28.01.2013 Views

Preventive effects of chebulic acid isolated from Terminalia chebula ...

Preventive effects of chebulic acid isolated from Terminalia chebula ...

Preventive effects of chebulic acid isolated from Terminalia chebula ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

Journal <strong>of</strong> Ethnopharmacology 131 (2010) 567–574<br />

Contents lists available at ScienceDirect<br />

Journal <strong>of</strong> Ethnopharmacology<br />

journal homepage: www.elsevier.com/locate/jethpharm<br />

<strong>Preventive</strong> <strong>effects</strong> <strong>of</strong> <strong>chebulic</strong> <strong>acid</strong> <strong>isolated</strong> <strong>from</strong> <strong>Terminalia</strong> <strong>chebula</strong> on advanced<br />

glycation endproduct-induced endothelial cell dysfunction<br />

Hyun-Sun Lee a , Yoon-Chang Koo d , Hyung Joo Suh b , Kyung-Yong Kim c , Kwang-Won Lee d,∗<br />

a Institute <strong>of</strong> Health Science, College <strong>of</strong> Health Science, Korea University, Seoul 136-703, Republic <strong>of</strong> Korea<br />

b Department <strong>of</strong> Food and Nutrition, College <strong>of</strong> Health Science, Korea University, Seoul 136-703, Republic <strong>of</strong> Korea<br />

c Department <strong>of</strong> Anatomy, College <strong>of</strong> Medicine, Chung-Ang University, Seoul 156-756, Republic <strong>of</strong> Korea<br />

d Division <strong>of</strong> Food Bioscience & Technology, College <strong>of</strong> Life Science and Biotechnology, Korea University, 5-ga, Anam-dong, Sungbuk-ku, Seoul 136-701, Republic <strong>of</strong> Korea<br />

article info<br />

Article history:<br />

Received 15 April 2010<br />

Received in revised form 15 July 2010<br />

Accepted 16 July 2010<br />

Available online 24 July 2010<br />

Keywords:<br />

Terminaila <strong>chebula</strong><br />

Chebulic <strong>acid</strong><br />

Diabetic complications<br />

Advanced glycation endproducts<br />

Endothelial dysfunction<br />

1. Introduction<br />

abstract<br />

The dried ripe fruits <strong>of</strong> <strong>Terminalia</strong> <strong>chebula</strong> Retzius (T. <strong>chebula</strong><br />

Retz.) (Combretaceae), which is a plant native plant to India and<br />

Southeast Asia, are commonly known as black Myroblans in English<br />

and Harad in Hindi (Rao and Nammi, 2006), and have traditionally<br />

been used as a popular folk medicine for alternative, astringement,<br />

denrifrice, purgative, stomachic, tonic, antiseptic, cardiotonic and<br />

laxative purposes. This fruit is also useful for burns, digestive disorders,<br />

diabetes, eye diseases, weak eye sight, fever, skin diseases<br />

and kidney dysfunction along with other herbs (Saleh et al., 1952;<br />

Rao and Nammi, 2006). <strong>Terminalia</strong> <strong>chebula</strong> has exhibited in vitro<br />

antioxidant and free radical scavenging activities (Naik et al., 2005).<br />

In addition, its antimicrobial (Sato et al., 1997), antiviral (Badmaev<br />

and Nowakowski, 2000), anticancer (Saleem et al., 2002), antianaphylaxis<br />

(Shin et al., 2001) and anti-diabetic (Sabu and Kuttan,<br />

∗ Corresponding author. Tel.: +82 2 3290 3027; fax: +82 2 953 0737.<br />

E-mail address: kwangwon@korea.ac.kr (K.-W. Lee).<br />

0378-8741/$ – see front matter © 2010 Elsevier Ireland Ltd. All rights reserved.<br />

doi:10.1016/j.jep.2010.07.039<br />

Aim <strong>of</strong> the study: The aqueous extract <strong>of</strong> <strong>Terminalia</strong> <strong>chebula</strong>r fruits was reported to have antihyperglycemia<br />

and anti-diabetic complication <strong>effects</strong>. The present study therefore investigated the<br />

protective mechanism <strong>of</strong> <strong>chebulic</strong> <strong>acid</strong>, a phenolcarboxylic <strong>acid</strong> compound <strong>isolated</strong> <strong>from</strong> the ripe fruits <strong>of</strong><br />

<strong>Terminalia</strong> <strong>chebula</strong> against advanced glycation endproducts (AGEs)-induced endothelial cell dysfunction.<br />

Materials and methods: To investigate the protective mechanism <strong>of</strong> <strong>chebulic</strong> <strong>acid</strong> against vascular endothelial<br />

dysfunction human umbilical vein endothelial cells (HUVEC) were treated with <strong>chebulic</strong> <strong>acid</strong> in the<br />

presence/absence <strong>of</strong> glyceraldehyde-related AGEs (glycer-AGEs).<br />

Results: HUVEC incubated with 100 �g/ml <strong>of</strong> glycer-AGEs had significantly enhanced reactive oxygen<br />

species formation, whereas the treatment <strong>of</strong> <strong>chebulic</strong> <strong>acid</strong> dose-dependently reduced glycer-AGEinduced<br />

formation to 108.2 ± 1.9% for 25 �M versus 137.8 ± 1.1% for glycer-AGEs treated alone. The<br />

transendothelial electrical resistance (TER) value <strong>of</strong> the glycer-AGEs group was dramatically decreased to<br />

76.9 ± 2.2% compared to the control, whereas <strong>chebulic</strong> <strong>acid</strong> treatment prevented glycer-AGE-induced TER<br />

change with a value <strong>of</strong> 91.3 ± 5.3%. The incubation <strong>of</strong> confluent HUVEC with 100 �g/ml <strong>of</strong> glycer-AGEs for<br />

24 h remarkably increased the adhesion <strong>of</strong> human monocytic THP-1 cells compared to non-stimulated<br />

HUVEC. These increases in HUVEC adhesiveness were dose-dependently reduced by <strong>chebulic</strong> <strong>acid</strong>.<br />

Conclusions: The present study shows the <strong>effects</strong> <strong>of</strong> <strong>chebulic</strong> <strong>acid</strong> against the progression <strong>of</strong> AGE-induced<br />

endothelial cell dysfunction suggesting that this compound may constitute a promising intervention<br />

agent against diabetic vascular complications.<br />

© 2010 Elsevier Ireland Ltd. All rights reserved.<br />

2002; Rao and Nammi, 2006; Murali et al., 2007) activities have<br />

been reported.<br />

The majority <strong>of</strong> diabetic complications are related to pathophysiological<br />

alterations in the vasculature, with macro- and<br />

microvascular disease being the most common cause <strong>of</strong> morbidity<br />

and mortality in patients with diabetes mellitus (Calcutt et<br />

al., 2009). Clinical and animal model data indicate that chronic<br />

hyperglycemia appears to be the central initiating factor for all<br />

types <strong>of</strong> diabetic microvascular disease (Zannad, 2008; Calcutt et<br />

al., 2009). Diabetes is also associated with accelerated atherosclerotic<br />

macrovascular disease affecting arteries that supply the<br />

heart, brain and lower extremities. As a result, patients with diabetes<br />

have a much higher risk <strong>of</strong> myocardial infarction, stroke<br />

and limb amputation (Laakso and Kuusisto, 1996). A hypothesis<br />

for the initial lesion <strong>of</strong> macrovascular disease such as<br />

atherosclerosis is endothelial dysfunction, defined pragmatically<br />

as changes in the concentration <strong>of</strong> chemical messengers produced<br />

by the endothelial cells and/or by blunting <strong>of</strong> the nitric<br />

oxide-dependent vasodilatory response to hyperemia (Cohen,<br />

2007).


568 H.-S. Lee et al. / Journal <strong>of</strong> Ethnopharmacology 131 (2010) 567–574<br />

Advanced glycation endproducts (AGEs) accumulate in the vessel<br />

wall, where they may perturb cell structure and function. AGEs<br />

have been implicated in both the microvascular and macrovascular<br />

complications <strong>of</strong> diabetes (Brownlee, 1995). The broad consequences<br />

<strong>of</strong> the receptor <strong>of</strong> AGEs (RAGE)–ligand interaction for<br />

cellular properties are emphasized by a spectrum <strong>of</strong> signaling<br />

mechanisms. One such consequence <strong>of</strong> this interaction can lead<br />

to the generation <strong>of</strong> reactive oxygen species (ROS). Other consequences<br />

include the production <strong>of</strong> growth factors and cytokines,<br />

chronic inflammatory responses, and cellular and vascular dysfunction<br />

associated with diabetic complications. The adhesion<br />

<strong>of</strong> leukocytes to the activated endothelium, their migration into<br />

the vessel wall, subsequent infiltration, and differentiation into<br />

macrophages are key events in the development <strong>of</strong> atherosclerotic<br />

cardiovascular disease, which is the leading cause <strong>of</strong> premature<br />

death in patients with diabetes (Renier et al., 2003).<br />

Murali et al. (2007) reported that oral administration <strong>of</strong> <strong>Terminalia</strong><br />

<strong>chebula</strong> extract once daily for two months reduced elevated<br />

blood glucose, and significantly reduced increases in glycosylated<br />

hemoglobin (HbA 1c). Shaila et al. (1998) also reported that the<br />

aortas <strong>of</strong> <strong>Terminalia</strong> <strong>chebula</strong> treated rabbits showed decreased<br />

athermatous plaque formation compared to control group. In previous<br />

studies, we identified <strong>chebulic</strong> <strong>acid</strong> <strong>isolated</strong> <strong>from</strong> <strong>Terminalia</strong><br />

<strong>chebula</strong>, and presented data on its antioxidant activity (Lee et al.,<br />

2007). In the present study, the anti-diabetic complication properties<br />

<strong>of</strong> <strong>chebulic</strong> <strong>acid</strong> were evaluated by assaying anti-glycation<br />

activity, ROS scavenging activity, normalized activity, and antiadhesion<br />

activity against AGEs.<br />

2. Materials and methods<br />

2.1. Chemicals<br />

dl-Glyceraldehyde, 2-deoxy-d-ribose, bovine serum albumin<br />

(BSA, low-endotoxin, fatty <strong>acid</strong> free), 2 ′ ,7 ′ -bis-(2-carboxyethyl)-5-<br />

(and-6)-carboxyfluorescein acetoxymethyl ester (BCECF-AM), and<br />

2 ′ ,7 ′ -dichlor<strong>of</strong>luorescein diacetate (DCFH-DA) were obtained <strong>from</strong><br />

Sigma Chemical Co. (St. Louis, MO, USA). An endothelial cell basal<br />

medium-2 (EBM-2) bullet kit was purchased <strong>from</strong> Clonetics (San<br />

Diago, CA, USA), and all other tissue culture reagents were obtained<br />

<strong>from</strong> GIBCO-BRL (Gaithersburg, MD, USA). All reagents were <strong>of</strong> analytical<br />

grade.<br />

2.2. Preparation <strong>of</strong> <strong>chebulic</strong> <strong>acid</strong> <strong>isolated</strong> <strong>from</strong> <strong>Terminalia</strong> <strong>chebula</strong><br />

The dried ripe fruits <strong>of</strong> <strong>Terminalia</strong> <strong>chebula</strong> cultivated in Taiwan<br />

were purchased <strong>from</strong> a local market (Kyungdong Herb-Market,<br />

Seoul, Korea), and were identified by Pr<strong>of</strong>. B.W. Kang (College<br />

<strong>of</strong> Life Sciences and Biotechnology, Korea University). The isolation<br />

procedures for <strong>chebulic</strong> <strong>acid</strong> were followed as described<br />

previously (Lee et al., 2007). Briefly, the dried fruits <strong>of</strong> <strong>Terminalia</strong><br />

<strong>chebula</strong> were extracted twice with 97% ethanol. The extract was<br />

combined, lyophilized, resuspended in H 2O, and then extracted<br />

successively with n-hexane, chlor<strong>of</strong>orm, ethyl acetate (EtOAc), and<br />

n-butanol. The EtOAc-soluble portion was applied to a column<br />

<strong>of</strong> silica gel, and eluted by a gradient with increasing amounts<br />

<strong>of</strong> methanol in ethyl acetate. The active fraction <strong>of</strong> EtOAc and<br />

methanol (70:30, v/v) was further purified by Sephadex TM LH-20<br />

(Amersham Biosciences, Uppsala, Sweden) column chromatography<br />

using methanol as the eluent, followed by PR-�-BondaPak<br />

C18 column (300 mm × 3.9 mm, 10 �m) (Waters, Milford, MA, USA)<br />

chromatography. The purified compound was identified as <strong>chebulic</strong><br />

<strong>acid</strong> (Fig. 1) using NMR spectra, which were interpreted by Pr<strong>of</strong>.<br />

Bong-Sik Yun (Chonbuk National University, Korea).<br />

Fig. 1. Structure <strong>of</strong> <strong>chebulic</strong> <strong>acid</strong> <strong>isolated</strong> <strong>from</strong> <strong>Terminalia</strong> <strong>chebula</strong>.<br />

2.3. Quantitative assay for cross-linking and AGE formation<br />

The quantitative assay for cross-linking was followed as<br />

described previously (Lee et al., 1998; Lehman and Ortwerth, 2001)<br />

with several modifications. Reaction mixtures were prepared containing<br />

10 mM threose, fatty <strong>acid</strong> free bovine serum albumin (BSA;<br />

5 mg/ml), 2.5 �Ci [ 14 C]lysine (1.0 mCi/mmol), 1.0 mM diethylenetriaminepentaacetic<br />

<strong>acid</strong> (DTPA), and 0.1 M phosphate buffer (pH<br />

7.4) and various concentrations <strong>of</strong> <strong>chebulic</strong> <strong>acid</strong> or aminoguanidine<br />

as a positive control. Each reaction mixture was filter sterilized into<br />

a 2.0 ml sterile Nunc tube, capped, and incubated at 37 ◦ C for 4 days.<br />

Aliquots <strong>of</strong> 50 �l were spotted on 2.5 cm × 3 mm filter paper discs<br />

suspended on pins. After 30 s, each filter was placed in a basket<br />

suspended in 300 ml <strong>of</strong> ice-cold 10% trichloroacetic <strong>acid</strong> (TCA) to<br />

precipitate the protein within the paper disc. At the end <strong>of</strong> sampling,<br />

each group <strong>of</strong> discs was washed in a 5% TCA solution with<br />

stirring at room temperature for 15 min, followed by 5% TCA at<br />

70 ◦ C for 15 min and ethanol:ether (2:1) for 15 min to remove the<br />

TCA and water <strong>from</strong> the discs. The discs were dried under an incandescent<br />

lamp, and protein-bound radioactivity was determined in<br />

a liquid scintillation counter (Beckman LS 5000TD, Beckman Instruments<br />

Inc., Canada). Reaction mixtures for AGE formation were<br />

prepared with 5 mg/ml <strong>of</strong> BSA, 10 mM threose, and 1 mM DTPA<br />

in 1.0 ml <strong>of</strong> 0.1 M and incubated at 37 ◦ C for 7 days in an incubator.<br />

Triplicate 100 �l aliquots were removed, and their fluorescence<br />

intensity was measured at an excitation <strong>of</strong> 370 nm and an emission<br />

<strong>of</strong> 440 nm with a spectr<strong>of</strong>luorometer (VICTOR3 TM , PerkinElmer,<br />

USA). The percentage <strong>of</strong> inhibition <strong>of</strong> glycation was calculated by<br />

the following equation (Choi et al., 2008):<br />

Inhibition (%)<br />

�<br />

�<br />

result <strong>of</strong> sample mixture − result <strong>of</strong> no sugar mixture<br />

= 1 −<br />

result <strong>of</strong> glycation mixture − result <strong>of</strong> no sugar mixture<br />

×100<br />

The calculated IC 50 values denote the concentration <strong>of</strong> sample<br />

required to inhibition 50% <strong>of</strong> glycation.<br />

2.4. Preparation <strong>of</strong> glyceraldehyde-induced AGEs (glycer-AGEs)<br />

Glycer-AGEs were prepared as described previously (Takeuchi<br />

et al., 2000). BSA was incubated with 0.1 M dl-glyceraldehyde,<br />

10 mg/ml BSA, and 5 mM DTPA in 0.2 M phosphate buffer (pH 7.4)<br />

in sterile conditions in the dark at 37 ◦ C for 7 days. Before incubation,<br />

the solution was sterile filtered by passing it through a 0.2 �m<br />

filter. Unmodified BSA was treated under the same conditions without<br />

glyceraldehyde as a control. At the end <strong>of</strong> the incubation period,<br />

unincorporated glyceraldehyde and low molecular reactants were<br />

removed by dialysis against 0.1 M phosphate buffer. We chose a<br />

concentration <strong>of</strong> 100 �g/ml <strong>of</strong> glycer-AGEs to treat the cultured<br />

cells as described previously (Yamagishi et al., 2002).


Table 1<br />

Anti-glycation activity <strong>of</strong> <strong>chebulic</strong> <strong>acid</strong>.<br />

50% <strong>of</strong> inhibitory<br />

concentration on<br />

cross-linking (mM) a<br />

H.-S. Lee et al. / Journal <strong>of</strong> Ethnopharmacology 131 (2010) 567–574 569<br />

50% <strong>of</strong> inhibitory<br />

concentration on<br />

formation <strong>of</strong> AGEs (mM) b<br />

Chebulic <strong>acid</strong> 17.1 ± 0.7 1.32 ± 0.10<br />

Aminoguanidine 21.3 ± 1.4 2.37 ± 0.09<br />

a The effect <strong>of</strong> inhibitors on the cross-linking <strong>of</strong> [ 14 C]lysine to BSA proteins. Reaction<br />

mixtures contained 5.0 mg/ml <strong>of</strong> BSA, 5.0 mM l-threose, 2.5 �Ci [ 14 C]lysine,<br />

10 mM inhibitor, and 1.0 mM DTPA in 0.1 M phosphate buffer, pH 7.0, and were<br />

incubated at 37 ◦ C for 4 days. Protein-bound radioactivity was determined in a liquid<br />

scintillation counter.<br />

b The <strong>effects</strong> <strong>of</strong> inhibitors on the formation <strong>of</strong> AGEs. The reaction mixtures for<br />

AGE formation contained 5.0 mg/ml <strong>of</strong> BSA, 5.0 mM l-threose, 10 mM inhibitor, and<br />

1.0 mM DTPA in 0.1 M phosphate buffer, pH 7.0, and were incubated at 37 ◦ C for 7<br />

days. The intensity <strong>of</strong> AGE formation was measured at an excitation <strong>of</strong> 370 nm and<br />

an emission <strong>of</strong> 440 nm with a spectr<strong>of</strong>luorometer.<br />

Table 2<br />

Cytotoxicity <strong>of</strong> glycer-induced AGEs on HUVEC.<br />

Concentration (�g/ml) BSA Glycer-AGEs<br />

0 100.0 ± 2.0 a 100.0 ± 2.7 a<br />

100 98.3 ± 2.5 a 84.5 ± 1.3 b<br />

250 99.4 ± 2.3 a 79.8 ± 1.7 c<br />

500 99.0 ± 4.3 a 78.9 ± 3.4 c<br />

750 102.7 ± 5.1 a 82.7 ± 0.6 b<br />

1000 103.9 ± 1.9 a 85.3 ± 1.4 b<br />

Glycer-induced AGEs were prepared by incubating BSA (10 mg/ml) with 0.1 M glyceraldehyde<br />

for 7 days. HUVEC incubated with various concentrations <strong>of</strong> glycer-AGEs<br />

or BSA for 24 h. Cell viability was determined by the MTT assay. Results were<br />

expressed as means ± S.D. (n = 3 or more). Means with different superscript letters<br />

are significantly different at p < 0.05 by Duncan’s multiple range tests.<br />

Table 3<br />

Cytotoxicity <strong>of</strong> <strong>chebulic</strong> <strong>acid</strong> with or without glyceraldehyde-induced AGEs on<br />

HUVEC.<br />

Chebulic <strong>acid</strong><br />

levels (�M)<br />

Chebulic <strong>acid</strong> Chebulic <strong>acid</strong> with<br />

100 �g/ml <strong>of</strong> glycer-AGEs<br />

0 100.0 ± 2.2 a 94.4 ± 1.3 c<br />

1 101.0 ± 0.5 a 95.9 ± 1.2 b<br />

5 100.0 ± 2.0 a 97.4 ± 0.7 b<br />

10 100.1 ± 2.8 a 99.4 ± 1.2 a<br />

25 101.0 ± 2.3 a 101.4 ± 1.2 a<br />

50 86.2 ± 4.5 d –<br />

100 69.6 ± 3.3 e –<br />

250 66.6 ± 9.8 e –<br />

Cells were incubated with various concentrations <strong>of</strong> <strong>chebulic</strong> <strong>acid</strong> with/without<br />

100 �g/ml <strong>of</strong> glycer-induced AGEs for 24 h. Cell viability was determined by the<br />

MTT assay. Results are expressed as means ± S.D. (n = 3 or more). Means with different<br />

superscript letters are significantly different at p < 0.05 by Duncan’s multiple<br />

range tests.<br />

2.5. Cell culture<br />

Human umbilical vein endothelial cells (HUVEC) were purchased<br />

<strong>from</strong> Biobud Co. (Seoul, Korea), were grown on 1.5%<br />

gelatin-coated culture dishes or plates in EBM-2, and used for the<br />

experiments within the first 5–6 passages. The cells were supplemented<br />

at 37 ◦ C in a humidified atmosphere <strong>of</strong> 5% CO 2. Cells <strong>of</strong> the<br />

human acute monocytic leukemia THP-1 cell line (ATCC TIB202)<br />

were obtained <strong>from</strong> the American Type Culture Collection (ATCC;<br />

Rockville, MD, USA), and were cultured in RPMI-1640 medium<br />

supplemented with 10% (v/v) heat-inactivated fetal bovine serum<br />

(FBS), 100 U/ml penicillin, and 100 �g/ml streptomycin.<br />

Fig. 2. Intracellular reactive oxygen species (ROS) production by glyceraldehyde-induced AGEs (glycer-AGEs) on human umbilical vein endothelial cells (HUVEC). Dichlor<strong>of</strong>luorescein<br />

diacetate (DCFH-DA)-loaded HUVEC were exposed to BSA (A), 50 �g/ml <strong>of</strong> glycer-AGEs (B), 100 �g/ml <strong>of</strong> glycer-AGEs (C), and 200 �g/ml <strong>of</strong> glycer-AGEs (D)<br />

for 24 h. Data are expressed as a histogram <strong>of</strong> the fluorescence peak channel by flow cytometry. The horizontal axis represents the intensity <strong>of</strong> cellular fluorescence (FL1-H),<br />

the vertical axis represents the number <strong>of</strong> cells detected for a given fluorescent value. Given the fixed intervals <strong>of</strong> FL1-H, the relative cell distributions are indicated for each<br />

treatment.


570 H.-S. Lee et al. / Journal <strong>of</strong> Ethnopharmacology 131 (2010) 567–574<br />

Fig. 3. ROS scavenging activities <strong>of</strong> <strong>chebulic</strong> <strong>acid</strong> against glycer-AGEs. DCFH-DA-loaded HUVEC were exposed to BSA (A), 100 �g/ml <strong>of</strong> glycer-AGEs (B), 100 �g/ml <strong>of</strong><br />

glycer-AGEs + 5 �M <strong>chebulic</strong> <strong>acid</strong> (C), 100 �g/ml <strong>of</strong> glycer-AGEs + 10 �M <strong>chebulic</strong> <strong>acid</strong> (D), and 100 �g/ml <strong>of</strong> glycer-AGEs + 25 �M <strong>chebulic</strong> <strong>acid</strong> (E) for 24 h. Data (A–E) were<br />

expressed as a histogram <strong>of</strong> the fluorescence peak channel by flow cytometry, and their fluorescence intensities were assayed with fluorescence spectrometry (F).<br />

2.6. Cytotoxicity <strong>of</strong> glycer-AGEs and <strong>chebulic</strong> <strong>acid</strong><br />

The cytotoxicities <strong>of</strong> the glycer-AGEs and various concentrations<br />

(1–100 �M) <strong>of</strong> <strong>chebulic</strong> <strong>acid</strong> were analyzed by MTT colorimetric<br />

assays (Mosmann, 1983). The cells were incubated with <strong>chebulic</strong><br />

<strong>acid</strong> with/without 100 �g/ml <strong>of</strong> glycer-AGEs for 24 h. At the end <strong>of</strong><br />

the treatment time, all the wells were aspirated, refilled with MTT<br />

solution, and incubated for 3 h (R<strong>of</strong>fey et al., 2007).<br />

2.7. Detection <strong>of</strong> ROS in HUVEC<br />

The production <strong>of</strong> intracellular ROS was determined by DCFH-<br />

DA by fluorescence spectrophotometry (Shukla et al., 2006), and<br />

flow cytometry. DCFH-DA was dissolved in dimethyl sulfoxide<br />

(DMSO) as a 10 mM stock solution, and kept frozen at −20 ◦ C. To<br />

load the cells, DCFH-DA <strong>from</strong> the stock solution was mixed with<br />

M199 medium. Next, 20 �M DCFH-DA-loaded HUVEC were incubated<br />

for 30 min, washed twice with M199, and exposed to various<br />

concentrations <strong>of</strong> <strong>chebulic</strong> <strong>acid</strong> and/or 100 �g/ml <strong>of</strong> glycer-AGEs.<br />

After 24 h, the cells were harvested by trypsinization, and washed<br />

with cold phosphate buffered saline (PBS) twice. ROS were measured<br />

by flow cytometry (FACS Calibur, Becton Dickinson, NJ, USA)<br />

at a level <strong>of</strong> 10,000 events for each test. Fluorescence was obtained<br />

by a histogram <strong>of</strong> the peak channel using the CellQuest program<br />

(Becton Dickinson), and the fluorescence intensity was determined<br />

with excitation <strong>of</strong> 485 nm and emission <strong>of</strong> 530 nm using a plate<br />

reader (VICTOR3 TM , PerkinElmer, MA, USA).<br />

2.8. Assessment <strong>of</strong> transendothelial electrical resistance (TER)<br />

The <strong>effects</strong> <strong>of</strong> glycer-AGEs on TER were measured in HUVEC<br />

according to the method <strong>of</strong> Kazak<strong>of</strong>f et al. (1995). The HUVEC were<br />

seeded at 3 × 10 5 cells per inset on a type 1 rat tail collagen coated<br />

membrane (pore size, 0.4 �m) contained in the apical chamber <strong>of</strong><br />

a 12-well trans-well system (Corning, NY, USA). After monolayer<br />

confluence was achieved (>120 cm 2 ), the HUVEC were treated<br />

with <strong>chebulic</strong> <strong>acid</strong> and glycer-AGEs. TER was measured every 4 h<br />

over 24 h. The measurements were taken in triplicate, and the data<br />

were expressed as relative percentages.<br />

2.9. Adhesion assay <strong>of</strong> THP-1<br />

The monocyte adhesion assay was performed as described previously<br />

(Hiraoka et al., 2004). Briefly, THP-1 cells were prepared


y combining the cells with BCECF-AM, and then the cells were<br />

washed three times with 1% FBS in PBS. The HUVEC grown to confluence<br />

in a 24-well plate were pretreated with <strong>chebulic</strong> <strong>acid</strong> and<br />

glycer-AGEs at 37 ◦ C for 24 h for the adhesion assay. The BCECF-AM<br />

labeled THP-1 cells were co-incubated with the HUVEC for 60 min at<br />

37 ◦ C. After incubation, nonadherent cells were removed by washing<br />

each well three times with 1% FBS in PBS. The attached cells<br />

were examined via confocal microscopy (LSM 510; Carl Zeiss Micro-<br />

Imaging, Inc., NY, USA). The fluorescence intensity <strong>of</strong> each well<br />

was measured using a fluorescence multiwell plate reader (Wallac<br />

1420, VICTOR3 TM ) at excitation and emission wave lengths <strong>of</strong><br />

485 and 530 nm, respectively.<br />

2.10. Statistical analysis<br />

All statistical analyses were performed using the Statistical<br />

Package for Social Sciences (SPSS) version 12.0 (SPSS Inc., IL, USA).<br />

The differences among groups were evaluated by one-way analysis<br />

<strong>of</strong> variance (ANOVA) and Duncan’s multiple range tests. All data<br />

were reported as means ± standard deviation (S.D.).<br />

3. Results<br />

3.1. Anti-glycation activity <strong>of</strong> <strong>chebulic</strong> <strong>acid</strong><br />

Table 1 shows that <strong>chebulic</strong> <strong>acid</strong> had IC 50 values <strong>of</strong> 17.1 mM for<br />

protein cross-linking and 1.32 mM for AGE formation. As a positive<br />

control, aminoguanidine had IC 50 values <strong>of</strong> 21.3 mM and 2.37 mM,<br />

respectively. However, based on these millimolar levels <strong>of</strong> <strong>chebulic</strong><br />

<strong>acid</strong>, it seemed to have very low potency regarding with inhibitory<br />

activities on glycation-induced protein cross-linking and AGE formation.<br />

3.2. Effects on the formation <strong>of</strong> AGEs and toxicity<br />

The <strong>effects</strong> <strong>of</strong> the AGEs and toxicity were assessed using various<br />

sugars such as ribose (a pentose), glyceraldehydes, glycoaldehyde<br />

(a short chain sugar), and glyoxal (a dicarbonyl compound) for the<br />

glycation <strong>of</strong> HUVEC. Ribose and glyoxal produced few AGEs, and<br />

did not induce cell death at the tested concentrations <strong>of</strong> 10–50 mM<br />

(data not shown). Glycoaldehyde and glyceraldehydes caused<br />

more potent cytotoxicity in a concentration-dependent manner.<br />

Overall, glyceraldehyde caused the fastest rate <strong>of</strong> AGE formation<br />

in a concentration-dependent manner in the HUVEC (data not<br />

shown).<br />

3.3. Glyer-AGEs cytotoxicity and ROS formation<br />

As shown in Table 2, glycer-AGEs caused significant cytotoxicity<br />

in a dose-dependent manner up to 500 �g/ml <strong>of</strong> glycer-AGEs. These<br />

levels <strong>of</strong> glycer-AGEs were used for intracellular ROS generation,<br />

and its ROS occurred in a dose-dependent manner (Fig. 2). In previous<br />

in vitro studies (Twigg et al., 2002; Ohashi et al., 2004; Xu et al.,<br />

2004), the concentration <strong>of</strong> AGEs was used mostly at 50–200 �g/ml.<br />

Therefore, based on these results, the concentration <strong>of</strong> glycer-AGEs<br />

at 100 �g/ml was used in our next studies.<br />

3.4. Cytotoxicity by <strong>chebulic</strong> <strong>acid</strong> with or without Glyer-AGEs<br />

Chebulic <strong>acid</strong> had slight toxic <strong>effects</strong> on the HUVEC above a concentration<br />

<strong>of</strong> 50 �M. Therefore, 25 �M <strong>chebulic</strong> <strong>acid</strong> was used for<br />

the subsequent experiments (Table 3). In addition, the results <strong>from</strong><br />

Tables 2 and 3 suggest that the apparent cytotoxic effect <strong>of</strong> <strong>chebulic</strong><br />

<strong>acid</strong> may reflect the variability <strong>of</strong> the effect <strong>of</strong> glycer-AGEs.<br />

H.-S. Lee et al. / Journal <strong>of</strong> Ethnopharmacology 131 (2010) 567–574 571<br />

Fig. 4. Changes in permeability by glycer-AGEs and <strong>chebulic</strong> <strong>acid</strong>. HUVEC were<br />

seeded at 3 × 10 5 cells per inset on a type 1 rat tail collagen coated membrane<br />

contained in the apical chamber <strong>of</strong> a 12-well trans-well system. After monolayer<br />

confluence was achieved (>120 cm 2 ), the HUVEC were treated with <strong>chebulic</strong> <strong>acid</strong><br />

and/or glycer-AGEs. Changes in transendothelial electric resistance (TER) were measured<br />

every 4 h over 24 h. Measurements were taken in triplicate and the data are<br />

expressed as relative percentages. *Significant difference compared to glycer-ACEs<br />

only treated group at p < 0.05.<br />

3.5. Intracellular ROS scavenging activities<br />

Intracellular ROS was estimated by flow cytometry using DCFH<br />

as a probe. Levels <strong>of</strong> ROS were low in the unstimulated HUVEC<br />

(Fig. 3(A)), and the HUVEC treated with 100 �g/ml <strong>of</strong> glycer-<br />

AGEs (Fig. 3(B)) significantly increased ROS formation. As shown<br />

in Fig. 3F, <strong>chebulic</strong> <strong>acid</strong> dose-dependently prevented glycer-<br />

AGE-induced ROS generation, which was significantly reduced to<br />

122.0 ± 3.9% for 5 �M, to 120.0 ± 3.3% for 10 �M, and to 108.2 ± 1.9%<br />

for 25 �M versus 137.8 ± 1.1% for the control treated with glycer-<br />

AGEs alone (p < 0.01 versus glycer-AGEs alone).<br />

3.6. <strong>Preventive</strong> <strong>effects</strong> on TER against AGEs<br />

As shown in Fig. 4, the control showed no significant<br />

changes in TER during 24 h. But the TER value <strong>of</strong> the glycer-<br />

AGEs group dramatically decreased in 4 h, and remained at<br />

98.0 ± 2.8 cm 2 (76.9 ± 2.2%) after 24 h as compared to the TER<br />

value <strong>of</strong> the control group (116.0 ± 2.8 cm 2 , 97.9 ± 2.4%). However,<br />

the treatment <strong>of</strong> <strong>chebulic</strong> <strong>acid</strong> dose-dependently recovered<br />

TER to 114.7 ± 8.7 cm 2 (91.3 ± 5.3%).<br />

3.7. Reduction <strong>of</strong> adhesion THP-1 cells on HUVEC<br />

The incubation <strong>of</strong> confluent HUVEC with 100 �g/ml <strong>of</strong> glycer-<br />

AGEs for 24 h caused a significant increase in the adhesion <strong>of</strong> THP-1<br />

monocytic cells compared to non-stimulated HUVEC (Fig. 5A versus<br />

Fig. 5B). This increased cell adhesion was dose-dependently<br />

reduced by <strong>chebulic</strong> <strong>acid</strong> treatments (Fig. 5C and D). As shown<br />

in Fig. 5E, <strong>chebulic</strong> <strong>acid</strong> reduced glycer-AGE-induced fluorescence<br />

intensity to 23600 ± 6600 for 5 �M, 22600 ± 36000 for 10 �M, and<br />

to 134000 ± 4100 �M for 25 �M versus 269000 ± 37000 �M for the<br />

control treated with glycer-AGEs alone.<br />

4. Discussion<br />

Diabetic patients have a 2- to 4-fold increased risk <strong>of</strong> subsequently<br />

developing microvascular complications such as renal,<br />

neuronal and retinal diseases, as well as macrovascular problems<br />

such as atherosclerosis and coronary heart disease (Calcutt et al.,<br />

2009). Unfortunately, these complications may develop in both


572 H.-S. Lee et al. / Journal <strong>of</strong> Ethnopharmacology 131 (2010) 567–574<br />

Fig. 5. Effects <strong>of</strong> <strong>chebulic</strong> <strong>acid</strong> on THP-1 cell adhesion to HUVEC stimulated by glycer-AGEs. HUVEC treated with BSA (A), 100 �g/ml <strong>of</strong> glycer-AGEs (B), 100 �g/ml <strong>of</strong><br />

glycer-AGEs + 10 �M <strong>chebulic</strong> <strong>acid</strong> (C), and 100 �g/ml <strong>of</strong> glycer-AGEs + 25 �M <strong>chebulic</strong> (D) for 24 h to the adhesion assay. After stimulation, the HUVEC were incubated with<br />

BCECF-AM-labeled THP-1. The adherent cells were collected by treatment with 0.1% SDS and measured using a fluorescence multiwell plate reader (Wallac 1420, Germany)<br />

at excitation and emission wave lengths <strong>of</strong> 485 and 530 nm, respectively, and the fluorescence intensity was expressed as a histogram (E). Shared letters represent means<br />

that were not significantly different (p < 0.05).<br />

type 1 and type 2 diabetic patients even with careful control <strong>of</strong><br />

their glycemic level, blood pressure, and lipid pr<strong>of</strong>ile, and such vascular<br />

diseases are an important cause <strong>of</strong> morbidity and mortality in<br />

diabetic patients (Laakso and Kuusisto, 1996; Cohen, 2007; Calcutt<br />

et al., 2009). The authors <strong>of</strong> one particular diabetes complications<br />

and control trial: The Epidemiology <strong>of</strong> Diabetes Interventions and<br />

Complications Trial, suggested that the influence <strong>of</strong> early glucose<br />

control on the development <strong>of</strong> macrovascular events may become<br />

more obvious with a longer follow-up time (The Diabetes Control<br />

and Complications Trial/Epidemiology <strong>of</strong> Diabetes Interventions<br />

and Complications Research Group, 1993, 2005; Writing Team<br />

for the Diabetes Control and Complications Trial/Epidemiology <strong>of</strong><br />

Diabetes Interventions and Complications Research Group, 2003).<br />

These findings suggest that advanced protein glycation, a pro-


cess involving the nonenzymatic modification <strong>of</strong> tissue proteins<br />

by physiological sugars in vivo, appears to play a central role in<br />

the pathogenesis <strong>of</strong> diabetic complications; this is because the<br />

concept <strong>of</strong> ‘glycemic memory’ is well-fitting to the in vivo formation<br />

and accumulation <strong>of</strong> AGEs and their roles (Brownlee, 1995).<br />

In addition, one must take into consideration that AGE formation<br />

can arise not only with glucose, but also with short chain sugars<br />

such as glycolaldehyde, and dicarbonyl compounds such as methylglyoxal<br />

derived <strong>from</strong> sugar autoxidation and other metabolic<br />

pathways (Takeuchi and Yamagishi, 2004). Inside cells, these can<br />

much readily accelerate AGE formation with amino groups <strong>of</strong> proteins<br />

as compared to glucose alone (Takeuchi et al., 2000). Thus,<br />

controlling AGE formation as well as blood glucose levels are essential<br />

aspects in diabetes treatment for the prevention <strong>of</strong> diabetic<br />

complications.<br />

AGEs mediate their <strong>effects</strong> directly and via their engagement<br />

with receptors for AGEs (RAGE). It was reported that AGE crosslinking<br />

to collagen can decrease vascular elasticity, and increases<br />

vascular stiffness in the vessel wall, as well as glomerulosclerosis<br />

in the kidney and vascular hyperpermeability in the retina<br />

(Wendt et al., 2003). The binding <strong>of</strong> AGEs to RAGE results in<br />

intracellulatr ROS generation, which seems to be linked to the<br />

NAD(P)H-oxidase system (Wautier et al., 2001), along with subsequent<br />

activation <strong>of</strong> the redox-sensitive transcription factor nuclear<br />

factor-kB in vascular wall cells and the expression <strong>of</strong> a variety<br />

<strong>of</strong> athrosclerosis-related genes, including intercellular adhesion<br />

molecule-1 (ICAM-1), vsasular cell adhesion molecule-1 (VCAM-<br />

1), monocyte chemoattractant protein-1 (MCP-1), plasminogen<br />

activator inhibitor-1 (PAI-1), vascular endothelial growth factor<br />

(VGEF) and RAGE (Basta, 2008; Yamagishi et al., 2008). As a<br />

candidate inhibitor for AGE formation, aminoguanidine, a nucleophilic<br />

hydrazine compound that confines reactive carbonyls<br />

during glycation reactions thus inhibiting the conversion <strong>of</strong> these<br />

intermediates into AGEs, was reported to enhance vascular elasticity<br />

and decrease vascular permeability in diabetic rats (Huijberts<br />

et al., 1993). However, clinical trials for aminoguanidine were<br />

stopped due to reported side <strong>effects</strong> such as gastrointestinal disturbance<br />

(Thornalley, 2003). Alternatively, the search for AGE<br />

inhibitors, by examining naturally occurring compounds used<br />

traditionally was started. In this regard, <strong>chebulic</strong> <strong>acid</strong> <strong>isolated</strong><br />

<strong>from</strong> <strong>Terminalia</strong> <strong>chebula</strong>r has been investigated by our research<br />

group (Lee et al., 2007). This compound belongs to the phenolcarboxylic<br />

<strong>acid</strong> group, and its structure is simpler compared<br />

to other compounds such as <strong>chebula</strong>nin, <strong>chebula</strong>gic <strong>acid</strong>, and<br />

casuarinin (Juang et al., 2004). We previously reported that the<br />

antioxidant activity <strong>of</strong> <strong>chebulic</strong> <strong>acid</strong> was evaluated by examining<br />

2,2-diphenyl-1-picrylhydrazyl radical (DPPH • ) scavenging activity,<br />

ferric reducing/antioxidant power (FRAP), and the specific<br />

ESR spectrum (Lee et al., 2007). In the present study, <strong>chebulic</strong><br />

<strong>acid</strong> inhibited glycation-mediated cross-linking more actively<br />

than aminoguanidine (1.2-fold-lower IC 50, 17.1 ± 0.7 mM versus<br />

21.3 ± 1.4 mM; Table 1) based on cross-linking assays, which<br />

were carried out using a system measuring the incorporation <strong>of</strong><br />

[ 14 C]lysine into BSA. Also, <strong>chebulic</strong> <strong>acid</strong>’s IC 50 value (mM) for inhibition<br />

<strong>of</strong> AGE formation was 1.8-fold-lower (1.32 ± 0.10 mM) than<br />

the IC 50 <strong>of</strong> aminoguanidine (2.37 ± 0.09 mM). Whiles such millimolar<br />

concentrations <strong>of</strong> <strong>chebulic</strong> <strong>acid</strong> seem to indicate relatively low<br />

potency with respect to inhibitory <strong>effects</strong> against cross-linking and<br />

AGE formation, effective levels <strong>of</strong> <strong>chebulic</strong> <strong>acid</strong> in this micromolar<br />

range appear to be markedly more potent in reducing AGE-induced<br />

cytotoxicity, AGE-induced ROS formation, AGE-induced reductions<br />

in endothelial cell electrical resistance and AGE-induced monocyte<br />

adhesion (Tables 2 and 3 and Figs. 2–5).<br />

There is accumulating evidence that RAGE interact with AGEs,<br />

which is described by the term ‘AGE-RAGE-oxidative stress axis’,<br />

and induce increased levels <strong>of</strong> intracellular reactive oxygen species<br />

H.-S. Lee et al. / Journal <strong>of</strong> Ethnopharmacology 131 (2010) 567–574 573<br />

(ROS) and subsequently evoke inflammatory responses leading<br />

to the development <strong>of</strong> vascular complications in diabetes (Basta,<br />

2008). This increase in ROS in turn activates a complex cascade<br />

<strong>of</strong> signal transduction pathways, and subsequently enhances the<br />

expression <strong>of</strong> many genes that are highly relevant for inflammation,<br />

immunity, and atherosclerosis, such as cell adhesion<br />

molecules, tissue factors, adhesion molecules (e.g., ICAM-1 and<br />

VCAM-1), cytokines, as well as RAGE (Basta, 2008; Yamagishi<br />

et al., 2008). These up-regulations are associated with endothelial<br />

adherence and the migration <strong>of</strong> leukocytes, which are the<br />

first steps in atherosclerosis, thus priming and sustaining vascular<br />

wall injury and the inflammatory response and causing diabetic<br />

vascular lesions (Jay et al., 2006). Interestingly, our preliminary<br />

experiment (data not shown), which employed streptozotocininduced<br />

diabetic rats, showed that the group orally supplemented<br />

with the ethylacetate-soluble layer <strong>of</strong> ehtanolic extract <strong>of</strong> <strong>Terminalia</strong><br />

<strong>chebula</strong>r containing <strong>chebulic</strong> <strong>acid</strong> had significantly reduced<br />

mRNA levels <strong>of</strong> RAGE and ICAM-1 in the thoracic aorta, which<br />

were increased only in the streptozotocin-treated group. However,<br />

it should be noted that our present in vitro results, which<br />

showed <strong>chebulic</strong> <strong>acid</strong> had activity for normalizing the permeability<br />

<strong>of</strong> endothelial cells and reduced the adhesion <strong>of</strong> monocytes<br />

to endothelial cells treated with AGEs, must be reproduced in<br />

vivo to form the basis for a completely novel anti-diabetic therapy<br />

that not only reduces blood glucose, but also reduces the<br />

action <strong>of</strong> AGEs. At this point, the activities <strong>of</strong> <strong>chebulic</strong> <strong>acid</strong> are<br />

not yet fully understood. However, the present results suggest that<br />

<strong>chebulic</strong> <strong>acid</strong> <strong>from</strong> <strong>Terminalia</strong> <strong>chebula</strong> may act as an anti-diabetic<br />

complication agent through the extracellular inhibition <strong>of</strong> AGE<br />

formation as well as intracellular ROS scavenging in endothelial<br />

cells.<br />

5. Conclusion<br />

Inhibitors <strong>of</strong> AGE formation to reduce tissue accumulation<br />

<strong>of</strong> AGEs in diabetes have potentially beneficial <strong>effects</strong> in innate<br />

chronic atherosclerosis. Alternatively, interference on RAGE-AGE<br />

axis may be a promising novel therapeutic strategy for the treatment<br />

<strong>of</strong> acute vascular injury. Our in vitro results with <strong>chebulic</strong><br />

<strong>acid</strong> <strong>isolated</strong> <strong>from</strong> <strong>Terminalia</strong> <strong>chebula</strong> may constitute a promising<br />

intervention agent in these two setting relevant to diabetic vascular<br />

complications. Further in vivo studies are required to prove<br />

whether <strong>chebulic</strong> <strong>acid</strong> could inhibit the risk <strong>of</strong> diabetic vascular<br />

complications beyond blood glucose lowering effect.<br />

Acknowledgements<br />

This work supported by grant number PJ007100201003 <strong>from</strong><br />

the BioGreen 21 program, Rural Development Administration,<br />

Republic <strong>of</strong> Korea Research Fund. The authors thank the Korea<br />

University-CJ Food Safety Center (Seoul, South Korea) for providing<br />

the equipment and facilities.<br />

Appendix A. Supplementary data<br />

Supplementary data associated with this article can be found, in<br />

the online version, at doi:10.1016/j.jep.2010.07.039.<br />

References<br />

Badmaev, V., Nowakowski, M., 2000. Protection <strong>of</strong> epithelial cells against influenza A<br />

virus by a plant derived biological response modifier Ledretan-96. Phytotherapy<br />

Research 14, 245–249.<br />

Basta, G., 2008. Receptor for advanced glycation endproducts and atherosclerosis:<br />

<strong>from</strong> basic mechanisms to clinical implications. Atherosclerosis 196, 9–21.<br />

Brownlee, M., 1995. Advanced protein glycosylation in diabetes and aging. Annual<br />

Review <strong>of</strong> Medicine 46, 223–234.


574 H.-S. Lee et al. / Journal <strong>of</strong> Ethnopharmacology 131 (2010) 567–574<br />

Calcutt, N.A., Cooper, M.E., Kern, T.S., Schmidt, A.M., 2009. Therapies for<br />

hyperglycaemia-induced diabetic complications: <strong>from</strong> animal models to clinical<br />

trials. Nature Reviews Drug Discovery 8, 417–429.<br />

Choi, S.Y., Jung, S.H., Lee, H.S., Park, K.W., Yun, B.S., Lee, K.W., 2008. Glycation<br />

inhibitory activity and the identification <strong>of</strong> an active compound in Plantago<br />

asiatica extract. Phytotherapy Research 22, 323–329.<br />

Cohen, J.D., 2007. Overview <strong>of</strong> physiology, vascular biology, and mechanisms <strong>of</strong><br />

hypertension. Journal <strong>of</strong> Managed Care Pharmacy 13, S6–S8.<br />

Hiraoka, M, Nitta, N., Nagai, M., Shimokado, K., Yoshida, M., 2004. MCP-1-induced<br />

enhancement <strong>of</strong> THP-1 adhesion to vascular endothelium was modulated<br />

by HMG-CoA reductase inhibitor through RhoA GTPase-, but not ERK1/2dependent<br />

pathway. Life Science 75, 1333–1341.<br />

Huijberts, M.S., Wolffenbuttel, B.H., Boudier, H.A., Crijns, F.R., Kruseman, A.C.,<br />

Poitevin, P., Levy, B.I., 1993. Aminoguanidine treatment increases elasticity and<br />

decreases fluid filtration <strong>of</strong> large arteries <strong>from</strong> diabetic rats. Journal <strong>of</strong> Clinical<br />

Investigation 92, 1407–1411.<br />

Jay, D., Hitomi, H., Griendling, K.K., 2006. Oxidative stress and diabetic cardiovascular<br />

complications. Free Radical Biology and Medicine 40, 183–192.<br />

Juang, L.J., Sheu, S.J., Lin, T.C., 2004. Determination <strong>of</strong> hydrolyzable tannins<br />

in the fruit <strong>of</strong> <strong>Terminalia</strong> <strong>chebula</strong> Retz. by high-performance liquid chromatography<br />

and capillary electrophoresis. Journal <strong>of</strong> Separation Science 27,<br />

718–724.<br />

Kazak<strong>of</strong>f, P.W., McGuire, T.R., Hoie, E.B., Cano, M., Iversen, P.L., 1995. An in vitro<br />

model for endothelial permeability: assessment <strong>of</strong> monolayer integrity. In Vitro<br />

Cellular and Developmental Biology: Animal 31, 846–852.<br />

Laakso, M., Kuusisto, J., 1996. Epidemiological evidence for the association <strong>of</strong><br />

hyperglycaemia and atherosclerotic vascular disease in non-insulin-dependent<br />

diabetes mellitus. Annals <strong>of</strong> Medicine 28, 415–418.<br />

Lee, H.S., Jung, S.H., Yun, B.S., Lee, K.W., 2007. Isolation <strong>of</strong> <strong>chebulic</strong> <strong>acid</strong> <strong>from</strong> <strong>Terminalia</strong><br />

<strong>chebula</strong> Retz. and its antioxidant effect in <strong>isolated</strong> rat hepatocytes. Archives<br />

<strong>of</strong> Toxicology 81, 211–218.<br />

Lee, K.-W., Mossine, V., Ortwerth, B.J., 1998. The relative ability <strong>of</strong> glucose and ascorbate<br />

to glycate and crosslink lens proteins in vitro. Experimental Eye Research<br />

67, 95–104.<br />

Lehman, T.D., Ortwerth, B.J., 2001. Inhibitors <strong>of</strong> advanced glycation end productassociated<br />

protein cross-linking. Biochimica et Biophysica Acta (BBA): Molecular<br />

Basis <strong>of</strong> Disease 1535, 110–119.<br />

Mosmann, T., 1983. Rapid colorimetric assay for cellular growth and survival:<br />

application to proliferation and cytotoxicity assays. Journal <strong>of</strong> Immunological<br />

Methods 65, 55–63.<br />

Murali, Y.K., Anand, P., Tandon, V., Singh, R., Chandra, R., Murthy, P.S., 2007.<br />

Long-term <strong>effects</strong> <strong>of</strong> <strong>Terminalia</strong> <strong>chebula</strong> Retz. on hyperglycemia and associated<br />

hyperlipidemia, tissue glycogen content and in vitro release <strong>of</strong> insulin in streptozotocin<br />

induced diabetic rats. Experimental Clinical Endocrinology & Diabetes<br />

115, 641–646.<br />

Naik, G.H., Priyadarsini, K.I., Bhagirathi, R.G., Mishra, B., Mishra, K.P., Banavalikar,<br />

M.M., Mohan, H., 2005. In vitro antioxidant studies and free radical reactions <strong>of</strong><br />

triphala, an ayurvedic formulation and its constituents. Phytotherapy Research<br />

19, 582–586.<br />

Ohashi, S., Abe, H., Takahashi, T., Yamamoto, Y., Takeuchi, M., Arai, H., Nagata, K., Kita,<br />

T., Okamoto, H., Yamamoto, H., Doi, T., 2004. Advanced glycation end products<br />

increase collagen-specific chaperone protein in mouse diabetic nephropathy.<br />

Journal <strong>of</strong> Biological Chemistry 279, 19816–19823.<br />

Rao, N.K., Nammi, S., 2006. Antidiabetic and renoprotective <strong>effects</strong> <strong>of</strong> the chlor<strong>of</strong>orm<br />

extract <strong>of</strong> <strong>Terminalia</strong> <strong>chebula</strong> Retz. seeds in streptozotocin-induced diabetic rats.<br />

BMC Complementary and Alternative Medicine 6, 17–22.<br />

Renier, G., Mamputu, J.C., Serri, O., 2003. Benefits <strong>of</strong> gliclazide in the atherosclerotic<br />

process: decrease in monocyte adhesion to endothelial cells. Metabolism 52,<br />

13–18.<br />

R<strong>of</strong>fey, B.W.C., Atwal, A.S., Johns, T., Kubow, S., 2007. Water extracts <strong>from</strong> Momordica<br />

charantia increase glucose uptake and adiponectin secretion in 3T3-L1 adipose<br />

cells. Journal <strong>of</strong> Ethnopharmacology 112, 77–84.<br />

Sabu, M.C., Kuttan, R., 2002. Anti-diabetic activity <strong>of</strong> medicinal plants and its relationship<br />

with their antioxidant property. Journal <strong>of</strong> Ethnopharmacology 81,<br />

155–160.<br />

Saleem, A., Husheem, M., Harkonen, P., Pihlaja, K., 2002. Inhibition <strong>of</strong> cancer cell<br />

growth by crude extract and the phenolics <strong>of</strong> <strong>Terminalia</strong> <strong>chebula</strong> Retz. fruit.<br />

Journal <strong>of</strong> Ethnopharmacology 81, 327–336.<br />

Saleh, H.H., Rashad, H., Khafaga, S., 1952. Pharmacological action <strong>of</strong> <strong>Terminalia</strong><br />

<strong>chebula</strong>. Journal <strong>of</strong> the Egyptian Medical Association 35, 763–771.<br />

Sato, Y., Oketani, H., Singyouchi, K., Ohtsubo, T., Kihara, M., Shibata, H., Higuti,<br />

T., 1997. Extraction and purification <strong>of</strong> effective antimicrobial constituents <strong>of</strong><br />

<strong>Terminalia</strong> <strong>chebula</strong> Rets. against methicillin-resistant Staphylococcus aureus. Biological<br />

and Pharmaceutical Bulletin 20, 401–404.<br />

Shaila, H.P., Udupa, S.L., Udupa, A.L., 1998. Hypolipidemic activity <strong>of</strong> three indigenous<br />

drugs in experimentally induced atherosclerosis. International Journal <strong>of</strong><br />

Cardiology 67, 119–124.<br />

Shin, T.Y., Jeong, H.J., Kim, D.K., Kim, S.H., Lee, J.K., Chae, B.S., Kim, J.H., Kang, H.W., Lee,<br />

C.M., Lee, K.C., Park, S.T., Lee, E.J., Lim, J.P., Kim, H.M., Lee, Y.M., 2001. Inhibitory<br />

action <strong>of</strong> water soluble fraction <strong>of</strong> <strong>Terminalia</strong> <strong>chebula</strong> on systemic and local<br />

anaphylaxis. Journal <strong>of</strong> Ethnopharmacology 74, 133–140.<br />

Shukla, S.K., Chaudhary, P., Prem Kumar, I., Afrin, F., Puri, S.C., Qazi, G.N., Sharma,<br />

R.K., 2006. Cytotoxic and radioprotective <strong>effects</strong> <strong>of</strong> Podophyllum hexandrum.<br />

Environmental Toxicology and Pharmacology 22, 113–120.<br />

Takeuchi, M., Bucala, R., Suzuki, T., Ohkubo, T., Yamazaki, M., Koike, T., Kameda, Y.,<br />

Makita, Z., 2000. Neurotoxicity <strong>of</strong> advanced glycation end-products for cultured<br />

cortical neurons. Journal <strong>of</strong> Neuropathology and Experimental Neurology 59,<br />

1094–1105.<br />

Takeuchi, M., Yamagishi, S., 2004. Alternative routes for the formation <strong>of</strong><br />

glyceraldehyde-derived AGEs (TAGE) in vivo. Medical Hypotheses 63, 453–455.<br />

The Diabetes Control and Complications Trial/Epidemiology <strong>of</strong> Diabetes Interventions<br />

and Complications (DCCT/EDIC) Study Research Group, 2005. Intensive<br />

diabetes treatment and cardiovascular disease in patients with type 1 diabetes.<br />

New England Journal <strong>of</strong> Medicine 353, 2643–2653.<br />

Thornalley, P.J., 2003. Use <strong>of</strong> aminoguanidine (Pimagedine) to prevent the formation<br />

<strong>of</strong> advanced glycation endproducts. Archives <strong>of</strong> Biochemistry and Biophysics<br />

419, 31–40.<br />

Twigg, S.M., Cao, Z., McLennan, S.V., Burns, W.C., Brammar, G., Forbes, J.M., Cooper,<br />

M.E., 2002. Renal connective tissue growth factor induction in experimental<br />

diabetes is prevented by aminoguanidine. Endocrinology 143, 4907–4915.<br />

Wautier, M.-P., Chappey, O., Corda, S., Stern, D.M., Schmidt, A.M., Wautier, J.-L., 2001.<br />

Activation <strong>of</strong> NADPH oxidase by AGE links oxidant stress to altered gene expression<br />

via RAGE. American Journal <strong>of</strong> Physiology, Endocrinology and Metabolism<br />

280, E685–E694.<br />

Wendt, T., Tanji, N., Guo, J., Hudson, B.I., Bierhaus, A., Ramasamy, R., Arnold, B.,<br />

Nawroth, P.P., Yan, S.F., D’Agati, V., Schmidt, A.M., 2003. Glucose, glycation,<br />

and RAGE: implications for amplification <strong>of</strong> cellular dysfunction in diabetic<br />

nephropathy. Journal <strong>of</strong> the American Society <strong>of</strong> Nephrology 14, 1383–1395.<br />

Writing Team for the Diabetes Control and Complications Trial/Epidemiology <strong>of</strong> Diabetes<br />

Interventions and Complications Research Group, 2003. Sustained effect<br />

<strong>of</strong> intensive treatment <strong>of</strong> type 1 diabetes mellitus on development and progression<br />

<strong>of</strong> diabetic nephropathy: the Epidemiology <strong>of</strong> Diabetes Interventions and<br />

Complications (EDIC) Study. Journal <strong>of</strong> the American Medical Association 290,<br />

2159–2167.<br />

Xu, S.W., Howat, S.L., Renzoni, E.A., Holmes, A., Pearson, J.D., Dashwood, M.R., Bou-<br />

Gharios, G., Denton, C.P., du Bois, R.M., Black, C.M., Leask, A., Abraham, D.J., 2004.<br />

Endothelin-1 induces expression <strong>of</strong> matrix-associated genes in lung fibroblasts<br />

through MEK/ERK. Journal <strong>of</strong> Biological Chemistry 279, 23098–23103.<br />

Yamagishi, S., Inagaki, Y., Okamoto, T., Amano, S., Koga, K., Takeuchi, M., Makita, Z.,<br />

2002. Advanced glycation end product-induced apoptosis and overexpression<br />

<strong>of</strong> vascular endothelial growth factor and monocyte chemoattractant protein-<br />

1 in human-cultured mesangial cells. Journal <strong>of</strong> Biological Chemistry 277,<br />

20309–20315.<br />

Yamagishi, S., Nakamura, K., Matsui, T., Ueda, S., Fukami, K., Okuda, S., 2008. Agents<br />

that block advanced glycation end product (AGE)-RAGE (receptor for AGEs)oxidative<br />

stress system: a novel therapeutic strategy for diabetic vascular<br />

complications. Expert Opinion on Investigational Drugs 17, 983–996.<br />

Zannad, F., 2008. Implications <strong>of</strong> the ADVANCE study for clinical practice. Journal <strong>of</strong><br />

Hypertension 26 (Suppl. 3), S31–S34.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!