s) 29.8 1.04 (3H, s) 23.6

12 1.38 (3H, s) 29.9 1.03 (3H, s) 24.8

13 2.14 (3H, s) 22.9 1.92 (3H, s) 19.3

10 4.38 (1H, d, J = 7.5 Hz) 103.1 4.34 (1H, d, J = 7.5 Hz) 102.9

20 3.20 (1H, dd, J = 7.5, 9.0 Hz) 75.1 3.21 (1H, dd, J = 7.5, 9.0 Hz) 75.4

30 3.37 (1H, dd, J = 9.0, 9.0 Hz) 78.1 3.38 (1H, dd, J = 9.0, 9.0 Hz) 78.2

40 3.29 (1H, dd, J = 9.0, 9.0 Hz) 71.7 3.30 (1H, dd, J = 9.0, 9.0 Hz) 71.8

50 3.29 (1H, ddd, J = 2.0, 5.0, 9.0 Hz) 78.0 3.29 (1H, ddd, J = 2.0, 5.0, 9.0 Hz) 78.2

60 3.67 (1H, dd, J = 5.0, 11.5 Hz) 62.8 3.63 (1H, dd, J = 5.0, 11.5 Hz) 63.0

3.86 (1H, dd, J = 2.0, 11.5 Hz) 3.86 (1H, dd, J = 2.0, 11.5 Hz)

Megastigmane glycoside from Akebia quinata

123

(3H, s, H-8), 2.20 (1H, dd, J = 3.0, 13.5 Hz, H-6), 2.17

(2H, dd, J = 7.5, 13.5 Hz, H-2), 2.01 (1H, dd, J = 7.5,

13.5 Hz, H-6); 13C NMR (125 MHz, CD3OD) d: 175.6 (C-

7), 168.4 (C-90), 150.0 (C-40), 147.4 (C-70), 147.1 (C-30),127.7 (C-10), 123.1 (C-60), 116.7 (C-50), 115.2 (C-20), 115.1

(C-80), 75.9 (C-1), 72.6 (C-4), 72.3 (C-3), 70.4 (C-5), 53.1

(C-8), 38.2 (C-6), 37.9 (C-2).

3,4,5-Trimethoxyphenyl-b-D-glucopyranoside (5)

White powder; [a]D20 -22.3� (MeOH; c 0.38); EI-MS m/z:

346 [M]?; IR mmax (film) cm-l: 3,410, 2,910, 1,650, 1,075;1H NMR (500 MHz, CD3OD) d: 6.49 (2H, s, H-2, 5), 4.81

(1H, d, J = 7.5 Hz, H-10), 3.92 (1H, dd, J = 2.0, 12.0 Hz,

H-60), 3.81 (6H, s, 3, 5-OCH3), 3.70 (3H, s, 4-OCH3), 3.66

(1H, dd, J = 6.5, 12.0 Hz, H-60), 3.33–3.47 (4H, m, H-20,30, 40, and 50); 13C NMR (125 MHz, CD3OD) d: 156.2 (C-

1), 154.9 (C-3), 154.9 (C-5), 134.6 (C-4), 103.3 (C-10), 96.2

(C-2), 96.2 (C-6), 78.6 (C-50), 78.2 (C-30), 75.1 (C-20), 71.8

(C-40), 62.9 (C-60), 61.4 (4-OCH3), 56.7 (3, 5-OCH3).

Cuneataside D (6)

White amorphous powder; [a]D20 -60.3� (H2O; c 0.10);

ESI–MS m/z: 471 [M ? Na]?; IR mmax (film) cm-l; 3,400,

1,618, 1,514, 1,441, 1,250, 1,203, 1,055; 1H NMR

(500 MHz, CD3OD) d: 6.74 (1H, d, J = 2.5 Hz, H-5), 6.71

(1H, d, J = 8.5 Hz, H-2), 6.58 (1H, dd, J = 2.5, 8.5 Hz,

H-6), 4.71 (1H, d, J = 7.5 Hz, H-10), 4.71 (1H, d,

J = 1.5 Hz, H-100), 4.02 (1H, dd, J = 1.5, 11.0 Hz, H-60),3.83 (3H, s, 3-OCH3), 3.82 (1H, m, H-200), 3.57–3.68 (3H,

m, H-300, 500, and H-50), 3.36–3.51 (4H, m, H-20, 30, 40, and

H-400), 1.22 (3H, d, J = 6.0 Hz, H-600); 13C NMR

(125 MHz, CD3OD) d: 152.6 (C-1), 149.4 (C-3), 143.2 (C-

4), 116.2 (C-5), 110.2 (C-6), 104.1 (C-2), 103.9 (C-10),102.3 (C-100), 78.1 (C-30), 77.0 (C-50), 75.1 (C-20), 74.2 (C-

400), 72.5 (C-500), 72.3 (C-200), 71.7 (C-40), 70.0 (C-300), 68.1

(C-60), 56.6 (3-OCH3), 18.1 (C-600).

3,4-Dimethoxyphenyl-6-O-(a-L-rhamnopyranosyl)-b-D-

glucopyranoside (7)

Amorphous solid; [a]D20 -56.8� (MeOH; c 1.2); ESI–MS m/z:

485 [M ? Na]?; IR mmax (film) cm-l: 3,410, 1,614, 1,506,

1,447, 1,241, 1,213, 1,055; 1H NMR (500 MHz, CD3OD)

d: 6.87 (1H, d, J = 9.0 Hz, H-5), 6.76 (1H, d, J = 2.5 Hz,

H-2), 6.66 (1H, dd, J = 2.5, 9.0 Hz, H-6), 4.75 (1H, d,

J = 7.5 Hz, H-10), 4.71 (1H, d, J = 1.5 Hz, H-100), 4.02

(1H, dd, J = 1.5, 11.0 Hz, H-60), 3.82 (1H, dd, J = 1.5,

10.0 Hz, H-200), 3.81 (3H, s, 3-OCH3), 3.78 (3H, s,

4-OCH3), 3.57–3.69 (3H, m, H-300, 500 and H-50), 3.34–3.55

(4H, m, H-20, 30, 40 and H-400), 1.21 (3H, d, J = 6.0 Hz,

H-600); 13C NMR (125 MHz, CD3OD) d: 153.9 (C-1), 151.2

(C-3), 146.3 (C-4), 114.1 (C-5), 109.4 (C-6), 104.4 (C-2),

103.5 (C-10), 102.3 (C-100), 78.1 (C-30), 77.1 (C-50), 75.1

(C-20), 74.2 (C-400), 72.5 (C-500), 72.3 (C-200), 71.7 (C-40),70.0 (C-300), 68.1 (C-60), 57.3 (3-OCH3), 56.7 (4-OCH3),

18.1 (C-600).

Acidic hydrolysis of 1

Compound 1 (2 mg) was dissolved in 1 N HCl (1 mL) and

MeOH (1 mL) and refluxed at 90 �C for 90 min (Kim et al.

2004). The reaction solution was evaporated under reduced

pressure, and the hydrolysate was extracted with EtOAc

(3 mL 9 3). The aqueous fraction was neutralized with

Ag2CO3, filtered, and the filtrate was concentrated under

reduced pressure. The residue was compared with standard

sugar using TLC (CHCl3:MeOH:H2O, 6/4/1), which

showed the sugar to be (?)-D-glucose (Rf = 0.13) in 1.

Bioassay of IL-6

IL-6 bioassay was carried out using a slight modification of

an established method (Kim et al. 2003; Liu et al. 2006).

Briefly, 500 lL of the MG-63 cells (3 9 104 cells/mL) in

DMEM containing 10 % FBS were dispensed into a 24-well

plate; the culture was incubated for 24 h at 37 �C. Then,

5 lL of TNF-a (10 ng/mL), 5 lL of BAY 11-7085 (10 ng/mL),

and 5 lL of the DMSO with or without the compounds

(100 lg/mL) were added. After incubation at 37 �C with

5 % CO2 for 24 h, the medium was stored at -20 �C until

measurement. The IL-6 content of the medium was measured

in an ELISA procedure. 96-well plates were coated with

100 lL of purified rat anti-human IL-6 monoclonal antibody

in 0.1 M NaHCO3 (pH 9.6) by overnight incubation at 4 �C.

The wells were blocked with 200 lL of 3 % BSA in PBS for

2 h at room temperature (RT) and then incubated with

100 lL of specific antibody for 2 h at RT. 100 lL of HRP

conjugated rabbit anti-goat IgG (1:1000 dilution) was added

to each well and incubated for 2 h at RT. 100 lL of TMB

(3,30,5,50-tetramethyl-benzidine) substrate solution was

added and incubated for 10 min at RT. The color reaction

was stopped with 50 lL of 0.4 N HCl and the optical density

was read at 450 nm using a Microplate Reader (Molecular

Devices Co., Ltd., USA).

Results and discussion

Repeated column chromatography of the n-BuOH soluble

fraction of the stem of A. quinata yielded a new meg-

astigmane glycoside, named akequintoside D (1), as well as

six known phenolic compounds 2–7 (Fig. 1).

Compound 1 was obtained as colorless gum, [a]D20

-19.6� (MeOH). Its molecular formula was determined to

H.-G. Jin et al.

123

be C19H30O8 by HR-ESI–MS data at m/z 409.1841

[M ? Na]? (calcd for C19H30O8Na 409.1838). In the IR

spectrum, the absorption bands for the hydroxyl

(3,410 cm-1), and carbonyl (1,655 cm-1) groups were

observed. The 1H NMR spectrum (Table 1) of 1 showed

four methyl protons at dH 1.24 (3H, d, J = 6.5 Hz, H-10),

1.34 (3H, s, H-11), 1.38 (3H, s, H-12) and 2.14 (3H, s,

H-13), two oxymethine protons at dH 4.93 (1H, dd,

J = 4.0, 9.5 Hz, H-8) and 3.96 (1H, dd, J = 4.0, 6.5 Hz,

H-9), two olefinic protons at dH 5.96 (1H, s, H-4) and 6.10

(1H, d, J = 9.5 Hz, H-7), methylene protons at dH 2.37

(2H, s, H-2), in addition to a glucosyl anomeric proton at

dH 4.38 (1H, d, J = 7.5 Hz, H-10). Acid hydrolysis of 1 in

refluxing 1 N HCl/MeOH afforded (?)-D-glucose which

was detected by direct comparison with an authentic

sample using co-TLC (Kim et al. 2004). In the 13C NMR

spectrum (Table 1), 13 carbon signals appeared besides

those of sugar unit, which included one carbonyl carbon at

dC 202.1 (C-3), two oxygenated methine carbons at dC 71.4

(C-8) and 79.6 (C-9), two olefinic carbons at dC 127.2 (C-

4) and 135.5 (C-7), four methyl carbons at dC 15.6 (C-10),

29.8 (C-11), 29.9 (C-12) and 22.9 (C-13). These spectral

data indicated that 1 was to be a megastigman derivative

(Lee et al. 2011; 2012; Jin et al. 2012). In addition, the

signals from the sugar unit appeared at dH 4.38 (1H, d,

J = 7.5 Hz, H-10), 3.20 (1H, dd, J = 7.5, 9.0 Hz, H-20),3.37 (1H, dd, J = 9.0, 9.0 Hz, H-30), 3.29 (1H, dd, J = 9.0,

9.0 Hz, H-40), 3.29 (1H, ddd, J = 2.0, 5.0, 9.0 Hz, H-50),3.67 (1H, dd, J = 5.0, 11.5 Hz, H-60a), 3.86 (1H, dd,

J = 2.0, 11.5 Hz, H-60b) [dC 103.1 (C-10), 75.1 (C-20), 78.1

(C-30), 71.7 (C-40), 78.0 (C-50), 62.8 (C-60)] and acid

hydrolysis experiment strongly supported the presence of

D-glucopyranose (Ishimaru et al. 1987). The coupling

constant (J = 7.5 Hz) of the anomeric proton of D-glucose

indicated it to be the b-form (Ishimaru et al. 1987). Based

on the 1H and 13C NMR data, the structure of 1 was closely

related to reseoside II, which was isolated from Alangium

premnifolium except for the different locations of hydroxyl

group and double bond in 1 (Otsuka et al. 1995). The

glycosidic linkage was established by a HMBC experiment

and comparison of the reported NMR data. The HMBC

correlation between H-10 and C-9 suggested that glucose

was attached to C-9 of megastigmane moiety. Furthermore,

long-range correlations were observed between the fol-

lowing protons and carbons (H-4 and C-2/C-6/C-13; H-7

and C-1/C-9; H-8 and C-6/C-9/C-10). In the 1H–1H COSY

spectrum, the olefinic proton of H-7 showed couplings with

H-8, H-9 and H-10 (Fig. 2). The relative configuration of 1

was proposed from a NOESY experiment (Fig. 3), and a

comparison of the observed and reported NMR data (Ot-

suka et al. 1992; Yajima et al. 2009; Lee et al. 2011). In the

NOESY spectrum, correlations between H-10, tentatively

assigned an a-orientation (Lee et al. 2011), and H-9, H-8

indicated that these are on the same side (a). These NOE

correlations suggested the relative configuration at C-8 and

C-9 as shown in Fig. 3. Accordingly, the structure of 1 is

proposed to be 8S*,9R*-megastigman-3-one-4,6-diene-8,9-

diol-9-O-b-D-glucopyranoside, named, akequintoside D.

Compound 2 was obtained as colorless gum, [a]D20

?82.9� (MeOH), and has the same molecular formula

(C19H30O8) as 1 by HR-ESI–MS (m/z 385.1862 [M-H]-).

The 1H, and 13C NMR spectra showed a typical pattern of

megastigmane glycoside. The 1H NMR spectrum (Table 1)

of 2 showed four methyls [dH 1.29 (3H, d, J = 6.5 Hz,

H-10), 1.04 (3H, s, H-11), 1.03 (3H, s, H-12), 1.92 (3H, s,

H-13)], one oxymethine [dH 4.42 (1H, qdd, J = 1.5, 3.0,

6.5 Hz, H-9)], three methines [dH 5.86 (1H, s, H-4), 5.87

(1H, d, J = 1.5 Hz, H-7), 5.87 (1H, d, J = 3.0 Hz, H-8)],

one methylene [dH 2.15 (1H, d, J = 17.0 Hz, H-2a), 2.53

(1H, d, J = 17.0 Hz, H-2b)], and a b-D-glucopyranosyl

moiety [dH 4. 34 (1H, d, J = 7.5 Hz, H-10)] (Ishimaru et al.

1987). In the 13C NMR spectrum (Table 1), 13 carbon

signals appeared beside the b-D-glucose moiety, which

included one carbonyl (dC 201.3), one oxygenated methine

(dC 77.4), three methines (dC 127.3, 131.7, 135.4), four

quaternary carbons (dC 42.6, 167.4, 80.1), and four methyls

(dC 21.3, 23.6, 24.8, 19.3). The 1H,13C NMR, HSQC, and

HMBC spectra indicated that compound 2 had the same

planar ‘‘roseoside’’ skeleton (Otsuka et al. 1995). Further-

more, the CD spectrum of 2 showed a positive Cotton

effect at 241 nm and a negative effect at 318 nm, which

indicated the absolute configurations of 2 to be 6S, 9R-

OO

OH

OH

OH

OH

O

HO

Fig. 2 Selected 1H–1H COSY (dark line), HMBC (H ? C) corre-

lations of compound 1

OO

OH

OH

OH

OH

O

HO

Fig. 3 Key NOESY correlations of compound 1

Megastigmane glycoside from Akebia quinata

123

configurations (Otsuka et al. 1995; Yamano and Ito 2005).

Based on the above results, the structure of 2 was identified

as reseoside II, which was isolated from Vinca rosea and

Alangium premnifolium (Bhakuni et al. 1974; Otsuka et al.

1995).

The structures of five known compounds were identified

as 3-O-caffeoylquinic acid (3) (Nishimura et al. 1984; Teng

et al. 2002), methyl-3-O-caffeoylquinate (4) (Ida et al.

1994), 3,4,5-trimethoxyphenyl-b-D-glucopyranoside (5)

(Shimomura et al. 1988), cuneataside D (6), (Chang and

Case 2005) and 3,4-dimethoxyphenyl-6-O-(a-L-rhamno-

pyranosyl)-b-D-glucopyranoside (7) (Graikou et al. 2005),

by comparing their spectroscopic data with those reported

in the literature.

IL-6 is a cytokine, originally identified as a T cell

derived factor that regulates B-cell growth and differenti-

ation (Hirano et al. 1986). Human IL-6 is an important

component of the inflammatory cascade. Dysregulation of

IL-6 production has been implicated in a variety of

inflammatory/autoimmune disease states, including rheu-

matoid arthritis, cardiac myxoma, Castleman’s disease, and

mesangial proliferative glomerulonephritis (Hirano et al.

1990). The proinflammatory cytokines IL-1 and TNF-amarkedly stimulate the production IL-6 (Van Damme et al.

1987).

The inhibitory activity of the isolated compounds (1–7)

against IL-6 production in TNF-a stimulated MG-63 cells

was examined. Among them, compound 1 showed mod-

erate inhibitory activity against IL-6 production in TNF-astimulated MG-63 cells, while compounds 2, 4, 5, and 7

showed negligible activity (Fig. 3; Table 2).

Table 2 Inhibitory effect of compounds 1–7 against IL-6 production

in TNF-a stimulated MG 63 cells

Treatment IL-6 (pg/mL) Inhibition (%)

None 50.12 ± 1.8 –

TNF-a 250.6 ± 24.8 –

Bay 11-7085 30.2 ± 2.1** 87.9**

1 175.4 ± 6.8** 30.1**

2 210.5 ± 4.9** 15.9**

3 233.1 ± 9.8 7.39

4 213.0 ± 7.0 15.35**

5 220.5 ± 4.3** 12.5**

6 255.6 ± 4.9 0.0

7 213.0 ± 10.7* 14.8*

MG-63 cells (3 9 104) were incubated for 24 h. Cultures were

incubated with or without compounds (100 lg/mL) for 30 min and

then stimulated with TNF-a (10 ng/mL) for 24 h. IL-6 in the super-

natant was measured by ELAISA as described in Materials and

Methods. Results are expressed as the mean ± SE from three dif-

ferent experiments. BAY 11-7085 was used as a positive control

*P \ 0.05 or **P \ 0.01 compared with TNF-a treated value

Fig. 4 Inhibitory effect of compounds 1–7 against IL-6 production in

TNF-a sitimulated MG-63cells. MG-63 cells (3 9 104) were incu-

bated for 24 h. Cultures were incubated with or without compounds

(100 lg/mL) for 30 min and then stimulated with TNF-a (10 ng/mL)

for 24 h. IL-6 in the supernatant was measured by ELAISA as

described in Materials and Methods. Results are expressed as the

mean ± SE from three different experiments. BAY 11-7085 was used

as a positive control. *P \ 0.05 or **P \ 0.01 compared with TNF-atreated value

H.-G. Jin et al.

123

In conclusion, this paper reports the isolation, charac-

terization, and inhibitory activity of 7 isolates, including a

new compound and six known compounds, from A. qui-

nata. Fig. 4.

Acknowledgments This work was supported by research funds

from Chosun University in 2013.

References

Ahn, D.K. 1998. Illustrated book of Korean medicinal herbs. Seoul:

Kyo-Hak Publishing.

Bensky, D., S. Clavey, and E. Stoger. 2004. Chinese herbal medicine:

materia medica, 3rd ed. Seattle: Eastland Press.

Bhakuni, D.S., P.P. Joshi, H. Uprety, and R.S. Kapil. 1974.

Roseoside—A C13 glycoside from Vinca rosea. Phytochemistry

13: 2541–2543.

Chang, J., and R. Case. 2005. Phenolic glycosides and ionone

glycoside from the stem of Sargentodoxa cuneata. Phytochem-

istry 66: 2752–2758.

Gao, H., and Z. Wang. 2006. Triterpenoid saponins and phenyleth-

anoid glycosides from stem of Akebia trifoliata var. australis.

Phytochemistry 67: 2697–2705.

Graikou, K., N. Aligiannis, I. Chinou, A.-L. Skaltsounis, F. Tillequin,

and M. Litaudon. 2005. Chemical constituents from Croton

insularis. Helvetica Chimica Acta 88: 2654–2660.

Hirano, T., K. Yasukawa, H. Harada, T. Taga, Y. Watanabe, T.

Matsuda, S. Kashiwamura, K. Nakajima, K. Koyama, A.

Iwamatsu, S. Tsunsawa, F. Sakiyama, H. Matsui, Y. Takahara,

T. Taniguchi, and T. Kishimoto. 1986. Complementary DNA for

a novel human interleukin (BSF-2) that induced lymphocytes to

produce immunoglobulin. Nature 324: 73–76.

Hirano, T., S. Akira, T. Taga, and T. Kishimoto. 1990. Biological and

clinical aspects of interleukin 6. Immunology Today 11:

443–449.

Ida, Y., Y. Satoh, M. Ohtsuka, M. Nagasao, and J. Shoji. 1994.

Phenolic constituents of Phellodendron amurense bark. Phyto-

chemistry 35: 209–215.

Ishimaru, K., G.-I. Nonaka, and I. Nishioka. 1987. Phenolic

glucosides gallates from Quercus mongolica and Q. acutissima.

Phytochemistry 26: 1147–1152.

Jin, Q., H.-G. Jin, A.R. Kim, and E.-R. Woo. 2012. A new

megastigmane palmitate and a new oleanane triterpenoid from

Aster yomena Makino. Helvetica Chimica Acta 95: 1455–1460.

Jung, H.-J., C.O. Lee, K.-T. Lee, J. Choi, and H.-J. Park. 2004.

Structure-activity relationship of oleanane disaccharides isolated

from Akebia quinata versus cytotoxicity against cancer cells and

NO inhibition. Biological & Pharmaceutical Bulletin 27:

744–747.

Kim, B.H., E.Y. Chung, J.-C. Ryu, S.-H. Jung, K.R. Min, and Y. Kim.

2003. Anti-inflammatory mode of isoflavone glycosides sopho-

ricoside by inhibition of interleukin-6 and cyclooxygenase-2 in

inflammatory response. Archives of Pharmacal Research 26:

306–311.

Kim, M.R., H.-I. Moon, J.H. Chung, Y.H. Moon, K.-S. Hahm, and E.-

R. Woo. 2004. Matrix Metalloproteinase-1 inhibitor from the

stem bark of Styrax japonica S. et Z. Chemical & Pharmaceu-

tical Bulletin 52: 1466–1469.

Lee, I.K., K.H. Kim, S.Y. Lee, S.U. Choi, and K.R. Lee. 2011. Three

new megastigmane glucopyranosides from the Cardamine

komarovii. Chemical & Pharmaceutical Bulletin 59: 773–779.

Lee, S.Y., I.K. Lee, S.U. Choi, and K.R. Lee. 2012. A new

megastigmane glucoside from the aerial parts of Erythronium

japonicum. Natural Product Sciences 18: 166–170.

Lee, T.B. 2003. Illustrated flora of Korea. Seoul: Hyang-Moon

Publishing.

Liu, Q.H., J.-E. Jeong, E.J. Choi, Y.H. Moon, and E.-R. Woo. 2006. A

new furofuran lignan from Geranium thunbergii. Archives of

Pharmacal Research 29: 1109–1113.

Mimaki, Y., M. Kuroda, A. Yokosuka, H. Hiroshi, M. Fukushima,

and Y. Sashida. 2003. Triterpenes and triterpene saponins from

the stems of Akebia trifoliata. Chemical & Pharmaceutical

Bulletin 51: 960–965.

Mimaki, Y., S. Doi, M. Kuroda, and A. Yokosuka. 2007. Triterpene

glycosides from the stems of Akebia quinqta. Chemical &

Pharmaceutical Bulletin 55: 1319–1324.

Nishimura, H., G.-I. Nonaka, and I. Nishioka. 1984. Seven quinic acid

gallates from Quercus stenophylla. Phytochemistry 23:

2621–2623.

Otsuka, H., M. Yao, K. Kamada, and Y. Takeda. 1995. Alangionoside

G-M: Glycosides of megastigmane derivatives from the leaves of

Alangium premnifolium. Chemical & Pharmaceutical Bulletin

43: 754–759.

Otsuka, H., Y. Takeda, K. Yamasaki, and Y. Takeda. 1992. Structural

elucidation of dendranthemosides A and B: Two new b-ionone

glucosides from Dendranthema shiwogiku. Planta Medica 58:

373–375.

Shimomura, H., Y. Sashida, M. Oohara, and H. Tenma. 1988.

Phenolic glucosides from Parabenzoin praecox. Phytochemistry

27: 644–646.

Teng, R.-W., Z.-H. Zhou, D.-Z. Wang, and C.-R. Yang. 2002. Four

caffeoylquinic acids from Morina nepalensis var. alba Hand.-

Mazz. Chinese Journal of Magnetic Resonance 19: 167–174.

Yamano, Y., and M. Ito. 2005. Synthesis of optically active

vomifoliol and roseoside stereoisomers. Chemical & Pharma-

ceutical Bulletin 53: 541–546.

Van Damme, J., G. Opdenakker, R.J. Simpson, M.R. Rubira, S.

Cayphas, A. Vink, A. Billiau, and J. Van Snick. 1987.

Identification of the human 26-kD protein, interferon beta 2

(IFN-beta 2), as a B cell hybridoma/plasmacytoma growth factor

induced in interleukin 1 and tumor necrosis factor. Journal of

Experimental Medicine 165: 914–919.

Yajima, A., Y. Oono, R. Nakagawa, T. Nukada, and G. Yabuta. 2009.

A simple synthesis of four stereoisomers of roseoside and their

inhibitory activity on leukotriene release from mice bone

marrow-derived cultured mast cells. Bioorganic & Medicinal

Chemistry 17: 189–194.

Megastigmane glycoside from Akebia quinata

123