27.06.2013 Views

Planta Medica

Planta Medica

Planta Medica

SHOW MORE
SHOW LESS

Create successful ePaper yourself

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

<strong>Planta</strong> <strong>Medica</strong><br />

Volume 77, Issue 08, May 2011<br />

de la Garza, Ana Laura; Milagro, Fermín I.; Boque, Noemí; Campión, Javier;<br />

Martínez, J. Alfredo:<br />

Natural Inhibitors of Pancreatic Lipase as New Players in Obesity Treatment<br />

Wang, Cai-Ping; Wang, Yemin; Wang, Xing; Zhang, Xian; Ye, Jian-Feng; Hu,<br />

Lin-Shui; Kong, Ling-Dong:<br />

Mulberroside A Possesses Potent Uricosuric and Nephroprotective Effects in<br />

Hyperuricemic Mice<br />

Chow, Nicholas K.; Fretz, Michael; Hamburger, Matthias; Butterweck,<br />

Veronika:<br />

Telemetry as a Tool to Measure Sedative Effects of a Valerian Root Extract<br />

and Its Single Constituents in Mice<br />

Ghosh, Tirtha; Maity, Tapan Kumar; Singh, Jagadish:<br />

Antihyperglycemic Activity of Bacosine, a Triterpene from Bacopa monnieri, in<br />

Alloxan-Induced Diabetic Rats<br />

Shao, Meng; Qu, Kai; Liu, Ke; Zhang, Yuqin; Zhang, Ling; Lian, Zeqin; Chen,<br />

Tingting; Liu, Junfeng; Wu, Aili; Tang, Yue; Zhu, Haibo:<br />

Effects of Ligustilide on Lipopolysaccharide-Induced Endotoxic Shock in<br />

Rabbits<br />

Hong, Feng; Xiao, Weilie; Ragupathi, Govind; Lau, Clara B. S.; Leung, Ping<br />

Chung; Yeung, K. Simon; George, Constantine; Cassileth, Barrie; Kennelly,<br />

Edward; Livingston, Philip O.:<br />

The Known Immunologically Active Components of Astragalus Account for<br />

Only a Small Proportion of the Immunological Adjuvant Activity When<br />

Combined with Conjugate Vaccines<br />

Venâncio, Antônio Medeiros; Onofre, Alexandre Sherlley Casimiro; Lira,<br />

Amintas Figueiredo; Alves, Péricles Barreto; Blank, Arie Fitzgerald; Antoniolli,<br />

Ângelo Roberto; Marchioro, Murilo; dos Santos Estevam, Charles; Santos de<br />

Araujo, Brancilene:<br />

Chemical Composition, Acute Toxicity, and Antinociceptive Activity of the<br />

Essential Oil of a Plant Breeding Cultivar of Basil (Ocimum basilicum L.)


Halder, Sumita; Mehta, Ashish Krishan; Kar, Rajarshi; Mustafa, Mohammad;<br />

Mediratta, Pramod Kumari; Sharma, Krishna Kishore:<br />

Clove Oil Reverses Learning and Memory Deficits in Scopolamine-Treated<br />

Mice<br />

Ettefagh, Keivan A.; Burns, Johnna T.; Junio, Hiyas A.; Kaatz, Glenn W.;<br />

Cech, Nadja B.:<br />

Goldenseal (Hydrastis canadensis L.) Extracts Synergistically Enhance the<br />

Antibacterial Activity of Berberine via Efflux Pump Inhibition<br />

Yang, Heejung; Sung, Sang Hyun; Kim, Jinwoong; Kim, Young Choong:<br />

Neuroprotective Diarylheptanoids from the Leaves and Twigs of Juglans<br />

sinensis against Glutamate-Induced Toxicity in HT22 Cells<br />

Liu, Hai; Chou, Gui-Xin; Wang, Jun-Ming; Ji, Li-Li; Wang, Zheng-Tao:<br />

Steroidal Saponins from the Rhizomes of Dioscorea bulbifera and Their<br />

Cytotoxic Activity<br />

Schmidt, Thomas J.; Kaiser, Marcel; Brun, Reto:<br />

Complete Structural Assignment of Serratol, a Cembrane-Type Diterpene<br />

from Boswellia serrata, and Evaluation of Its Antiprotozoal Activity<br />

Zhao, Jianping; Avula, Bharathi; Joshi, Vaishali C.; Techen, Natascha; Wang,<br />

Yan-Hong; Smillie, Troy J.; Khan, Ikhlas A.:<br />

NMR Fingerprinting for Analysis of Hoodia Species and Hoodia Dietary<br />

Products<br />

Turski, Michal P.; Turska, Monika; Zgrajka, Wojciech; Bartnik, Magdalena;<br />

Kocki, Tomasz; Turski, Waldemar A.:<br />

Distribution, Synthesis, and Absorption of Kynurenic Acid in Plants<br />

Yang, Shuting; Chen, Chuan; Zhao, Yunpeng; Xi, Wang; Zhou, Xiaolong;<br />

Chen, Binlong; Fu, Chengxin:<br />

Association between Chemical and Genetic Variation of Wild and Cultivated<br />

Populations of Scrophularia ningpoensis Hemsl.


Natural Inhibitors of Pancreatic Lipase<br />

as New Players in Obesity Treatment<br />

Authors Ana Laura de la Garza, Fermín I. Milagro, Noemí Boque, Javier Campión, J. Alfredo Martínez<br />

Affiliation Department of Nutrition and Food Sciences, Physiology and Toxicology, University of Navarra, Pamplona, Spain<br />

Key words<br />

l " Orlistat<br />

l " high fat diet<br />

l " polyphenols<br />

l " saponins<br />

l " obesity<br />

l " fat digestion<br />

received Dec. 7, 2010<br />

revised February 11, 2011<br />

accepted February 21, 2011<br />

Bibliography<br />

DOI http://dx.doi.org/<br />

10.1055/s-0030-1270924<br />

Published online March 16,<br />

2011<br />

<strong>Planta</strong> Med 2011; 77: 773–785<br />

© Georg Thieme Verlag KG<br />

Stuttgart · New York ·<br />

ISSN 0032‑0943<br />

Correspondence<br />

Prof. J. Alfredo Martinez<br />

Department of Nutrition<br />

and Food Sciences,<br />

Physiology and Toxicology<br />

University of Navarra<br />

c/Irunlarrea 1<br />

31008 Pamplona<br />

Spain<br />

Phone: + 34 94842 56 00<br />

Fax: + 3494842 5649<br />

jalfmtz@unav.es<br />

Abstract<br />

!<br />

Obesity is a multifactorial disease characterized<br />

by an excessive weight for height due to an enlarged<br />

fat deposition such as adipose tissue, which<br />

is attributed to a higher calorie intake than the energy<br />

expenditure. The key strategy to combat obesity<br />

is to prevent chronic positive impairments in<br />

the energy equation. However, it is often difficult<br />

to maintain energy balance, because many available<br />

foods are high-energy yielding, which is usually<br />

accompanied by low levels of physical activity.<br />

The pharmaceutical industry has invested many<br />

efforts in producing antiobesity drugs; but only a<br />

lipid digestion inhibitor obtained from an actino-<br />

Introduction<br />

!<br />

Obesity is becoming one of the greatest threats to<br />

global health in this century, with more than 1.5<br />

billion overweight adults and at least 400 million<br />

of clinically obese subjects [1]. Due to these increasing<br />

obesity rates, the World Health Organization<br />

(WHO) has prompted to consider it as the<br />

epidemic of XXI century and to promote strategies<br />

to prevent and control its progress [2].<br />

The development of obesity is characterized by a<br />

chronic imbalance between energy intake and<br />

energy expenditure [3–5], and it is often ascribed<br />

to changing lifestyles and inadequate dietary habits<br />

[3]. Also, decreased energy expenditure is<br />

often associated with an inherited low basal metabolic<br />

rate, low energy cost of physical activity,<br />

and low capacity for fat oxidation [6]. To reduce<br />

body weight and adiposity, a change in lifestyle<br />

habits is still the crucial cornerstone [7]. Physical<br />

activity might be helpful in the prevention of obesity<br />

by elevating the average daily metabolic rate<br />

and increasing energy expenditure [3]. Unfortunately,<br />

this clinical approach is not long-term lasting,<br />

and weight regain is often seen. Drugs that<br />

Reviews<br />

bacterium is currently approved and authorized<br />

in Europe for obesity treatment. This compound<br />

inhibits the activity of pancreatic lipase, which is<br />

one of the enzymes involved in fat digestion.<br />

In a similar way, hundreds of extracts are currently<br />

being isolated from plants, fungi, algae, or<br />

bacteria and screened for their potential inhibition<br />

of pancreatic lipase activity. Among them,<br />

extracts isolated from common foodstuffs such<br />

as tea, soybean, ginseng, yerba mate, peanut, apple,<br />

or grapevine have been reported. Some of<br />

them are polyphenols and saponins with an inhibitory<br />

effect on pancreatic lipase activity, which<br />

could be applied in the management of the obesity<br />

epidemic.<br />

prevent weight regain appear necessary in obesity<br />

treatment [7]. Thus, the development of natural<br />

products for the treatment of obesity is a<br />

challenging task, which can be launched faster<br />

and cheaper than conventional single-entity<br />

pharmaceuticals [8]. Many medicinal plants may<br />

provide safe, natural, and cost-effective alternatives<br />

to synthetic drugs [9, 10]. Currently, one of<br />

the most important strategies in the treatment of<br />

obesity includes development of inhibitors of nutrient<br />

digestion and absorption. For example,<br />

acarbose is an antidiabetic drug that inhibits glycoside<br />

hydrolases, thus preventing the digestion<br />

of complex carbohydrates and decreasing postprandial<br />

hyperglycemia [11, 12]. Similar compounds<br />

with alpha-amylase inhibiting activity<br />

that can be used for diabetes control are being isolated<br />

from different plants. The list includes valoneaic<br />

acid dilactone [13], obtained from banaba<br />

(Lagerstroemia speciosa), the ethanol extract obtained<br />

from chestnut astringent skin [14], or the<br />

purified pancreatic alpha-amylase inhibitor isolated<br />

from white beans (Phaseolus vulgaris),<br />

which is able to reduce glycemia in both nondiabetic<br />

and diabetic rats [15].<br />

de la Garza AL et al. Natural Inhibitors of … <strong>Planta</strong> Med 2011; 77: 773–785<br />

773


774<br />

Reviews<br />

In this context, since dietary lipids represent the major source of<br />

unwanted calories, the inhibition of fat digestion is an interesting<br />

approach for reducing fat absorption [16]. Orlistat is the only authorized<br />

antiobesity drug in Europe and has been shown to act<br />

through inhibition of pancreatic lipase (PL), which is a key enzyme<br />

for the digestion of dietary triglycerides [17]. Orlistat is<br />

the saturated derivative of lipstatin, an inhibitor of PL isolated<br />

from the bacterium Streptomyces toxytricini [18]. This molecule<br />

exerts a modest weight lowering effect when accompanying a<br />

suitable dietary advice. Thus, in a recent meta-analysis [19], the<br />

mean BMI change with Orlistat (120 mg three times daily) was a<br />

reduction of 0.83 kg m −2 (95% CI: 0.47–1.19) compared with placebo.<br />

Accompanying this antiobesity action, Orlistat is also able<br />

to modestly reduce blood pressure, improve oral glucose tolerance<br />

and prevent the onset of type 2 diabetes [20, 21].<br />

Now, extracts from hundreds of species of medicinal plants, vegetables,<br />

and fruits [22] as well as products from microorganisms<br />

[9], fungi [23] and marine algae [24] are being screened for potential<br />

lipase inhibitory activity. Ideally, these treatments will be<br />

viewed as adjuncts to behavioral and lifestyle changes aimed at<br />

maintenance of weight loss and improved health [8].<br />

Obesity and High-Fat Diets<br />

!<br />

Epidemiological studies have shown a direct relation between<br />

the incidence of overweight/obesity and dietary fat consumption<br />

[3, 6,25].<br />

Humans are frequently exposed to fat rich foods, which are usually<br />

associated with a high-energy intake [6, 26]. Thus, those<br />

foods with a high-energy and dietary fat content are considered<br />

to promote body fat storage and weight gain in humans [8]. One<br />

explanation is that, in commercially available food items, the percentage<br />

of energy derived from fat is highly correlated with energy<br />

density. Given that fat contains 9 kcal/g compared with<br />

4 kcal/g for carbohydrates and proteins, foods rich in fat are often<br />

high in energy density. Thus, when a similar volume of food is<br />

consumed, energy intake will be higher in high-fat diets compared<br />

with low-fat diets [3].<br />

On the other hand, independently of an increased energy intake,<br />

specific dietary constituents may promote the development of<br />

obesity. This statement means that even consuming an equal<br />

amount of energy, the diet composition is important, especially<br />

the balance between nutrients [27, 28].<br />

Thus, a macronutrient profile (high-protein, high-carbohydrate,<br />

and high-lipid diets) can affect diet-induced thermogenesis, the<br />

oxidation pathway, energy intake, gene expression, or the level<br />

of some hormones [29]. Following a high-fat diet, the diet-induced<br />

thermogenesis is lower than following high-protein and<br />

carbohydrate diets, and also fat is more effectively absorbed from<br />

the gastrointestinal tract than are carbohydrates, which translates<br />

into lower energy expenditure when following a high-fat diet<br />

[26]. So, high-fat diets produce a metabolically more efficient<br />

state, at least in part because of the lower postprandial thermogenic<br />

effect of lipids in comparison with carbohydrates [30].<br />

Furthermore, the consumption of a high-fat diet has the capacity<br />

to modulate the gastrointestinal responses to ingested fat and,<br />

thereby, may lead to impairments in appetite regulation that favour<br />

the development of obesity. Dietary fat usually implies an<br />

increase in energy consumption because it has a lower potential<br />

for inducing satiety than carbohydrates and protein [6,31].<br />

de la Garza AL et al. Natural Inhibitors of… <strong>Planta</strong> Med 2011; 77: 773–785<br />

Hence, high-fat diets may play an important role in the increased<br />

prevalence of obesity and can be a triggering factor in the development<br />

of hyperglycemia and hyperinsulinemia [3, 32]. Moreover,<br />

the intake of dietary fats is usually accompanied by a higher<br />

intake of refined sweet carbohydrates (fast food, desserts), where<br />

the high intake of sucrose promotes weight gain, visceral adiposity,<br />

and the development of diseases that are related with obesity,<br />

such as diabetes and cardiovascular diseases [33]. Therefore, lowfat<br />

diets often are prescribed in the prevention and treatment of<br />

overweight and obesity because a reduction in dietary lipids<br />

without restriction of total energy intake could cause weight loss<br />

[26].<br />

Fat digestion<br />

Recent studies indicate that fat digestion is a prerequisite for the<br />

effects of fat on gastric emptying, gastrointestinal hormone secretion,<br />

appetite, and energy intake [6]. An increasing number of<br />

gastrointestinal enzymes involved in nutrient digestion are being<br />

identified and characterized, representing a rich pool of potential<br />

therapeutic targets for obesity and other metabolic disorders [9].<br />

Especially interesting are those enzymes that are related with dietary<br />

fat, which includes pre-duodenal lipases (lingual and gastric<br />

lipases), pancreatic lipase (PL), cholesterol-ester lipase, and<br />

bile-salt stimulated lipase [34].<br />

Most dietary fat is ingested as triglycerides (90–95%), and their<br />

hydrolysis starts in the mouth, then goes on through the stomach<br />

by an acid stable gastric lipase, and continues in the duodenum<br />

through the synergistic actions of gastric and colipase-dependent<br />

pancreatic lipases (PL), leading to the formation of monoglycerides<br />

and free fatty acids (FFA) (l " Fig. 1). FFA are absorbed<br />

by the enterocyte to synthesize new triglyceride molecules,<br />

which are transported to the different organs via lipoproteins, especially<br />

chylomicrons, after a meal [34].<br />

Pancreatic lipase (PL), encoded by the PNLIP gene in humans,<br />

plays a key role in the efficient digestion of triglycerides [35]. It<br />

is secreted into the duodenum through the duct system of the<br />

pancreas and is responsible for the hydrolysis of 50–70% of total<br />

dietary fats [9]. This enzyme has been widely used for the determination<br />

of the potential efficacy of natural products as antiobesity<br />

agents [36].<br />

Orlistat is currently the only clinically approved drug for obesity<br />

management in Europe. This molecule acts by inhibiting PL activity<br />

and the reduction of triglyceride absorption, and its long-term<br />

administration accompanying an energy restricted diet, results in<br />

weight loss [37]. Reduction on intestinal lipid digestion has been<br />

related to a decrease in the intra-abdominal fat content [7]. Thus,<br />

this compound is associated with a small, but statistically significant<br />

weight loss of about 3% more than diet alone in overweight<br />

and obese people [1]. In addition to losing weight, Orlistat within<br />

a prescribed diet has been shown to be safe and more effective<br />

than diet alone in modifying some of the risk of coronary artery<br />

disease and other obesity-related comorbidities. The most commonly<br />

reported adverse effects of Orlistat are a range of gastrointestinal<br />

side effects, including steathorrhea, bloating, oily spotting,<br />

fecal urgency, and fecal incontinence, as well as hepatic adverse<br />

effects [19, 38]. These adverse effects are similar to those<br />

observed for other lipase inhibitors tested in phase II studies,<br />

such as Cetilistat (ATL-962) [39].<br />

On the other hand, the inhibition of fat absorption could be accompanied<br />

by fat-soluble vitamin deficiencies, which could be<br />

prevented by the vitamin supplementation strategy, as other au-


Fig. 1 Fat metabolism in humans. Dietary fats are hydrolyzed in the gastrointestinal<br />

tract, where some lipases are involved.<br />

thors have recommended when vitamin deficiency occurs in patients<br />

undergoing Orlistat therapy [40].<br />

Hence the interest in the search for new natural substances that<br />

show potent inhibitory activity against PL and have fewer side effects<br />

than the current ones.<br />

Natural inhibitors of pancreatic lipase<br />

!<br />

In the continued search for effective antiobesity agents, several<br />

bacterial, fungal, and marine species have been screened to find<br />

new compounds with PL inhibitory activity.<br />

Many metabolic products from microorganisms, such as different<br />

kinds of Streptomyces (toxytricini, sp. NR 0619, albolongus, aburaviensis,<br />

andlavendulae) have a potent inhibitory activity of PL<br />

[9]. Lipstatin was isolated from an actinobacterium, Streptomyces<br />

toxytricini, and the catalytic hydrogenation product of lipstatin is<br />

the approved antiobesity drug tetrahydrolipstatin (Orlistat; marketed<br />

by Roche as Xenical) [18]. Panclicins, analogs of tetrahydrolipstatin<br />

isolated from Streptomyces sp. NR0619, also present<br />

strong anti-lipase activity [41]. Other compounds which also act<br />

as potent inhibitors of PL, at least in vitro, are ebelactones A and B,<br />

isolated from Streptomyces aburaviensis [42], and vibralactone,<br />

isolated from the culture broth of the polypore Boreostereum vibrans<br />

[43]. Finally, other examples of lipase inhibitors have been<br />

obtained from yeasts and fungi such as Candida antarctica, Candida<br />

rugosa, Gestrichum candidum, Humicola lanuginose, and<br />

Pseudomonas glumae, which have received special attention and<br />

are widely used in the pharmaceutical industry [44].<br />

Due to the biodiversity and unexplored resources, the fungal<br />

kingdom has been particularly searched to find new substances<br />

with lipase inhibitory activity. In a thorough screening of lipase<br />

inhibitors of fungal origin in Slovenia [23], extracts obtained from<br />

three species, Laetiporus sulphureus, Tylopilus felleus, andHygrocybe<br />

conica, exhibited very high lipase inhibitory activities (83%<br />

± 5%, 96% ± 3%, and 97% ± 5%, respectively), even higher than Orlistat.<br />

Pleurotus eryngii water extract also shows a significant in-<br />

Reviews<br />

hibitory activity against PL, preventing postprandial hyperlipidemia<br />

through low intestinal absorption of dietary fat [45]. Finally,<br />

the water and ethanol extracts from fruiting bodies of Phellinus<br />

linteus show a potent lipase inhibitory and antiobesity effect<br />

[46]. A special case is that of monascus pigments from Monascus<br />

sp., which have been used for many years as natural colorants and<br />

as a healthy food in East Asia, being employed in the production<br />

of certain fermented foods. Various monascus derivatives with<br />

incorporated unnatural amino acids show inhibitory activities<br />

against lipase [47].<br />

In the same way, marine products are an especially rich source of<br />

bioactive compounds [48]. In a milestone study, Bitou et al. [24]<br />

screened the lipase inhibitory activities of methanol and ethyl<br />

acetate extracts from 54 species of marine algae. These investigations<br />

observed a very high activity (almost 100% inhibition) in<br />

the methanol extracts from Caulerpa taxifolia and Asparagopsis<br />

tociformis, although the methanolic extracts of other Chlorophyta<br />

(i.e., Caulerpa okamurae or Codium latum), Rhodophyta (i.e.,<br />

Gloiopeltis tenax or Hypnea charoides), and Phaeophyta (i.e., Sargassum<br />

muticum, Dictyopteris latiuscula, orCutleria cylindrica),<br />

were also very promising. In this sense, Phaeophyta generally<br />

contains large amounts of polyphenols, such as tannins, with lipase-inhibiting<br />

activity. In fact, most compounds with a porphyrin<br />

structure are able to inhibit lipase activity [49]. Two algae<br />

whose extracts inhibit gastric and pancreatic lipases are Caulerpa<br />

prolifera, which may be a source of a potential antiobesity agent<br />

[50], and Caulerpa taxifolia, which synthesizes the toxin caulerpenyne<br />

[24]. On the other hand, carotenoids from Undaria pinnatifida<br />

and Sargassum fulvellum, specifically fucoxanthin that is<br />

metabolized in vivo to fucoxanthinol, suppress triglyceride absorption<br />

via the inhibition of PL in the intestinal lumen [51].<br />

Medicinal plants have been used as dietary supplements for body<br />

weight management and control in many countries. In this sense,<br />

presence of PL inhibitors has been demonstrated in different<br />

plant species (l " Table 1), although more research is needed for<br />

identifying and characterizing effective lipase inhibitors [52]. Lipase<br />

inhibitors of plant origin include certain proteins, such as<br />

those from soybean [53] and from wheat bran and germ [54].<br />

Other proteins that strongly inhibit hydrolysis of triglycerides<br />

are the basic protein protamine [55] and ε-polylysine [56], which<br />

could act, as several amphiphilic proteins like ovoalbumin and βlactoglobulin<br />

[57], by the desorption of lipase from its substrate<br />

due to a change in interfacial quality [58].<br />

Other lipase inhibitors from plant origin are basic polysaccharides,<br />

especially chitosan oligosaccharides, water-soluble chitosan<br />

(46 kDa) and polydextrose when a basic group is introduced<br />

[59, 60], phytic acid and other myoinositol phosphate esters [61],<br />

phenylboronic acid, a potent inhibitor of lipase from Oryza sativa<br />

[62], and carnosic acid, a diterpene isolated from the methanolic<br />

extract of the leaves of sage (Salvia officinalis) and rosemary [63].<br />

Korean and Chinese researchers have been very active in the<br />

search of new lipase inhibitors of herbal origin. Among the most<br />

promising compounds, there are platycodin D, isolated from the<br />

fresh roots of Platycodon grandiflorum [64, 65], dioscin from Dioscorea<br />

nipponica [66], licochalcone A from the roots of Glycyrrhiza<br />

uralensis [67], phenolic constituents from the leaves of Nelumbo<br />

nucifera [68], the aqueous ethanol extracts of Juniperus communis<br />

or common juniper (bark) and Illicium religiosum (wood)<br />

[69], the ethanol extract from stem bark and leaves from mango<br />

tree (Mangifera indica), which is able to prevent weight gain induced<br />

by feeding a high-fat diet to Wistar rats [70], a pomegranate<br />

leaf extract rich in ellagic acid and tannins [71], Rhei rhizoma<br />

de la Garza AL et al. Natural Inhibitors of … <strong>Planta</strong> Med 2011; 77: 773–785<br />

775


776<br />

Reviews<br />

Table 1 Plant extracts that showed over 40% inhibitory activity in vitro of pancreatic lipase and part of the plant from which the extract has been isolated.<br />

Family Scientific name Common Part Ref Family Scientific name Common Part Ref<br />

name<br />

of plant<br />

name of plant<br />

Aeraceae Acer pseudosiebol- Korean maple Whole [138] Lamiaceae Spirodela polyrhiza Common Whole [138]<br />

dianum<br />

duckmeat<br />

Anacardiaceae Pistacia vera Pistachio Fruits hull [52] Lamiaceae Thymus pulegoides Lemon thyme Whole [22]<br />

Apiaceae Levisticum officinale Garden lovage Whole [52] Lauraceae Cinnamomum<br />

zeylanicum<br />

Cinnamon Derm [52]<br />

Apiaceae Sanicula chinensis Bian Dou Cai Whole [138] Lauraceae Lindera glauca Grayblue<br />

spicebush<br />

Whole [138]<br />

Araliaceae Eleutherococcus Siberian ginseng Leaves [114] Liliaceae Asparagus<br />

Shiny asparagus Radix [138]<br />

senticosus<br />

cochinchinesis<br />

Aspidiaceaes Cyrtomium falcatum Japanese holly fern Whole [138] Liliaceae Scilla scilloides Chinese scilla Whole [138]<br />

Asteraceae Artemisia scoparia Redstem Whole [138] Linaceae Linum<br />

Oil flax Seed [139]<br />

wormwood<br />

usitatissimum<br />

Asteraceae Helianthus annus Common Seed [139] Lythraceae Lythrum salicaria Purple Whole [138]<br />

sunflower<br />

loosestrife<br />

Brassicaceae Brassica nigra Black mustard Radix [22] Musaeae Musa sapientum French plantain Fructus [22]<br />

Brassicaceae Brassica oleracea<br />

capitata<br />

Cabbage Folium [22] Myricaceae Myrika spp Bayberry Bark [140]<br />

Brassicaceae Raphanus sativus Radish Radix [22] Myrtaceae Myrtus communis True myrtle Leaves [52]<br />

Caprifoliaceae Lonicera japonica Japanese honeysuckle<br />

Whole [138] Myrtaceae Solanum tuberosum Potato Flowers [22]<br />

Celastraceae Euonymus<br />

sachalinensis<br />

Spindletree Whole [138] Oleaceae Olea europeae Olive Folium [22]<br />

Crassulaceae Rhodiola rosea Roseroot Whole [141] Orchida- Gastrodia elata Tien Ma Whole [138]<br />

stonecrop<br />

ceae<br />

Cucurbitaceae Cucurbita pepo Field pumpkin Whole [138] Oxalidaceae Oxalis corniculata Sleeping<br />

beauty<br />

Whole [138]<br />

Cucurbitaceae Momordica<br />

Spiny<br />

Whole [138] Poaceae Eriochloa villosa Hairy cup- Whole [138]<br />

cochinchinensis bittergourd<br />

grass<br />

Cyperaceae Bulbostylis barbata Watergrass Whole [138] Poaceae Hemarthria sibirica Weed Whole [138]<br />

Cyperaceae Carex kobomugi Japanese Whole [138] Poaceae Panicum dichotomi- Fall panic- Whole [138]<br />

sedge<br />

florumgrass<br />

Cyperaceae Cyperus amuricus Asian<br />

Whole [138] Poaceae Setaria italica Foxtail bris- Whole [138]<br />

flatsedge<br />

tlegrass<br />

Eleagnaceae Elaeagnus macro- Oleaster Whole [138] Polygala- Polygala tenuifolia Yuan Zhi Whole [138]<br />

phyllaceae<br />

Ericaceae Arctostaphylos uva-ursi Bear berry Folium [22] Polygona- Reynoutria elliptica Black bind- Whole [138]<br />

ceaeweed<br />

Ericaceae Vaccinium myrtillus Bilberry Fructus [22] Polygona- Rheum ribes Rhubarb Rhi- [52]<br />

ceaezomes<br />

Eriocaulaceae Eriocaulon siebol- Flattened Whole [138] Potamogetona- Potamogeton distinctus Pondweed Whole [138]<br />

dianum<br />

pipewort<br />

ceae<br />

Fabaceae Alhagi camelorum Camelthorn Aerial<br />

parts<br />

[52] Rosaceae Rosa damascene Damask rose Floret [52]<br />

Fabaceae Glycyrrhiza uralensis Gan Cao Whole [138] Rosaceae Rubus idaeus Raspberry Fructus [22]<br />

Fabaceae Lespedeza cuneata Chinese bush<br />

clover<br />

Whole [138] Rosaceae Malus domestica Apple Fructus [22]<br />

Fabaceae Phaseolus vulgaris Common bean Whole [22] Rubiaceae Gardenia jasmi- Cape jas- Whole [138]<br />

noidesmine<br />

Fabaceae Pisum sativum Garden pea Fructus [22] Rubiaceae Rubia akane Asian madder<br />

Whole [138]<br />

Fabaceae Pueraria thunbergiana Kudzu Whole [138] Rutaceae Citrus aurantifolium Lime Whole [138]<br />

Fabaceae Quercus infectoria Aleppo oak Galls [52] Rutaceae Murraya koeninggi Curryleaf<br />

tree<br />

Leaves [142]<br />

Juncaceae Juncus effusus Soft rush Whole [138] Rutaceae Orixa japonica Pearl frost Whole [138]<br />

Lamiaceae Agastache rugosa Purple giant Whole [138] Saxifragaceae Chrysosplenium Golden saxifrage Whole [138]<br />

hyssop<br />

grayanum<br />

Lamiaceae Origanum vulgare Oregano Herba [22] Simaroubaceae Ailanthus altissima Tree of heaven Whole [138]<br />

Lamiaceae Prunella vulgaris Common selfheal Whole [73] Tiliaceae Tilia platyphyllos Largeleaf linden Whole [22]<br />

Lamiaceae Rosmarinus officinalis Rosemary Folium [22] Urticaceae Urtica urens Dwarf nettle Aerial parts [52]<br />

Lamiaceae Salvia officinalis Salvia Folium [22] Zingiberaceae Afromomum<br />

Meleguetta Seed [143]<br />

meleguetta<br />

pepper<br />

de la Garza AL et al. Natural Inhibitors of… <strong>Planta</strong> Med 2011; 77: 773–785


Table 2 Some classes of natural compounds that have been reported to inhibit pancreatic lipase activity in vitro and species from which the compound has been<br />

obtained.<br />

Metabolites Scientific name Common name Family References<br />

Flavonoids Alpinia officinarum Lesser galangal Zingiberaceae [144,145]<br />

Flavonoids Taraxacum officinale Dandelion Asteraceae [103]<br />

Flavonoids, triterpenes Actinidia arguta Kiwi Actinidiaceae [146]<br />

Polyphenols Arachis hypogaea Peanut Fabaceae [9]<br />

Polyphenols Mangifera indica Mango Anacardiaceae [9]<br />

Polyphenols <strong>Medica</strong>go sativa Alfalfa Fabaceae [78]<br />

Polyphenols Nelumbo nucifera Sacred lotus Nelumbonaceae [9]<br />

Polyphenols Salacia reticulate Kotala himbutu Celastraceae [101]<br />

Polyphenols Salix matsudana Corkscrew willow Salicaceae [147]<br />

Polyphenols, proanthocyanidins,<br />

catechins<br />

Camellia sinensis Green, black, oolong tea Theaceae [89]<br />

Polyphenols, saponins Ilex paraguariensis Yerba mate Aquifoliaceae [99]<br />

Proanthocyanidins Cassia mimosoides Nomame herba Fabaceae [148]<br />

Proanthocyanidins Cinnamomum sieboldii Cinnamon Lauraceae [86]<br />

Proanthocyanidins Theobroma cacao Cocoa Malvaceae [86]<br />

Proanthocyanidins, saponins Vitis vinifera Grape vine Vitaceae [79,104]<br />

Saponins Aesculus hippocastanum Horse chestnut Sapindaceae [32]<br />

Saponins Aesculus turbinate Japanese horse chestnut Hippocastanaceae [110]<br />

Saponins Arctostaphylos uva-ursi Bearberry Ericaceae [32]<br />

Saponins Ardisia japonica Marlberry Myrsinaceae [152]<br />

Saponins Avena sativa Oat Poaceae [149]<br />

Saponins Coffea Arabica Coffee Rubiaceae [32]<br />

Saponins Cyclocarya paliurus Wheel wingnut Juglandaceae [9]<br />

Saponins Dioscorea nipponica Yam Dioscoreaceae [9]<br />

Saponins Eleutherococcus senticosus Siberian ginseng Araliaceae [114]<br />

Saponins Eleutherococcus sessiliflorus Sessiloside Araliaceae [9]<br />

Saponins Gardenia jasminoides Cape jasmine Rubiaceae [118]<br />

Saponins Gypsophila oldhamiana Oldhamʼs babyʼs-breath Caryophyllaceae [119]<br />

Saponins Kochia scoparia Burningbush Chenopodiaceae [150]<br />

Saponins Malus domestica Apple Rosaceae [32]<br />

Saponins Momordica charantia Balsampear Cucurbitaceae [151]<br />

Saponins Olea europeae Olive Oleaceae [32]<br />

Saponins Panax ginseng Ginseng Araliaceae [109]<br />

Saponins Panax japonicus Japanese ginseng Araliaceae [120]<br />

Saponins Panax quinquefolium American ginseng Araliaceae [122]<br />

Saponins Platycodi radix Doraji Campanulaceae [64]<br />

Saponins Platycodon grandiflorum Balloon flower Campanulaceae [103]<br />

Saponins Sapindus rarak Soapberry Sapindaceae [127]<br />

Saponins Scabiosa tschiliensis Pincushions Dipsacaceae [9]<br />

Saponins Solanum lycopersicum Tomato Solanaceae [32]<br />

Terpenes Salvia officinalis Salvia Lamiaceae [32]<br />

Triterpenes Aloe vera Aloe vera Asphodelaceae [32]<br />

Triterpenes Betula alba Birch Betulaceae [32]<br />

Triterpenes Calendula officinalis Pot marigold Asteraceae [32]<br />

Triterpenes Melissa officinalis Lemon balm Lamiaceae [32]<br />

Triterpenes Origanum vulgare Oregano Lamiaceae [32]<br />

(rhubarb) and the combinatorial drug Chunghyuldan [72], Prunella<br />

vulgaris, Rheum palmatum, and other herbs [73]. Most of<br />

the common compounds that are found in different plant species<br />

are polyphenols, saponins, and terpenes (l " Table 2).<br />

In the following chapters more information will be given out<br />

about the most thoroughly studied compounds, classified according<br />

to their biochemical structure.<br />

Polyphenols<br />

A number of studies have revealed various health benefits of<br />

plant polyphenols and their importance in foods, beverages, and<br />

natural medicine. In this context, polyphenols have some potential<br />

efficacy for preventing obesity. They inhibit enzymes related<br />

to fat metabolism including PL, lipoprotein lipase, and glycero-<br />

Reviews<br />

phosphate dehydrogenase [74]. Polyphenol extracts are able to<br />

decrease the blood levels of glucose, triglycerides, and LDL cholesterol,<br />

increase energy expenditure and fat oxidation, and reduce<br />

body weight and adiposity [75, 76]. In fact, many polyphenols,<br />

including flavones, flavonols, tannins, and chalcones, have<br />

shown an inhibitory activity of PL [9, 22].<br />

Flavonoids are a type of plant secondary metabolites that are<br />

characterized as containing two or more aromatic rings, each<br />

bearing at least one aromatic hydroxyl and connected with a carbon<br />

bridge [76]. Some of them are polymerized into large molecules,<br />

either by the plants themselves or as a result of food processing.<br />

These polymers are called tannins, and three subclasses<br />

(condensed tannins, derived tannins, and hydrolysable tannins)<br />

exhibit a variety of beneficial effects on health [76]. A flavonoid<br />

de la Garza AL et al. Natural Inhibitors of … <strong>Planta</strong> Med 2011; 77: 773–785<br />

777


778<br />

Reviews<br />

with PL inhibitory activity is hesperidin, obtained from the peels<br />

of Citrus unshiu [77].<br />

Proanthocyanidins (PA), also known as condensed tannins, are<br />

the most common group of flavonoids in the Western diet. They<br />

consist of monomeric units of flavans linked through carbon-carbon<br />

and ether linkages, which are considered the second most<br />

abundant group of natural phenolics after lignins [78]. PA can be<br />

found in such common foodstuffs as cereals, legumes, fruits, vegetables,<br />

and beverages (red wine and tea in particular) [75, 79].<br />

They have a putative role as antioxidants, showing beneficial effects<br />

on inflammatory processes, cardiovascular diseases, and<br />

other pathological conditions [80, 81]. For example, these compounds<br />

actively reduce plasma triglycerides by inhibiting the absorption<br />

of dietary lipids [79] and possess inhibitory effects on<br />

different digestive enzymes, such as trypsin, amylase, and lipase<br />

[36].<br />

Some examples of polyphenols with inhibitory action on PL are<br />

proanthocyanidins from edible herbs, such as those from Cassia<br />

mimosoides [82], and tea catechins, especially (−)-catechin gallate<br />

and (−)-gallocatechin gallate, [83]. Some of the most thoroughly<br />

studied polyphenol extracts in relation to PL inhibition are the<br />

following:<br />

Arachis hypogaea: Peanut (Arachis hypogaea) shells (hulls, seed<br />

coats), which are by-products of the peanut industry, provide<br />

several compounds showing PL inhibitory activity in a dose dependent<br />

manner (1 mg/mL = 42% inhibitory effect) that are able<br />

to reduce body weight gain in rats fed a high-fat diet [84]. This<br />

plant contains several bioactive molecules, such as luteolin<br />

(l " Fig. 2), certain fatty acids, caffeic, ferulic, and benzoic acids,<br />

all of which are able to inhibit lipases [9]. Coumarin derivates<br />

and phenolic acids were assumed to be the major active constituents.<br />

However the authors have not examined the individual effects<br />

of each compound.<br />

Camellia sinensis: Camellia sinensis or tea plant (green tea, black<br />

tea, or oolong tea) contains over 60 polyphenols, some of them<br />

with a potent PL inhibitory activity. It is likely the plant whose<br />

extracts have been more thoroughly used for searching new PL<br />

inhibitors. The major polyphenols are catechins (l " Fig. 2), which<br />

constitute about one-third of its total dry weight. A serving of tea<br />

is moderate to high in flavonoid and/or tannin content [85–89].<br />

Nakai et al. [90] found that the polyphenols with more potent PL<br />

inhibitory effect were flavan-3-ol digallate esters isolated from<br />

oolong tea, such as (−)-epigallocatechin-3,5-digallate. Oolong<br />

tea-polymerized polyphenols reduced postprandial hypertriglyceridemia<br />

in olive oil-loaded rats and mice [91]. Also (−)-epigallocatechin,<br />

abundant in the green tea extract, is a weak inhibitor of<br />

PL and is able to decrease the postprandial hypertriglyceridemia<br />

in rodents [92].<br />

The administration of black-tea polyphenols suppressed postprandial<br />

hypertriglyceridemia in a dose-dependent manner in<br />

rats, with theaflavin-3,3′-digallate as the most effective PL inhibitor<br />

[93], whereas other authors point out to thearubigins [94].<br />

These extracts are able to prevent increases in body weight and<br />

adiposity in mice fed a high-fat diet [95]. The PL inhibitory and<br />

hypotriglyceridemic effects of tea extracts were corroborated by<br />

Tanaka et al. [96], who orally administered mixed fermented tea<br />

extracts and Loquat tea extracts to rats with a 10% soybean oil<br />

emulsion.<br />

Finally, cocoa tea extract (Camellia sinensis var. ptilophylla) is rich<br />

in polyphenols with PL inhibitory effect. A single oral administration<br />

of this extract produces an inhibition of plasma triglyceride<br />

levels in olive oil-loaded ICR mice and triolein-loaded rats [97].<br />

de la Garza AL et al. Natural Inhibitors of… <strong>Planta</strong> Med 2011; 77: 773–785<br />

Fig. 2 Selected polyphenols with PL inhibitory activity: Luteolin (1) from<br />

Arachis hypogaea, catechin (2) from Camellia sinensis, daidzein (3) from Glycine<br />

max, quercetin (4) from Ilex paraguariensis, structure of a procyanidin<br />

(5) from Vitis vinifera.<br />

Glycine max: Daidzein (l " Fig. 2) belongs to the group of isoflavones<br />

and is produced almost exclusively by the members of the<br />

Fabaceae/Leguminosae (bean) family such as soybean. In one<br />

study, Guo et al. [98] investigated the effects of daidzein on body<br />

weight, adipose tissue, blood, and liver lipid levels in obese mice<br />

fed a high-fat diet, finding that daidzein reduced body and white<br />

adipose tissue weights in obese mice and ameliorated the hyperlipidemia<br />

induced by the high-fat diet. The authors attributed<br />

this effect to the inhibition of PL activity and fat digestion.<br />

Ilex paraguariensis: Yerba mate (MT) is a plant from the subtropical<br />

region of South America that is widely consumed in Brazil,<br />

Argentina, Paraguay, and Uruguay. Yerba mate contains polyphenols,<br />

such as flavonoids (quercetin and rutin) (l " Fig. 2) and phenolic<br />

acids (chlorogenic and caffeic acids), and is also rich in caffeine<br />

and saponins [99]. These substances act on the lipid metabolism<br />

by inhibiting PL activity in a concentration value of 1.5 mg/<br />

mL [99]. Several triterpene saponins and monoterpene oligoglycosides<br />

from the leaves of yerba mate were found to exhibit potent<br />

inhibitory activity on porcine PL [100].<br />

Malus domestica: Apples (Malus domestica) belong to the Rosaceae<br />

family whose fruits contain several phenolic substances<br />

(cholorogenic acid, catechin, epicatechin, phloridzin, and procyanins).<br />

Procyanidins in apples are mainly composed of various<br />

polymerized catechins, with some of them showing a PL inhibitory<br />

activity and reducing triglyceride absorption [36]. In corn<br />

oil-loaded mice, a single oral administration of apple polyphenols<br />

reduced plasma triglyceride levels, and a test diet containing<br />

600 mg of apple polyphenols significantly inhibited triglyceride


elevation at 6 h after ingestion, indicating an inhibition of triglyceride<br />

absorption [36].<br />

Salacia reticulata: Salacia reticulata contains a high concentration<br />

of polyphenols, including catechins and condensed tannins.<br />

In hot water-soluble extract from the roots of Salacia reticulata<br />

(SRHW) the concentration is about 24% polyphenols [74]. The<br />

polyphenols from Salacia reticulata inhibit enzymes related to<br />

fat metabolism, including PL, lipoprotein lipase, and glycerophosphate<br />

dehydrogenase, and are effective in preventing obesity<br />

[101]. In fact, Salacia extract markedly improved metabolic syndrome<br />

symptoms (including body weight, adiposity, glucose intolerance,<br />

hypertension, and peripheral neuropathy) in TSOD<br />

mice [102].<br />

Taraxacum officinale: Dandelion (Taraxacum officinale) is a perennial<br />

herbaceous plant of the family Asteraceae that has been<br />

used as a phytomedicine due to its choleretic, antirhemetic, diuretic,<br />

and anti-inflammatory properties [103]. Extracts from<br />

this plant have shown hypolipidemic effects and an inhibitory activity<br />

of PL, decreasing AUC (area under curve) for the postprandial<br />

triglyceride response curve [103].<br />

Vitis vinifera: Grapevine (Vitis vinifera) has become a model plant<br />

for studying proanthocyanidin biosynthesis. Grapevine proanthocyanidins<br />

(l " Fig. 2) consist of two major flavan 3-ol monomers,<br />

catechin and epicatechin, that have inhibitory activity on<br />

PL [79, 104].<br />

Polyphenol-rich extracts from a range of berries, particularly<br />

cloudberry, are able to inhibit PL activity in vitro, which has been<br />

attributed to their content in ellagitannins and proanthocyanidins<br />

[105].<br />

Saponins<br />

Saponins are a major family of secondary metabolites that occur<br />

in a wide range of plants species [106]. These compounds have<br />

been isolated from different parts of the plants, including the<br />

roots, rhizomes, stems, bark, leaves, seeds, and fruits. Occasionally,<br />

the whole plant has been used [107].<br />

Saponins are categorized into two major classes, the triterpenoid<br />

and the steroid saponins, which are both derived from the 30 carbon<br />

atoms containing precursor oxidosqualene [107, 108]. Some<br />

of the triterpene-rich plant materials are common foodstuffs<br />

consumed in large amounts in Mediterranean countries. Therefore,<br />

the correlation of a triterpene-rich diet and the beneficial effects<br />

of consuming a Mediterranean diet should be investigated<br />

in more detail [32]. These types of plant secondary metabolites<br />

are found to inhibit PL and, thus, may represent potential effective<br />

treatments for obesity and related disorders [9, 22].<br />

Aesculus turbinata: The Japanese horse chestnut (Aesculus turbinata)<br />

is a medicinal plant widely used in East Asia. The saponin<br />

mixture extracted from the seeds is called escins and has a strong<br />

inhibitory activity on PL [110]. In mice fed a high-fat diet, total<br />

escins suppressed the increase in body weight, adiposity, and liver<br />

fat and increased triglyceride level in the feces, whereas it decreased<br />

plasma triglycerides after the oral administration of a lipid<br />

emulsion [111, 112].<br />

Dioscorea nipponica: The methanol extract of Dioscorea nipponica<br />

Makino powder has a potent inhibitory activity against porcine<br />

PL, with an IC 50 value of 5–10 µg/mL [66]. In fact, the saponin dioscin<br />

and its aglycone, diosgenin, both suppressed the increase of<br />

blood triacylglycerols when orally injected with corn oil to mice.<br />

Rats fed a high-fat diet containing 5% Dioscorea nipponica Makino<br />

gained significantly less body weight and adipose tissue than<br />

control animals [66], and a similar result has been observed after<br />

Reviews<br />

Fig. 3 Selected isoprenoids with PL inhibitory activity: Eleutheroside (6)<br />

from Eleutherococcus senticosus, geniposide (7) from Gardenia jasminoides,<br />

general structure of dammaran aglycons of ginsenosides (8)inPanax ginseng,<br />

betulin (9) from Betula alba.<br />

administering the aqueous extract of this rhizome to mice fed a<br />

high-fat diet [113].<br />

Eleutherococcus senticosus: Eleutherococcus senticosus is a shrub,<br />

belonging to the family Araliaceae, which is commonly distributed<br />

in north-eastern Asia. It is used as a traditional Chinese<br />

medicine against ischemic heart diseases, neurasthenia, hypertension,<br />

arthritis, and tumors [114]. At least fifteen triterpenoid<br />

saponins with in vitro PL inhibitory activity (l " Fig. 3) have been<br />

isolated from the fruits of Eleutherococcus senticosus [115]. The<br />

total saponin fraction obtained from the fruits of Eleutherococcus<br />

senticosus exhibits inhibitory activity on PL with an IC50 value of<br />

3.63 mg/mL [114].<br />

Eleutherococcus sessiliflorus: Different lupine-type triterpene triglycosides<br />

isolated from a hot water extract of Eleutherococcus<br />

sessiliflorus leaves are able to inhibit PL activity in vitro and to<br />

suppress the body weight gain of mice fed a high-fat diet [116].<br />

Gardenia jasminoides: Crocin is a glycosylated carotenoid extracted<br />

from the fructus of Gardenia jasminoides (l " Fig. 3). Gardeniae<br />

Fructus is used in Asian countries as a natural colorant,<br />

and in Chinese traditional medicine for its antioxidant, cytotoxic,<br />

antitumor, and neuroprotective effects. Crocin and crocetin are<br />

effective hypolipidemic agents that act by reducing the absorption<br />

of fat and cholesterol through inhibition of PL activity [117].<br />

Sheng et al. demonstrated that crocin selectively inhibited the activity<br />

of PL as a competitive inhibitor [118].<br />

Gypsophila oldhamiana: Gypsophila oldhamiana (Caryophyllaceae)<br />

is a plant distributed in the north of China whose roots have<br />

high amounts of saponins, sterols, and fatty acids. The extract<br />

from this plant shows a potent inhibitory activity of PL with an<br />

IC 50 value of 0.54 mg/ml [118,119] and different triterpenoid<br />

saponins, gypsosaponins A–C, as the more efficient compounds<br />

[119].<br />

Panax ginseng: Ginseng is one of the most popular medicinal<br />

herbs and is commonly consumed as powder, a beverage, or a<br />

food supplement. Roots of Panax ginseng contain high levels of<br />

de la Garza AL et al. Natural Inhibitors of … <strong>Planta</strong> Med 2011; 77: 773–785<br />

779


780<br />

Reviews<br />

ginsenosides (l " Fig. 3), which are steroidal saponins that show<br />

beneficial effects on lipid metabolism. Saponins from ginseng<br />

roots suppressed the expected increase in body weight and plasma<br />

triacylglycerols in mice following a high-fat diet, which was<br />

probably mediated by inhibiting PL with an IC50 value of 500 µg/<br />

mL [109].<br />

Panax japonicus: The rhizomes of Panax japonicus (Japanese ginseng)<br />

are used in folk medicine for the treatment of arteriosclerosis,<br />

hyperlipidemia, hypertension, and diabetes mellitus. Chikusetsusaponins<br />

prevented the increase in body weight and parametrial<br />

adipose tissue weight induced by a high-fat diet and inhibited<br />

the elevation of postprandial plasma triacylglycerols due<br />

to their inhibitory action of PL on dietary fat [120]. The delay in<br />

intestinal fat absorption was also behind the antiobesity effects<br />

observed for Korean white ginseng extract in high-fat diet-induced<br />

obese mice [121].<br />

Panax quinquefolium: American ginseng (Panax quinquefolium) is<br />

a native plant from North America. The saponins isolated from<br />

stems and leaves of Panax quinquefolium may prevent fat storage<br />

in adipose tissue and postprandial elevations of plasma triacylglycerols<br />

by inhibiting the intestinal absorption of dietary fat<br />

through the inhibition of PL activity [122].<br />

Platycodi grandiflorum: Platycodi radix, widely used in traditional<br />

Oriental medicines as a remedy for respiratory disorders, is<br />

rich in saponins, which are responsible for a diversity of effects<br />

including anti-inflammation, antiallergy, antitumor, and immunostimulation<br />

[64]. Given its inhibitory action on PL [123], with<br />

platycodin D as the most efficient compound [124], it ameliorated<br />

high fat-induced obesity in mice [125] and rats [64]. SK1 is<br />

an edible saponin-rich compound from Platycodi Radix that is<br />

able to reduce body weight and fat accumulation by increasing<br />

fecal lipid outputs in high-fat fed mice [126].<br />

Sapindus rarak: The methanolic extract from the pericarps of Sapindus<br />

rarak (Lerak) shows a PL inhibitory activity that is probably<br />

due to diverse saponins and sesquiterpene glycosides [127].<br />

Scabiosa tschiliensis: Different triterpenoid saponins isolated<br />

from the Mongol and Chinese traditional medicinal herb Scabiosa<br />

tschiliensis have shown strong inhibition of PL in vitro [128]. Due<br />

to the difficult task of isolating scabiosaponins and the scarceness<br />

of this type of saponin in nature, some of them have been successfully<br />

synthesized in the laboratory [129].<br />

Tea saponins: At least three kinds of tea (oolong, green, and black)<br />

have been used as healthy drinks. Tea saponins suppress the increases<br />

in body and parametrial adipose tissue weights and adipocyte<br />

diameters induced by a high-fat diet in mice by inhibiting PL<br />

and also reduce the elevation in plasma triacylglycerol levels after<br />

oral administration of a lipid emulsion. The Ki value of tea saponins<br />

was determined to be 0.25 mg/mL [85]. Thus, the crude saponin<br />

fraction from the flower buds of Chinese tea plant exhibits<br />

accelerating effects on gastrointestinal transit in mice and inhibitory<br />

effects against porcine PL, and three floratheasaponins (A–C)<br />

showed inhibitory effects on serum triglyceride elevation [130].<br />

Triterpenes<br />

Terpenes are the primary constituents of the essential oils of<br />

many types of plants and are classified by the number of terpene<br />

units in the molecule (diterpenes, triterpenes, among others).<br />

The pharmacological relevance of triterpenes has increased during<br />

the last two decades demonstrating multitarget properties<br />

such as wound healing, anti-inflammatory, antibacterial, antiviral,<br />

hepatoprotective, and antitumoral effects, combined with<br />

low toxicity [32]. Triterpene extracts are safe and provide a high<br />

de la Garza AL et al. Natural Inhibitors of… <strong>Planta</strong> Med 2011; 77: 773–785<br />

potential for further pharmaceutical and pharmacological research<br />

[131], some of them inhibiting PL activity.<br />

Betula alba: Bark of birch (Betula alba) contains pentacyclic triterpenes<br />

(l " Fig. 3). This triterpene extract is safe and provides a<br />

high potential for further pharmaceutical and pharmacological<br />

research [32, 131], displaying an inhibitory activity on PL [22].<br />

Clinical Studies about Pancreatic Lipase Inhibitors<br />

!<br />

A number of plants and natural products have been screened for<br />

their PL inhibitory activity but just some of them have gone up to<br />

clinical studies. In this line, only one product derived from natural<br />

compounds (Orlistat) is currently in clinical use, although<br />

others are under investigation. Some of them are Panax ginseng<br />

[132], Camellia sinensis [133], Eleutherococcus senticosus [134],<br />

Malus domestica [135], and Arachis hypogaea [136].<br />

In one study [132], the administration of an extract of Panax ginseng<br />

in humans for 8 weeks decreased circulating cholesterol, triglyceride,<br />

and low-density lipoprotein levels (LDL). Each subject<br />

ingested 2 g of Panax ginseng extract three times a day.<br />

Lee et al. [134] reported that healthy postmenopausal women<br />

treated for 6 months with Eleutherococcus senticosus supplementation<br />

showed significant decreases in serum LDL levels and LDL/<br />

HDL ratios.<br />

In other study, Sugiyama et al. [135] assessed six healthy male<br />

volunteers that followed a high-fat diet with 40 g of fat with 10<br />

control or 10 apple polyphenol (Malus domestica) capsules (600<br />

or 1500 mg). In this study, they demonstrated that apple polyphenols<br />

may prevent obesity in humans by a PL inhibitory mechanism.<br />

Green tea (Camellia sinensis) has been extensively studied in relation<br />

to obesity and other metabolic disorders. Thus, Chantre<br />

and Lairon [133] showed that green tea consumption may be<br />

useful to treat obesity by both, increasing thermogenesis and inhibiting<br />

PL. Thus, a green tea extract showed a direct in vitro inhibition<br />

of gastric and pancreatic lipases [133]. In moderately<br />

obese patients, green tea lowered body weight by stimulating<br />

thermogenesis and increasing energy expenditure when each<br />

subject received 2 times/d a green tea extract (2 capsules morning,<br />

2 capsules midday). Ingestion of 4 capsules containing AR25<br />

(Exolise) provided a daily total intake of 375 mg catechins, of<br />

which 270 mg was epigallocatechin gallate. Also, He et al. [137]<br />

administered daily 8 g of oolong tea for 6 weeks to 102 obese subjects.<br />

As a result, 70% of the obese subjects decreased more than<br />

1 kg in body weight. In vitro studies suggested that the effect of<br />

oolong tea on body weight could be partially attributed to the inhibition<br />

of PL [68].<br />

According to these data, a number of common herbal products<br />

that are being studied in animal (l " Table 3) and human models<br />

for obesity treatment contain different metabolites that act on<br />

lipid digestion and absorption. However, it is very difficult to establish<br />

in in vivo studies whether these antiobesity effects are only<br />

or mainly due to PL activity inhibition. The clinical implications<br />

of this therapeutic approach have yet to be determined.<br />

Conclusions<br />

!<br />

Orlistat is the only drug authorized and present in Europe for the<br />

treatment of obesity within an adequate energy intake, which<br />

acts by inhibiting the lipolytic activity of PL. With the aim of find-


Table 3 Plant extracts that showed in vivo inhibitory activity of pancreatic lipase, doses and effects. IC 50 is indicated when available.<br />

Scientific name Common name IC50 Doses Model Effects References<br />

Aesculus turbinate Japanese horse chestnut IC50 24 mg/mL 0.1–0.5% of diet DIO mice TG plasma levels and body weight gain [153]<br />

Arachis hypogaea Peanut IC50 0.029 µg/mL 1% of diet DIO rats Body weight gain [136]<br />

Camellia sinensis Green, black, oolong tea IC50 0.091 mg/mL 3% of HFD Rats Body weight gain<br />

and visceral fat<br />

[89]<br />

Cassia mimosoides Nomame herba IC50 0.1–0.71 mg/mL 1–3.5% of diet DIO rats Body weight gain [154]<br />

Coffea arabica Coffee 0.5% of standard<br />

diet<br />

Mice Body weight gain [155]<br />

Cyclocarya paliurus Wheel wingnut IC50 9.1 µg/mL 250 mg/kg; VO Mice TG plasma levels and blood glucose levels [156]<br />

Dioscorea nipponica Yam IC50 5–10 mg/mL 5% of HFD Rats TG plasma levels and body weight gain [157]<br />

Eleutherococcus senti- Siberian ginseng IC50 0.22–0.29 mM 12 mg/kg DIO rats Abdominal fat, TG in liver<br />

[158]<br />

cosus<br />

and serum and LDL in serum<br />

Eleutherococcus Sessiloside IC50 0.36–0.75 mg/mL 100–300 mg/kg; Mice TG plasma levels [159]<br />

sessiliflorus<br />

VO<br />

Gardenia jasminoides Cape jasmine IC50 2.1 mg/mL 50 mg/kg/d Mice Body weight gain [118]<br />

Humulus lupulus Common hop 0.2–1.2% (w/w)<br />

of extract<br />

Mice Body weight gain and blood glucose levels [160]<br />

Ilex paraguariensis Yerba mate 0.24% of HFD Rats Body weight gain [99]<br />

Kochia scoparia Burningbush 3% of HFD Mice Body weight gain [150]<br />

Malus domestica Apple IC50 5.6 µg/mL 200 mg/kg; VO Mice TG plasma levels [161]<br />

Myrica spp Bayberry – – TG plasma levels [140]<br />

Nelumbo nucifera Sacred lotus IC50 0.46 mg/mL 5% of diet Mice TG plasma levels and body weight gain [162]<br />

Panax ginseng Ginseng IC50 500 µg/mL 200 mg/kg with<br />

HFD<br />

Rats Body weight gain [109]<br />

Panax japonicus Japanese ginseng 1–3% of diet DIO mice Body weight gain [120]<br />

Platycodi radix Doraji Ki 0.18 mM 70 mg/kg, Sprague Body weight gain [64]<br />

with HFD Dawley rats<br />

Rhodiola rosea Roseroot stonecrop IC50 0.093 mM 150 mg/kg Mice TG plasma levels [141]<br />

Rosmarinus officinalis Rosemary 200 mg/kg HFD Mice Body weight and fat mass [163]<br />

Salacia reticulata Kotala himbutu IC50 264 mg/L 125 mg/kg;<br />

VO HFD<br />

Rats Body weight gain [101]<br />

Salix matsudana Corkscrew willow 5% of HFD Wistar rats Body weight gain [147]<br />

DIO: Diet-induced obesity; HFD: High-fat diet; VO: Via oral. (Daily food intake is approximately: rats: 20 g/day; mice: 4.5 g/day)<br />

ing new compounds more potent or with less secondary effects<br />

than Orlistat, new natural products are being identified and<br />

screened for their PL inhibitory potential. Some of these extracts<br />

are obtained from plants that are rich in polyphenols and saponins<br />

and show inhibitory effects on fat digestion, whereas other<br />

extracts come from algae, fungi, and microorganisms. Thus, natural<br />

products provide an exciting opportunity and promise for the<br />

development of new therapeutic approaches to the treatment of<br />

obesity by blocking the digestion and absorption of dietary lipids,<br />

and constitute a valuable alternative to other pharmacological<br />

agents. Some of the products reviewed in this article show potentially<br />

promising effects for weight control. In particular apple,<br />

green tea, soybean, and ginseng seem to have great potential as<br />

sources of molecules with PL inhibitory activity. For all of them<br />

more data are needed to define effects, optimal dose required,<br />

and mechanism of action, as well as their possible side or toxic<br />

effects.<br />

Thus, there is an urgent need to update the knowledge on the numerous<br />

natural sources that could act as inhibitors of PL in order<br />

to screen them as new potential therapeutic antiobesity agents<br />

with low secondary effects.<br />

Reviews<br />

Acknowledgements<br />

!<br />

The authors thank Línea Especial (LE/97) from the University of<br />

Navarra (Spain) and the CENIT PRONAOS Program (MICINN,<br />

Spain) for financial support. A. L. de la Garza and N. Boqué hold<br />

pre-doctoral grants from Ibercaja. We also acknowledge Marta<br />

Díaz Hernando for her contribution to the figures design.<br />

References<br />

1 Drew B, Dixon A, Dixon J. Obesity management: update on orlistat. Vasc<br />

Health Risk Manag 2007; 3: 817–821<br />

2 Brug J, Crawford D. The obesity pandemic. Is it bad or worse? Eur<br />

J Public Health 2009; 19: 570–571<br />

3 Schrauwen P, Westerterp KR. The role of high-fat diets and physical activity<br />

in the regulation of body weight. Br J Nutr 2000; 84: 417–427<br />

4 Voshol P, Rensen PCN, van Dijk K, Romijn J, Havekes L. Effect of plasma<br />

triglyceride metabolism on lipid storage in adipose tissue: studies using<br />

genetically engineered mouse models. Biochim Biophys Acta 2009;<br />

1791: 479–485<br />

5 Abete I, Astrup A, Martnez JA, Thorsdottir I, Zulet M. Obesity and the<br />

metabolic syndrome: role of different dietary macronutrient distribution<br />

patterns and specific nutritional components on weight loss and<br />

maintenance. Nutr Rev 2010; 68: 214–231<br />

6 Little T, Horowitz M, Feinle-Bisset C. Modulation by high-fat diets of gastrointestinal<br />

function and hormones associated with the regulation of<br />

energy intake: implications for the pathophysiology of obesity. Am<br />

J Clin Nutr 2007; 86: 531–541<br />

7 Rubio M, Gargallo M, Millán A, Moreno B. Drugs in the treatment of obesity:<br />

sibutramine, orlistat and rimonabant. Public Health Nutr 2007;<br />

10: 1200–1205<br />

de la Garza AL et al. Natural Inhibitors of … <strong>Planta</strong> Med 2011; 77: 773–785<br />

781


782<br />

Reviews<br />

8 Moreno D, Ilic N, Poulev A, Brasaemle D, Fried S, Raskin I. Inhibitory effects<br />

of grape seed extract on lipases. Nutrition 2003; 19: 876–879<br />

9 Birari R, Bhutani K. Pancreatic lipase inhibitors from natural sources:<br />

unexplored potential. Drug Discov Today 2007; 12: 879–889<br />

10 Sumantran V. Experimental approaches for studying uptake and action<br />

of herbal medicines. Phytother Res 2007; 21: 210–214<br />

11 Yamagishi S, Matsui T, Ueda S, Fukami K, Okuda S. Clinical utility of acarbose,<br />

an alpha-glucosidase inhibitor in cardiometabolic disorders. Curr<br />

Drug Metab 2009; 10: 159–163<br />

12 Raz I, Eldor R, Cernea S, Shafrir E. Diabetes: insulin resistance and derangements<br />

in lipid metabolism. Cure through intervention in fat<br />

transport and storage. Diabetes Metab Res 2005; 21: 3–14<br />

13 Hosoyama H, Sugimoto A, Suzuki Y, Sakane I, Kakuda T. [Isolation and<br />

quantitative analysis of the alpha-amylase inhibitor in Lagerstroemia<br />

speciosa (L.) Pers. (Banaba)]. Yakugaku Zasshi 2003; 123: 599–605<br />

14 Tsujita T, Takaku T, Suzuki T. Chestnut astringent skin extract, an alphaamylase<br />

inhibitor, retards carbohydrate absorption in rats and humans.<br />

J Nutr Sci Vitaminol 2008; 54: 82–88<br />

15 Tormo MA, Gil-Exojo I, de Tejada AR, Campillo JE. White bean amylase<br />

inhibitor administered orally reduces glycaemia in type 2 diabetic rats.<br />

Br J Nutr 2006; 96: 539–544<br />

16 Bray G, Ryan D. Drug treatment of the overweight patient. Gastroenterology<br />

2007; 132: 2239–2252<br />

17 McClendon K, Riche D, Uwaifo G. Orlistat: current status in clinical therapeutics.<br />

Expert Opin Drug Saf 2009; 8: 727–744<br />

18 Weibel EK, Hadvary P, Hochuli E, Kupfer E, Lengsfeld H. Lipstatin, an inhibitor<br />

of pancreatic lipase, produced by Streptomyces toxytricini.<br />

I. Producing organism, fermentation, isolation and biological activity.<br />

J Antibiot 1987; 40: 1081–1085<br />

19 Viner RM, Hsia Y, Tomsic T, Wong ICK. Efficacy and safety of anti-obesity<br />

drugs in children and adolescents: systematic review and meta-analysis.<br />

Obes Rev 2010; 11: 593–602<br />

20 Heymsfield SB, Segal KR, Hauptman J, Lucas CP, Boldrin MN, Rissanen A.<br />

Effects of weight loss with orlistat on glucose tolerance and progression<br />

to type 2 diabetes in obese adults. Arch Intern Med 2000; 160:<br />

1321–1326<br />

21 Torgerson J, Hauptman J, Boldrin M, Sjstrm L. Xenical in the prevention<br />

of diabetes in obese subjects (XENDOS) study: a randomized study of<br />

orlistat as an adjunct to lifestyle changes for the prevention of type 2<br />

diabetes in obese patients. Diabetes Care 2004; 27: 155–161<br />

22 Slanc P, Doljak B, Kreft S, Lunder M, Janes D, Strukelj B. Screening of selected<br />

food and medicinal plant extracts for pancreatic lipase inhibition.<br />

Phytother Res 2009; 23: 874–877<br />

23 Slanc P, Doljak B, Mlinaric A, Strukelj B. Screening of wood damaging<br />

fungi and macrofungi for inhibitors of pancreatic lipase. Phytother Res<br />

2004; 18: 758–762<br />

24 Bitou N, Ninomiya M, Tsujita T, Okuda H. Screening of lipase inhibitors<br />

from marine algae. Lipids 1999; 34: 441–445<br />

25 Abete I, Parra MD, Zulet MA, Martinez JA. Different dietary strategies for<br />

weight loss in obesity: role of energy and macronutrient content. Nutr<br />

Rev 2006; 19: 5–17<br />

26 Astrup A. The role of dietary fat in the prevention and treatment of<br />

obesity. Efficacy and safety of low-fat diets. Int J Obes 2001; 25: 46–50<br />

27 Lomba A, Martinez JA, Garcia-Daz D, Paternain L, Marti A, Campion J,<br />

Milagro FI. Weight gain induced by an isocaloric pair-fed high fat diet:<br />

a nutriepigenetic study on FASN and NDUFB6 gene promoters. Mol<br />

Genet Metab 2010; 101: 273–278<br />

28 Lomba A, Milagro FI, Garcia-Daz DF, Campion J, Marzo F, Martinez JA.<br />

A high-sucrose isocaloric pair-fed model induces obesity and impairs<br />

NDUFB6 gene function in rat adipose tissue. J Nutrigenet Nutrigenomics<br />

2009; 2: 267–272<br />

29 Hermsdorff HHM, Volp ACP, Bressan J. [Macronutrient profile affects<br />

diet-induced thermogenesis and energy intake]. Arch Latinoam Nutr<br />

2007; 57: 33–42<br />

30 Mobbs C, Mastaitis J, Yen K, Schwartz J, Mohan V, Poplawski M. Low-carbohydrate<br />

diets cause obesity, low-carbohydrate diets reverse obesity:<br />

a metabolic mechanism resolving the paradox. Appetite 2007; 48:<br />

135–138<br />

31 Rolls BJ. The role of energy density in the overconsumption of fat. J Nutr<br />

2000; 130: 268–271<br />

32 Jäger S, Trojan H, Kopp T, Laszczyk M, Scheffler A. Pentacyclic triterpene<br />

distribution in various plants – rich sources for a new group of multipotent<br />

plant extracts. Molecules 2009; 14: 2016–2031<br />

de la Garza AL et al. Natural Inhibitors of… <strong>Planta</strong> Med 2011; 77: 773–785<br />

33 Stevenson E, Astbury N, Simpson E, Taylor M, Macdonald I. Fat oxidation<br />

during exercise and satiety during recovery are increased following a<br />

low-glycemic index breakfast in sedentary women. J Nutr 2009; 139:<br />

890–897<br />

34 Armand M. Lipases and lipolysis in the human digestive tract: where do<br />

we stand? Curr Opin Clin Nutr Metab Care 2007; 10: 156–164<br />

35 Lowe M. The triglyceride lipases of the pancreas. J Lipid Res 2002; 43:<br />

2007–2016<br />

36 Sugiyama H, Akazome Y, Shoji T, Yamaguchi A, Yasue M, Kanda T. Oligomeric<br />

procyanidins in apple polyphenol are main active components<br />

for inhibition of pancreatic lipase and triglyceride absorption. J Agric<br />

Food Chem 2007; 55: 4604–4609<br />

37 Neovius M, Johansson K, Rssner S. Head-to-head studies evaluating efficacy<br />

of pharmaco-therapy for obesity: a systematic review and metaanalysis.<br />

Obes Rev 2008; 9: 420–427<br />

38 Filippatos T, Derdemezis C, Gazi I, Nakou E, Mikhailidis D, Elisaf M. Orlistat-associated<br />

adverse effects and drug interactions: a critical review.<br />

Drug Saf 2008; 31: 53–65<br />

39 Kopelman P, Bryson A, Hickling R, Rissanen A, Rossner S, Toubro S. Cetilistat<br />

(ATL‑962), a novel lipase inhibitor: a 12-week randomized, placebo-controlled<br />

study of weight reduction in obese patients. Int J Obes<br />

2007; 31: 494–499<br />

40 Melia AT, Koss-Twardy SG, Zhi J. The effect of orlistat, an inhibitor of<br />

dietary fat absorption, on the absorption of vitamins A and E in healthy<br />

volunteers. J Clin Pharmacol 1996; 36: 647–653<br />

41 Mutoh M, Nakada N, Matsukuma S, Ohshima S, Yoshinari K, Watanabe J.<br />

Panclicins, novel pancreatic lipase inhibitors. I. Taxonomy, fermentation,<br />

isolation and biological activity. J Antibiot 1994; 47: 1369–1375<br />

42 Nonaka Y, Ohtaki H, Ohtsuka E, Kocha T, Fukuda T, Takeuchi T. Effects of<br />

ebelactone B, a lipase inhibitor, on intestinal fat absorption in the rat.<br />

J Enzym Inhib 1996; 10: 57–63<br />

43 Liu D, Wang F, Liao T, Tang J, Steglich W, Zhu H. Vibralactone: a lipase<br />

inhibitor with an unusual fused beta-lactone produced by cultures of<br />

the basidiomycete Boreostereum vibrans. Org Lett 2006; 8: 5749–5752<br />

44 Weber HK, Zuegg J, Faber K, Pleiss J. Molecular reasons for lipase-sensitivity<br />

against acetaldehyde. J Mol Catal B 1997: 131–138<br />

45 Mizutani T, Inatomi S, Inazu A, Kawahara E. Hypolipidemic effect of<br />

Pleurotus eryngii extract in fat-loaded mice. J Nutr Sci Vitaminol 2010;<br />

56: 48–53<br />

46 Lee J, Song J, Lee J. Optimal extraction conditions of anti-obesity lipase<br />

inhibitor from Phellinus linteus and nutritional characteristics of the<br />

extracts. Mycobiology 2010; 38: 58–61<br />

47 Kim J, Kim H, Park H, Youn S, Choi D, Shin C. Development of inhibitors<br />

against lipase and alpha-glucosidase from derivatives of monascus pigment.<br />

FEMS Microbiol Lett 2007; 276: 93–98<br />

48 Jones A, Gu L, Sorrels C, Sherman D, Gerwick W. New tricks from ancient<br />

algae: natural products biosynthesis in marine cyanobacteria. Curr<br />

Opin Chem Biol 2009; 13: 216–223<br />

49 Bitou N, Ninomiya M, Tsujita T, Okuda H. Screening of lipase inhibitors<br />

from marine algae. Lipids 1999; 34: 441–445<br />

50 Ben Rebah F, Smaoui S, Frikha F, Gargouri Y, Miled N. Inhibitory effects of<br />

tunisian marine algal extracts on digestive lipases. Appl Biochem Biotechnol<br />

2008; 151: 71–79<br />

51 Matsumoto M, Hosokawa M, Matsukawa N, Hagio M, Shinoki A, Nishimukai<br />

M. Suppressive effects of the marine carotenoids, fucoxanthin<br />

and fucoxanthinol on triglyceride absorption in lymph duct-cannulated<br />

rats. Eur J Nutr 2010; 49: 243–249<br />

52 Gholamhoseinian A, Shahouzehi B, Sharifi-far F. Inhibitory effect of<br />

some plant extracts on pancreatic lipase. Int J Pharmacol 2010; 6: 18–<br />

24<br />

53 Gargouri Y, Julien R, Pieroni G, Verger R, Sarda L. Studies on the inhibition<br />

of pancreatic and microbial lipases by soybean proteins. J Lipid Res<br />

1984; 25: 1214–1221<br />

54 Lairon D, Lafont H, Vigne JL, Nalbone G, Lonardi J, Hauton JC. Effects of<br />

dietary fibers and cholestyramine on the activity of pancreatic lipase<br />

in vitro. Am J Clin Nutr 1985; 42: 629–638<br />

55 Tsujita T, Matsuura Y, Okuda H. Studies on the inhibition of pancreatic<br />

and carboxylester lipases by protamine. J Lipid Res 1996; 37: 1481–<br />

1487<br />

56 Tsujita T, Sumiyoshi M, Takaku T, Momsen W, Lowe M, Brockman H. Inhibition<br />

of lipases by epsilon-polylysine. J Lipid Res 2003; 44: 2278–<br />

2286


57 Ivanova M, Panaiotov I, Bois A, GArgouri Y, Verger R. Inhibitiion of pancreatic<br />

lipase by ovalbumin and ‑lactoglobulin A at the air-water interface.<br />

J Colloid Interface Sci 1990; 136: 363–374<br />

58 Gargouri Y, Julien R, Sugihara A, Verger R, Sarda L. Inhibition of pancreatic<br />

and microbial lipases by proteins. Biochim Biophys Acta 1984;<br />

795: 326–331<br />

59 Han LK, Kimura Y, Okuda H. Reduction in fat storage during chitin-chitosan<br />

treatment in mice fed a high-fat diet. Int J Obes 1999; 23: 174–<br />

179<br />

60 Tsujita T, Takaichi H, Takaku T, Sawai T, Yoshida N, Hiraki J. Inhibition of<br />

lipase activities by basic polysaccharide. J Lipid Res 2007; 48: 358–365<br />

61 Knuckles B. Effect of phytate and other myo-inositol phosphate esters<br />

on lipase activity. J Food Sci 1988; 53: 250–252<br />

62 Raghavendra MP, Prakash V. Phenylboronic acid–a potent inhibitor of<br />

lipase from Oryza sativa. J Agric Food Chem 2002; 50: 6037–6041<br />

63 Ninomiya K, Matsuda H, Shimoda H, Nishida N, Kasajima N, Yoshino T.<br />

Carnosic acid, a new class of lipid absorption inhibitor from sage. Bioorg<br />

Med Chem Lett 2004; 14: 1943–1946<br />

64 Zhao HL, Sim J, Shim SH, Ha YW, Kang SS, Kim YS. Antiobese and hypolipidemic<br />

effects of platycodin saponins in diet-induced obese rats:<br />

evidences for lipase inhibition and calorie intake restriction. Int J Obes<br />

2005; 29: 983–990<br />

65 Zhao H, Kim Y. Determination of the kinetic properties of platycodin D<br />

for the inhibition of pancreatic lipase using a 1,2-diglyceride-based<br />

colorimetric assay. Arch Pharm Res 2004; 27: 1048–1052<br />

66 Kwon C, Sohn H, Kim S, Kim J, Son K, Lee J. Anti-obesity effect of Dioscorea<br />

nipponica Makino with lipase-inhibitory activity in rodents. Biosci<br />

Biotechnol Biochem 2003; 67: 1451–1456<br />

67 Won S, Kim S, Kim Y, Lee P, Ryu J, Kim J. Licochalcone A: a lipase inhibitor<br />

from the roots of Glycyrrhiza uralensis. Food Res Int 2007; 40: 1046–<br />

1050<br />

68 Ono Y, Hattori E, Fukaya Y, Imai S, Ohizumi Y. Anti-obesity effect of Nelumbo<br />

nucifera leaves extract in mice and rats. J Ethnopharmacol 2006;<br />

106: 238–244<br />

69 Kim H, Kang M. Screening of Korean medicinal plants for lipase inhibitory<br />

activity. PTR. Phytother Res 2005; 19: 359–361<br />

70 Moreno D, Ripoll C, Ilic N, Poulev A, Aubin C, Raskin I. Inhibition of lipid<br />

metabolic enzymes using Mangifera indica extracts. J Food Agric Environ<br />

2006; 4: 21–26<br />

71 Lei F, Zhang XN, Wang W, Xing DM, Xie WD, Su H. Evidence of anti-obesity<br />

effects of the pomegranate leaf extract in high-fat diet induced<br />

obese mice. Int J Obes 2007; 31: 1023–1029<br />

72 Yang H, Kim Y, Bae H, Cho K, Shin J, Kim N, Kim D. Rhei Rhizoma and<br />

Chunghyuldan inhibit pancreatic lipase. Nat Prod Sci 2003; 9: 38–43<br />

73 Zheng C, Duan Y, Gao J, Ruan Z. Screening for anti-lipase properties of<br />

37 traditional Chinese medicinal herbs. J Chin Med Assoc 2010; 73:<br />

319–324<br />

74 Yoshikawa M, Shimoda H, Nishida N, Takada M, Matsuda H. Salacia reticulata<br />

and its polyphenolic constituents with lipase inhibitory and<br />

lipolytic activities have mild antiobesity effects in rats. J Nutr 2002;<br />

132: 1819–1824<br />

75 Terra X, Montagut G, Bustos M, Llopiz N, Ardvol A, Blad C. Grape-seed<br />

procyanidins prevent low-grade inflammation by modulating cytokine<br />

expression in rats fed a high-fat diet. J Nutr Biochem 2009; 20: 210–<br />

218<br />

76 García-Lafuente A, Guillamón E, Villares A, Rostagno M, Martínez J. Flavonoids<br />

as anti-inflammatory agents: implications in cancer and cardiovascular<br />

disease. Inflamm Res 2009; 58: 537–552<br />

77 Kawaguchi K, Mizuno T, Aida K, Uchino K. Hesperidin as an inhibitor of<br />

lipases from porcine pancreas and Pseudomonas. Biosci Biotechnol Biochem<br />

1997; 61: 102–104<br />

78 He F, Pan Q, Shi Y, Duan C. Biosynthesis and genetic regulation of proanthocyanidins<br />

in plants. Molecules 2008; 13: 2674–2692<br />

79 Quesada H, del Bas JM, Pajuelo D, Daz S, Fernandez-Larrea J, Pinent M.<br />

Grape seed proanthocyanidins correct dyslipidemia associated with a<br />

high-fat diet in rats and repress genes controlling lipogenesis and VLDL<br />

assembling in liver. Int J Obes 2009; 33: 1007–1012<br />

80 Lee Y, Cho E, Tanaka T, Yokozawa T. Inhibitory activities of proanthocyanidins<br />

from persimmon against oxidative stress and digestive enzymes<br />

related to diabetes. J Nutr Sci Vitaminol 2007; 53: 287–292<br />

81 de la Iglesia R, Milagro FI, Campion J, Boque N, Martinez JA. Healthy<br />

properties of proanthocyanidins. Biofactors 2010; 36: 159–168<br />

82 Yamamoto M, Shimura S, Itoh Y, Ohsaka T, Egawa M, Inoue S. Anti-obesity<br />

effects of lipase inhibitor CT‑II, an extract from edible herbs, Nomame<br />

Herba, on rats fed a high-fat diet. Int J Obes 2000; 24: 758–764<br />

Reviews<br />

83 Ikeda I, Tsuda K, Suzuki Y, Kobayashi M, Unno T, Tomoyori H. Tea catechins<br />

with a galloyl moiety suppress postprandial hypertriacylglycerolemia<br />

by delaying lymphatic transport of dietary fat in rats. J Nutr<br />

2005; 135: 155–159<br />

84 Moreno D, Ilic N, Poulev A, Raskin I. Effects of Arachis hypogaea nutshell<br />

extract on lipid metabolic enzymes and obesity parameters. Life<br />

Sci 2006; 78: 2797–2803<br />

85 Han LK, Kimura Y, Kawashima M, Takaku T, Taniyama T, Hayashi T.<br />

Anti-obesity effects in rodents of dietary teasaponin, a lipase inhibitor.<br />

Int J Obes 2001; 25: 1459–1464<br />

86 Tanaka T, Matsuo Y, Kouno I. Chemistry of secondary polyphenols<br />

produced during processing of tea and selected foods. Int J Mol Sci<br />

2009; 11: 14–34<br />

87 Koo S, Noh S. Green tea as inhibitor of the intestinal absorption of lipids:<br />

potential mechanism for its lipid-lowering effect. J Nutr Biochem<br />

2007; 18: 179–183<br />

88 Juhel C, Armand M, Pafumi Y, Rosier C, Vandermander J, Lairon D. Green<br />

tea extract (AR25) inhibits lipolysis of triglycerides in gastric and duodenal<br />

medium in vitro. J Nutr Biochem 2000; 11: 45–51<br />

89 Bose M, Lambert J, Ju J, Reuhl K, Shapses S, Yang C. The major green tea<br />

polyphenol, (−)-epigallocatechin-3-gallate, inhibits obesity, metabolic<br />

syndrome, and fatty liver disease in high-fat-fed mice. J Nutr 2008;<br />

138: 1677–1683<br />

90 Nakai M, Fukui Y, Asami S, Toyoda-Ono Y, Iwashita T, Shibata H. Inhibitory<br />

effects of oolong tea polyphenols on pancreatic lipase in vitro.<br />

J Agric Food Chem 2005; 53: 4593–4598<br />

91 Toyoda-Ono Y, Yoshimura M, Nakai M, Fukui Y, Asami S, Shibata H. Suppression<br />

of postprandial hypertriglyceridemia in rats and mice by<br />

oolong tea polymerized polyphenols. Biosci Biotechnol Biochem<br />

2007; 71: 971–976<br />

92 Ikeda I. Multifunctional effects of green tea catechins on prevention of<br />

the metabolic syndrome. Asia Pac J Clin Nutr 2008; 17: 273–274<br />

93 Kobayashi M, Ichitani M, Suzuki Y, Unno T, Sugawara T, Yamahira T.<br />

Black-tea polyphenols suppress postprandial hypertriacylglycerolemia<br />

by suppressing lymphatic transport of dietary fat in rats. J Agric<br />

Food Chem 2009; 57: 7131–7136<br />

94 Kusano R, Andou H, Fujieda M, Tanaka T, Matsuo Y, Kouno I. Polymerlike<br />

polyphenols of black tea and their lipase and amylase inhibitory<br />

activities. Chem Pharm Bull 2008; 56: 266–272<br />

95 Uchiyama S, Taniguchi Y, Saka A, Yoshida A, Yajima H. Prevention of<br />

diet-induced obesity by dietary black tea polyphenols extract in vitro<br />

and in vivo. Nutrition 2011; 27: 287–292<br />

96 Tanaka K, Tamaru S, Nishizono S, Miyata Y, Tamaya K, Matsui T. Hypotriacylglycerolemic<br />

and antiobesity properties of a new fermented tea<br />

product obtained by tea-rolling processing of third-crop green tea<br />

(Camellia sinensis) leaves and Loquat (Eriobotrya japonica) leaves.<br />

Biosci Biotechnol Biochem 2010; 74: 1606–1612<br />

97 Kurihara H, Shibata H, Fukui Y, Kiso Y, Xu J, Yao X. Evaluation of the hypolipemic<br />

property of Camellia sinensisvar. ptilophylla on postprandial<br />

hypertriglyceridemia. J Agric Food Chem 2006; 54: 4977–4981<br />

98 Guo Y, Wu G, Su X, Yang H, Zhang J. Antiobesity action of a daidzein<br />

derivative on male obese mice induced by a high-fat diet. Nutr Res<br />

2009; 29: 656–663<br />

99 Martins F, Noso T, Porto V, Curiel A, Gambero A, Bastos DHM. Maté tea<br />

inhibits in vitro pancreatic lipase activity and has hypolipidemic effect<br />

on high-fat diet-induced obese mice. Obesity 2010; 18: 42–47<br />

100 Sugimoto S, Nakamura S, Yamamoto S, Yamashita C, Oda Y, Matsuda H.<br />

Brazilian natural medicines. III. structures of triterpene oligoglycosides<br />

and lipase inhibitors from mate, leaves of Ilex paraguariensis.<br />

Chem Pharm Bull 2009; 57: 257–261<br />

101 Yoshikawa M, Shimoda H, Nishida N, Takada M, Matsuda H. Salacia reticulata<br />

and its polyphenolic constituents with lipase inhibitory and<br />

lipolytic activities have mild antiobesity effects in rats. J Nutr 2002;<br />

132: 1819–1824<br />

102 Akase T, Shimada T, Harasawa Y, Akase T, Ikeya Y, Nagai E. Preventive<br />

effects of Salacia reticulata on obesity and metabolic disorders in<br />

TSOD mice. Evid Based Complement Alternat Med, advance online<br />

publication 8 June 2009; doi: 10.1093/ecam/nep052<br />

103 Zhang J, Kang M, Kim M, Kim M, Song J, Lee Y. Pancreatic lipase inhibitory<br />

activity of Taraxacum officinale in vitro and in vivo. Nutr Res Pract<br />

2008; 2: 200–203<br />

104 Zhao J, Pang Y, Dixon R. The mysteries of proanthocyanidin transport<br />

and polymerization. Plant Physiol 2010; 153: 1–18<br />

105 McDougall GJ, Kulkarni NN, Stewart D. Berry polyphenols inhibit pancreatic<br />

lipase activity in vitro. J Food Chem 2009; 115: 193–199<br />

de la Garza AL et al. Natural Inhibitors of … <strong>Planta</strong> Med 2011; 77: 773–785<br />

783


784<br />

Reviews<br />

106 Sparg SG, Light ME, van Staden J. Biological activities and distribution<br />

of plant saponins. J Ethnopharmacol 2004; 94: 219–243<br />

107 Vincken J, Heng L, de Groot A, Gruppen H. Saponins, classification and<br />

occurrence in the plant kingdom. Phytochemistry 2007; 68: 275–297<br />

108 Haralampidis K, Trojanowska M, Osbourn A. Biosynthesis of triterpenoid<br />

saponins in plants. Adv Biochem Eng Biotechnol 2002; 75: 31<br />

109 Karu N, Reifen R, Kerem Z. Weight gain reduction in mice fed Panax<br />

ginseng saponin, a pancreatic lipase inhibitor. J Agric Food Chem<br />

2007; 55: 2824–2828<br />

110 Kimura H, Ogawa S, Jisaka M, Kimura Y, Katsube T, Yokota K. Identification<br />

of novel saponins from edible seeds of Japanese horse chestnut<br />

(Aesculus turbinata Blume) after treatment with wooden ashes and<br />

their nutraceutical activity. J Pharm Biomed Anal 2006; 41: 1657–<br />

1665<br />

111 Hu J, Zhu X, Han L, Saito M, Sun Y, Yoshikawa M. Anti-obesity effects of<br />

escins extracted from the seeds of Aesculus turbinata BLUME (Hippocastanaceae).<br />

Chem Pharm Bull 2008; 56: 12–16<br />

112 Kimura H, Ogawa S, Katsube T, Jisaka M, Yokota K. Antiobese effects of<br />

novel saponins from edible seeds of Japanese horse chestnut (Aesculus<br />

turbinata BLUME) after treatment with wood ashes. J Agric Food<br />

Chem 2008; 56: 4783–4788<br />

113 Song M, Lv N, Kim E, Kwon K, Yoo Y, Kim J. Antiobesity activity of aqueous<br />

extracts of Rhizoma Dioscoreae tokoronis on high-fat diet-induced<br />

obesity in mice. J Med Food 2009; 12: 304–309<br />

114 Li F, Li W, Fu H, Zhang Q, Koike K. Pancreatic lipase-inhibiting triterpenoid<br />

saponins from fruits of Acanthopanax senticosus. Chem Pharm<br />

Bull 2007; 55: 1087–1089<br />

115 Jiang W, Li W, Han L, Liu L, Zhang Q, Zhang S. Biologically active triterpenoid<br />

saponins from Acanthopanax senticosus. J Nat Prod 2006; 69:<br />

1577–1581<br />

116 Yoshizumi K, Hirano K, Ando H, Hirai Y, Ida Y, Tsuji T. Lupane-type saponins<br />

from leaves of Acanthopanax sessiliflorus and their inhibitory<br />

activity on pancreatic lipase. J Agric Food Chem 2006; 54: 335–341<br />

117 Lee I, Lee J, Baek N, Kim D. Antihyperlipidemic effect of crocin isolated<br />

from the fructus of Gardenia jasminoides and its metabolite Crocetin.<br />

Biol Pharm Bull 2005; 28: 2106–2110<br />

118 Sheng L, Qian Z, Zheng S, Xi L. Mechanism of hypolipidemic effect of<br />

crocin in rats: crocin inhibits pancreatic lipase. Eur J Pharmacol<br />

2006; 543: 116–122<br />

119 Zheng Q, Li W, Han L, Koike K. Pancreatic lipase-inhibiting triterpenoid<br />

saponins from Gypsophila oldhamiana. Chem Pharm Bull 2007; 55:<br />

646–650<br />

120 Han L, Zheng Y, Yoshikawa M, Okuda H, Kimura Y. Anti-obesity effects<br />

of chikusetsusaponins isolated from Panax japonicus rhizomes. BMC<br />

Complement Alternat Med 2005; 5: 9<br />

121 Lee Y, Cha B, Yamaguchi K, Choi S, Yonezawa T, Teruya T. Effects of Korean<br />

white ginseng extracts on obesity in high-fat diet-induced obese<br />

mice. Cytotechnology 2010; 62: 367–376<br />

122 Liu W, Zheng Y, Han L, Wang H, Saito M, Ling M. Saponins (Ginsenosides)<br />

from stems and leaves of Panax quinquefolium prevented<br />

high-fat diet-induced obesity in mice. Phytomedicine 2008; 15:<br />

1140–1145<br />

123 Xu B, Han L, Zheng Y, Lee J, Sung C. In vitro inhibitory effect of triterpenoidal<br />

saponins from Platycodi Radix on pancreatic lipase. Arch<br />

Pharm Res 2005; 28: 180–185<br />

124 Zhao H, Kim Y. Determination of the kinetic properties of platycodin D<br />

for the inhibition of pancreatic lipase using a 1,2-diglyceride-based<br />

colorimetric assay. Arch Pharm Res 2004; 27: 968–972<br />

125 Han L, Zheng Y, Xu B, Okuda H, Kimura Y. Saponins from platycodi radix<br />

ameliorate high fat diet-induced obesity in mice. J Nutr 2002; 132:<br />

2241–2245<br />

126 Kim J, Moon K, Seo K, Park K, Choi M, Do G. Supplementation of SK1<br />

from Platycodi radix ameliorates obesity and glucose intolerance in<br />

mice fed a high-fat diet. J Med Food 2009; 12: 629–636<br />

127 Morikawa T, Xie Y, Asao Y, Okamoto M, Yamashita C, Muraoka O. Oleanane-type<br />

triterpene oligoglycosides with pancreatic lipase inhibitory<br />

activity from the pericarps of Sapindus rarak. Phytochemistry<br />

2009; 70: 1166–1172<br />

128 Zheng Q, Koike K, Han L, Okuda H, Nikaido T. New biologically active<br />

triterpenoid saponins from Scabiosa tschiliensis. J Nat Prod 2004; 67:<br />

604–613<br />

129 Guo T, Liu Q, Wang P, Zhang L, Zhang W, Li Y. Facile synthesis of three<br />

bidesmosidic oleanolic acid saponins with strong inhibitory activity<br />

on pancreatic lipase. Carbohydr Res 2009; 344: 1167–1174<br />

de la Garza AL et al. Natural Inhibitors of… <strong>Planta</strong> Med 2011; 77: 773–785<br />

130 Yoshikawa M, Morikawa T, Yamamoto K, Kato Y, Nagatomo A, Matsuda<br />

H. Floratheasaponins A–C, acylated oleanane-type triterpene oligoglycosides<br />

with anti-hyperlipidemic activities from flowers of the<br />

tea plant (Camellia sinensis). J Nat Prod 2005; 68: 1360–1365<br />

131 Jäger S, Laszczyk M, Scheffler A. A preliminary pharmacokinetic study<br />

of betulin, the main pentacyclic triterpene from extract of outer bark<br />

of birch (Betulae alba cortex). Molecules 2008; 13: 3224–3235<br />

132 Kim S, Park K. Effects of Panax ginseng extract on lipid metabolism in<br />

humans. Pharmacol Res 2003; 48: 511–513<br />

133 Chantre P, Lairon D. Recent findings of green tea extract AR25 (Exolise)<br />

and its activity for the treatment of obesity. Phytomedicine<br />

2002; 9: 3–8<br />

134 Lee Y, Chung H, Kwak H, Yoon S. The effects of A. senticosus supplementation<br />

on serum lipid profiles, biomarkers of oxidative stress, and<br />

lymphocyte DNA damage in postmenopausal women. Biochem Biophys<br />

Res Commun 2008; 375: 44–48<br />

135 Sugiyama H, Akazome Y, Shoji T, Yamaguchi A, Yasue M, Kanda T. Oligomeric<br />

procyanidins in apple polyphenol are main active components<br />

for inhibition of pancreatic lipase and triglyceride absorption. J Agric<br />

Food Chem 2007; 55: 4604–4609<br />

136 Moreno D, Ilic N, Poulev A, Raskin I. Effects of Arachis hypogaea nutshell<br />

extract on lipid metabolic enzymes and obesity parameters. Life<br />

Sci 2006; 78: 2797–2803<br />

137 He R, Chen L, Lin B, Matsui Y, Yao X, Kurihara H. Beneficial effects of<br />

oolong tea consumption on diet-induced overweight and obese subjects.<br />

Chin J Integr Med 2009; 15: 34–41<br />

138 Sharma N, Sharma V, Seo S. Screening of some medicinal plants for<br />

anti-lipase activity. J Ethnopharmacol 2005; 97: 453–456<br />

139 Möller N, Roos N, Schrezenmeir J. Lipase inhibitory activity in alcohol<br />

extracts of worldwide occurring plants and propolis. Phytother Res<br />

2009; 23: 585–586<br />

140 Kobayashi K, Ihara S, Kobata A, Itoh K, Kusunoki N, Yoshizaki F. Inhibitory<br />

effect of Myrica bark on lipase activity in mouse plasma and gastrointestinal<br />

tract. J Med Food 2008; 11: 289–293<br />

141 Kobayashi K, Yamada K, Murata T, Hasegawa T, Takano F, Koga K. Constituents<br />

of Rhodiola rosea showing inhibitory effect on lipase activity<br />

in mouse plasma and alimentary canal. <strong>Planta</strong> Med 2008; 74: 1716–<br />

1719<br />

142 Birari R, Roy S, Singh A, Bhutani K. Pancreatic lipase inhibitory alkaloids<br />

of Murraya koenigii leaves. Nat Prod Commun 2009; 4: 1089–<br />

1092<br />

143 Ekanem AP, Wang M, Simon JE, Moreno DA. Antiobesity properties of<br />

two African plants (Afromomum meleguetta and Spilanthes acmella)<br />

by pancreatic lipase inhibition. Phytother Res 2007; 21: 1253–1255<br />

144 Shin J, Han M, Song M, Baek N, Kim D. 5-Hydroxy-7-(4′-hydroxy-3′methoxyphenyl)-1-phenyl-3-heptanone:<br />

a pancreatic lipase inhibitor<br />

isolated from Alpinia officinarum. Biol Pharm Bull 2004; 27: 138–<br />

140<br />

145 Shin J, Han M, Kim D. 3-Methylethergalangin isolated from Alpinia officinarum<br />

inhibits pancreatic lipase. Biol Pharm Bull 2003; 26: 854–<br />

857<br />

146 Jang D, Lee G, Kim J, Lee Y, Kim J, Kim Y. A new pancreatic lipase inhibitor<br />

isolated from the roots of Actinidia arguta. Arch Pharm Res 2008;<br />

31: 666–670<br />

147 Han L, Sumiyoshi M, Zhang J, Liu M, Zhang X, Zheng Y. Anti-obesity action<br />

of Salix matsudana leaves (Part 1). Anti-obesity action by polyphenols<br />

of Salix matsudana in high fat-diet treated rodent animals.<br />

Phytother Res 2003; 17: 1188–1194<br />

148 Tucci SA, Boyland EJ, Halford JC. The role of lipid and carbohydrate digestive<br />

enzyme inhibitors in the management of obesity: a review of<br />

current and emerging therapeutic agents. Diabetes Metab Syndr Obes<br />

2010; 3: 125–143<br />

149 Osbourn A. Saponins in cereals. Phytochemistry 2003; 62: 1–4<br />

150 Han L, Nose R, Li W, Gong X, Zheng Y, Yoshikawa M. Reduction of fat<br />

storage in mice fed a high-fat diet long term by treatment with saponins<br />

prepared from Kochia scoparia fruit. Phytother Res 2006; 20:<br />

877–882<br />

151 Oishi Y, Sakamoto T, Udagawa H, Taniguchi H, Kobayashi-Hattori K,<br />

Ozawa Y. Inhibition of increases in blood glucose and serum neutral<br />

fat by Momordica charantia saponin fraction. Biosci Biotechnol Biochem<br />

2007; 71: 735–740<br />

152 Chang X, Li W, Jia Z, Satou T, Fushiya S, Koike K. Biologically active triterpenoid<br />

saponins from Ardisia japonica. J Nat Prod 2007; 70: 179–<br />

187


153 Kimura H, Ogawa S, Katsube T, Jisaka M, Yokota K. Antiobese effects of<br />

novel saponins from edible seeds of Japanese horse chestnut (Aesculus<br />

turbinata BLUME) after treatment with wood ashes. J Agric Food<br />

Chem 2008; 56: 4783–4788<br />

154 Yamamoto M, Shimura S, Itoh Y, Ohsaka T, Egawa M, Inoue S. Anti-obesity<br />

effects of lipase inhibitor CT‑II, an extract from edible herbs, Nomame<br />

Herba, on rats fed a high-fat diet. Int J Obes 2000; 24: 758–764<br />

155 Shimoda H, Seki E, Aitani M. Inhibitory effect of green coffee bean extract<br />

on fat accumulation and body weight gain in mice. BMC Complement<br />

Alternat Med 2006; 6: 9<br />

156 Kurihara H, Asami S, Shibata H, Fukami H, Tanaka T. Hypolipemic effect<br />

of Cyclocarya paliurus (Batal) Iljinskaja in lipid-loaded mice. Biol<br />

Pharm Bull 2003; 26: 383–385<br />

157 Kwon C, Sohn H, Kim S, Kim J, Son K, Lee J. Anti-obesity effect of Dioscorea<br />

nipponica Makino with lipase-inhibitory activity in rodents.<br />

Biosci Biotechnol Biochem 2003; 67: 1451–1456<br />

158 Cha Y, Rhee S, Heo Y. Acanthopanax senticosus extract prepared from<br />

cultured cells decreases adiposity and obesity indices in C57BL/6J<br />

mice fed a high fat diet. J Med Food 2004; 7: 422–429<br />

Reviews<br />

159 Yoshizumi K, Murota K, Watanabe S, Tomi H, Tsuji T, Terao J. Chiisanoside<br />

is not absorbed but inhibits oil absorption in the small intestine<br />

of rodents. Biosci Biotechnol Biochem 2008; 72: 1126–1129<br />

160 Yajima H, Noguchi T, Ikeshima E, Shiraki M, Kanaya T, Tsuboyama-Kasaoka<br />

N. Prevention of diet-induced obesity by dietary isomerized<br />

hop extract containing isohumulones, in rodents. Int J Obes 2005;<br />

29: 991–997<br />

161 Sugiyama H, Akazome Y, Shoji T, Yamaguchi A, Yasue M, Kanda T. Oligomeric<br />

procyanidins in apple polyphenol are main active components<br />

for inhibition of pancreatic lipase and triglyceride absorption. J Agric<br />

Food Chem 2007; 55: 4604–4609<br />

162 Ono Y, Hattori E, Fukaya Y, Imai S, Ohizumi Y. Anti-obesity effect of<br />

Nelumbo nucifera leaves extract in mice and rats. J Ethnopharmacol<br />

2006; 106: 238–244<br />

163 Harach T, Aprikian O, Monnard I, Moulin J, Membrez M, Bolor J. Rosemary<br />

(Rosmarinus officinalis L.) leaf extract limits weight gain and liver<br />

steatosis in mice fed a high-fat diet. <strong>Planta</strong> Med 2010; 76: 566–571<br />

de la Garza AL et al. Natural Inhibitors of … <strong>Planta</strong> Med 2011; 77: 773–785<br />

785


786<br />

Original Papers<br />

Mulberroside A Possesses Potent<br />

Uricosuric and Nephroprotective Effects<br />

in Hyperuricemic Mice<br />

Authors Cai-Ping Wang 1 , Yemin Wang 1,2 , Xing Wang 1 , Xian Zhang 1 , Jian-Feng Ye 3 , Lin-Shui Hu 4 , Ling-Dong Kong 1<br />

Affiliations<br />

Key words<br />

l " mulberroside A<br />

l " hyperuricemia<br />

l " uricosuric<br />

l " nephroprotective<br />

l " organic anion transporters<br />

l " organic cation/carnitine<br />

transporters<br />

l " Morus alba L.<br />

l " Moraceae<br />

received July 23, 2010<br />

revised October 13, 2010<br />

accepted Nov. 9, 2010<br />

Bibliography<br />

DOI http://dx.doi.org/<br />

10.1055/s-0030-1250599<br />

Published online December 10,<br />

2010<br />

<strong>Planta</strong> Med 2011; 77: 786–794<br />

© Georg Thieme Verlag KG<br />

Stuttgart · New York ·<br />

ISSN 0032‑0943<br />

Correspondence<br />

Ling-Dong Kong<br />

School of Life Sciences<br />

Nanjing University<br />

No. 22, Hankou Road<br />

Nanjing 210093<br />

P. R. China<br />

Phone: + 86 2583 5946 91<br />

Fax: +862583594691<br />

kongld@nju.edu.cn<br />

1 State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, P. R. China<br />

2 Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA<br />

3 Zhejiang Institute of Traditional Chinese Medicine & Natural Drug, Hangzhou, P.R. China<br />

4 Zhejiang Conba Pharmaceutical Co., Ltd., Lanxi, P. R. China<br />

Abstract<br />

!<br />

Mulberroside A is a major stilbene glycoside of<br />

Morus alba L. (Moraceae), which is effectively<br />

used for the treatment of hyperuricemia and gout<br />

in traditional Chinese medicine. We examined<br />

whether mulberroside A had effects on renal urate<br />

underexcretion and dysfunction in oxonateinduced<br />

hyperuricemic mice and investigated the<br />

potential uricosuric and nephroprotective mechanisms<br />

involved. Mulberroside A at 10, 20, and<br />

40 mg/kg decreased serum uric acid levels and increased<br />

urinary urate excretion and fractional excretion<br />

of uric acid in hyperuricemic mice. Simultaneously,<br />

it reduced serum levels of creatinine<br />

and urea nitrogen (10–40 mg/kg), urinary N-acetyl-β-D-glucosaminidase<br />

activity (10–40 mg/kg),<br />

β2-microglobulin (10–40 mg/kg) and albumin<br />

(20–40 mg/kg), and increased creatinine clearance<br />

(10–40 mg/kg) in hyperuricemic mice. Furthermore,<br />

mulberroside A downregulated mRNA<br />

and protein levels of renal glucose transporter 9<br />

(mGLUT9) and urate transporter 1 (mURAT1),<br />

and upregulated mRNA and protein levels of renal<br />

organic anion transporter 1 (mOAT1) and organic<br />

Introduction<br />

!<br />

The prevalence of gout is rapidly increasing<br />

worldwide [1, 2]. Hyperuricemia is the first clinical<br />

manifestation of gout and contributes to the<br />

development of renal dysfunction [2]. In the majority<br />

of patients with primary gout, hyperuricemia<br />

results from the decreased excretion of renal<br />

uric acid [1]. Recent studies have demonstrated a<br />

pivotal role of organic ion transporters of the<br />

SLC2A and SLC22A gene families in renal urate excretion<br />

and function [3–7]. In renal proximal tubules,<br />

glucose transporter 9 (GLUT9, SLC2A9) and<br />

urate transporter 1 (URAT1, SLC22A12) at the<br />

brush border membranes, and organic anion<br />

Wang C-P et al. Mulberroside A Possesses… <strong>Planta</strong> Med 2011; 77: 786–794<br />

cation and carnitine transporters (mOCT1,<br />

mOCT2, mOCTN1, and mOCTN2) in hyperuricemic<br />

mice. This is the first study demonstrating<br />

that mulberroside A exhibits uricosuric and nephroprotective<br />

effects mediated in part by cooperative<br />

attenuation of the expression alterations of<br />

renal organic ion transporters in hyperuricemic<br />

mice. These data suggest that mulberroside A<br />

may be a new drug candidate for the treatment<br />

of hyperuricemia with renal dysfunction.<br />

Abbreviations<br />

!<br />

GAPDH: glyceraldehyde-3-phosphate<br />

dehydrogenase<br />

HRP: horseradish peroxidase<br />

IgG: immunoglobulin G<br />

Scr: serum creatinine level<br />

Ucr: urinary creatinine level<br />

Supporting information available online at<br />

http://www.thieme-connect.de/ejournals/toc/<br />

plantamedica<br />

transporter 1 (OAT1, SLC22A6) at the basolateral<br />

membranes, are potential molecular components<br />

in effective reabsorptive and secretory mechanisms<br />

of renal urate excretion [5–7]. Organic cation<br />

and carnitine transporters (OCTs and OCTNs)<br />

are responsible for renal uptake and excretion of<br />

organic cations [3]. The altered expressions of<br />

the basolateral membrane OCT1 (SLC22A1) and<br />

OCT2 (SLC22A2), and of the brush border membrane<br />

OCTN1 (SLC22A4) and OCTN2 (SLC22A5)<br />

are suggested to be associated with renal dysfunction<br />

[8–11].<br />

Hypouricemic drugs including uricosurics have<br />

some side effects, which restrict their uses in patients,<br />

such as inapplicability of probenecid in pa-


tients with renal calculi and gastrointestinal problems [12].<br />

Therefore, there is an urgent need for novel hypouricemic agents.<br />

Mulberroside A is a major stilbene glycoside isolated from Morus<br />

genus plants including M. alba L. (Moraceae) [13], which is<br />

widely used in many traditional Chinese medicine prescriptions<br />

to treat gout, arthritis, and rheumatism through purging diuresis<br />

and relieving edema. However, it is not clear whether mulberroside<br />

A plays a causal role in the treatment of hyperuricemia.<br />

In the present study, we evaluated the effects of mulberroside A<br />

on serum uric acid levels and renal urate excretion in oxonate-induced<br />

hyperuricemic mice, a model used to investigate therapies<br />

of hypouricemic agents [14, 15]. We also explored the potential<br />

molecular mechanisms of its uricosuric action by determining<br />

mRNA and protein levels of renal mGLUT9, mURAT1, and mOAT1<br />

in hyperuricemic mice treated with mulberroside A. In addition,<br />

the present study was conducted to assess renal function by measuring<br />

serum levels of creatinine (Cr) and urea nitrogen (BUN),<br />

creatinine clearance (Ccr), and urinary biochemical markers of<br />

renal damage, N-acetyl-β-D-glucosaminidase (NAG) activity, β2microglobulin<br />

(β2-MG), and albumin levels. Expression levels of<br />

renal mOCT1, mOCT2, mOCTN1, and mOCTN2 were simultaneously<br />

detected to investigate the possible mechanisms underlying<br />

kidney function improvement in hyperuricemic mice with<br />

mulberroside A treatment.<br />

Materials and Methods<br />

!<br />

Reagents<br />

Mulberroside A was purified from twigs of Morus alba L., which<br />

were purchased from Medicinal Materials Co. of Jiangsu Province,<br />

R.P. China, and authenticated by Prof. J. W. Cheng, Nanjing University<br />

of Traditional Chinese Medicine. A voucher specimen<br />

(NU-20010) of the herb was deposited in the Herbarium of the<br />

Nanjing University, Nanjing. Uric acid (purity > 99%) and potassium<br />

oxonate (purity > 97%) were purchased from Sigma. Probenecid<br />

(purity > 98.5%) was purchased from SanJiang Pharmaceutical<br />

Chemical Co., Ltd. The purified mulberroside A (purity > 97%,<br />

proved by HPLC) was used. Materials for CC were silica gel (45–<br />

75 µm; Qingdao Haiyang Chemical Co., Ltd.) and Sephadex LH-<br />

20 (40–70 µm; Pharmacia). Assay kits of creatinine, albumin,<br />

BUN, and NAG were purchased from Jiancheng Biotech. ELISA assay<br />

kit of β 2-MG was purchased from R & D. All other reagents<br />

were local analytical grade products.<br />

Extraction and isolation<br />

The air-dried twigs (6500 g) were broken into small pieces and<br />

successively extracted three times with 60% ethanol (45 500 mL)<br />

under reflux, and the solvent was removed under reduced pressure<br />

at 50°C to afford ethanol extract (563.8 g). The extract was<br />

dissolved in deionized water (1500 mL) and re-extracted to obtain<br />

a soluble portion by phase separation using petroleum ether<br />

(5 × 800 mL, 65.9 g), ethyl acetate (3 × 600 mL, 99.8 g), and n-butanol<br />

(3 × 500 mL, 71.6 g). The n-butanol soluble portion (71.6 g)<br />

was subjected to separation on silica gel column (112 × 10.8 cm)<br />

and successively eluted with a solvent system of chloroform/<br />

methanol/H 2O (9 : 1:1, 8 : 2:1, 7:3:1, 6:4:1, v/v/v) with a flow<br />

rate of 8 mL/min to yield one hundred and five fractions. The fractions<br />

so obtained were recombined into fifteen portions on the<br />

basis of bioassay guided TLC fractionation. The fraction 13 was<br />

further purified by gel filtration on a Sephadex LH-20 column<br />

(100 × 2.7 cm) eluted with methanol for isolation of mulberroside<br />

Original Papers<br />

Fig. 1 Structure of<br />

mulberroside A isolated<br />

from the twigs of Morus<br />

alba L.<br />

A (6.7 g), which was identified by comparing experimental data<br />

for MS, 1 H-NMR, and 13 C-NMR spectral data with those of the authentic<br />

mulberroside A. The structure of mulberroside A is<br />

shown in l " Fig. 1.<br />

Animals<br />

Male Kun-Ming mice (20 ± 2 g) were purchased from Henan Experimental<br />

Animal Center. They were housed in plastic cages<br />

with free access to food and tap water at room temperature<br />

(22 ± 2°C) with relative humidity of 55 ± 5%, under a normal 12h/12-h<br />

light/dark schedule with the lights on at 7:00 a.m. They<br />

were allowed at least 1 week to adapt to the environment before<br />

the experiment started. All animal procedures and the experimental<br />

protocols were approved by the Institutional Animal Care<br />

Committee at the Nanjing University and the China Council on<br />

Animal Care at the Nanjing University.<br />

Experimental protocols<br />

In the present study, hyperuricemia was induced by the uricase<br />

inhibitor, potassium oxonate, according to a previous report<br />

[16]. Probenecid was used as a positive control. All doses were<br />

expressed as mg/kg body weight of the respective drugs. Food,<br />

but not water, was withdrawn from the animals 1 h prior to the<br />

administration. Sixty-four mice were divided into a normal group<br />

receiving water (normal-vehicle) and 250 mg/kg oxonate-treated<br />

groups receiving water (hyperuricemia-vehicle), 5, 10, 20, and<br />

40 mg/kg mulberroside A, 50 and 100 mg/kg probenecid, respectively.<br />

Oxonate was administered by oral gavage once daily at<br />

8:00 a. m. for 7 consecutive days. Mulberroside A and probenecid<br />

were orally initiated at 9:00 a. m. on the day after oxonate was<br />

given, respectively. The dose volume 10 mL/kg body weight was<br />

kept constant for all treatment groups.<br />

Blood, urine, and tissue sample collection: During the 6th day of<br />

treatment, normal and hyperuicemic mice were housed in metabolic<br />

cages with free access to standard chow and tap water. The<br />

24-h urine was collected, recorded and centrifuged at 2000 × g for<br />

10 min to remove particulate contaminants. Whole blood samples<br />

were collected 1 h after final administration on the 7th day<br />

by tail vein bleeding, and then centrifuged at 10000 × g for 5 min<br />

to obtain the serum. Serum and urine samples were subjected to<br />

biochemical assays on the day of collection. Simultaneously, animals<br />

were killed to collect kidney cortex tissues that were then<br />

stored at −80°C for assays.<br />

Measurement of urine and blood biospecimens: Uric acid levels in<br />

serum (Sur) and urine (Uur) were determined as described previously<br />

[17]. Excretion of urate in 24 h was calculated using the formula:<br />

volume of urine in 24 h × Uur. Creatinine levels in serum<br />

(Scr) and urine (Ucr) were determined spectrophotometrically<br />

using a standard diagnostic kit. Fractional excretion of uric acid<br />

(FEUA) was calculated using the formula: FEUA = (Uur × Scr)/<br />

(Sur × Ucr) × 100, expressed as a percentage. Creatinine clearance<br />

was calculated using the formula: Ccr = (Uur × Volume of urine in<br />

Wang C-P et al. Mulberroside A Possesses … <strong>Planta</strong> Med 2011; 77: 786–794<br />

787


788<br />

Original Papers<br />

Table 1 Antibodies used for Western blot analyses.<br />

Company Description Catalog<br />

number<br />

SaiChi Biotech Rabbit mGLUT9 antibody 001052-R<br />

Rabbit mURAT1 antibody 001046-R<br />

Rabbit mOAT1 antibody 001019-R<br />

Rabbit mOCT1 antibody 001017-R<br />

Rabbit mOCT2 antibody 001018-R<br />

Alpha Diagnostic International, Inc. Rabbit mOCTN1 antibody OCTN11-A<br />

Rabbit mOCTN2 antibody OCTN21-A<br />

KangChen BIO-TECH Mouse mGAPDH<br />

monoclonal antibody<br />

KC-5G5<br />

Cell Signaling Technology, Inc. Rabbit mNa + -K + -ATPase<br />

antibody<br />

#3010S<br />

Jingmei Biotech Goat anti-rabbit-IgG-HRP SB-200<br />

24 h)/(Scr × 24 × 60). BUN levels were determined using a standard<br />

diagnostic kit. Urine NAG was measured by the enzymatic<br />

colorimetric method. One unit (U) of NAG enzyme activity was<br />

defined as 1 µmol p-nitrophenol hydrolyzed per minute from 1 L<br />

of reaction sample with substrate at 37°C. Urine β2-MG was measured<br />

using an ELISA kit. Urine albumin was measured by the<br />

bromocresol green colorimetry method. Urinary NAG, β2-MG,<br />

and albumin levels were adjusted with creatinine into NAG/creatinine<br />

(U/g cr), β2-MG/creatinine (ng/g cr), and albumin/creatinine<br />

(g/g cr), respectively.<br />

Histological analysis: Mouse kidneys were removed and immediately<br />

fixed in carnoy fixative (ethanol:chloroform : acetic acid =<br />

6: 3: 1) for 1 day at room temperature, and preserved in 70%<br />

ethanol. Renal biopsies were dehydrated with a graded series of<br />

alcohol and embedded in paraffin. Specimens were cut in 7-µmthick<br />

sections on a rotary microtome and mounted on APEScoated<br />

glass slides. Sections were deparaffinized in xylene, rehydrated<br />

in decreasing concentrations of alcohol in water and<br />

stained with hematoxylin-eosin reagent (Sigma Chemicals Co.).<br />

The slides were mounted with neutral balsam.<br />

Western blot analysis: Mouse kidney tissue samples for Western<br />

blot analyses of mGLUT9, mURAT1, mOAT1, mOCT1, mOCT2,<br />

mOCTN1, and mOCTN2 were prepared as described previously<br />

[18]. The antibodies are listed in l " Table 1.<br />

Semiquantitative reverse transcription-polymerase chain reaction<br />

(RT-PCR): Mouse kidney tissue samples for RT-PCR analyses of<br />

mGLUT9, mURAT1, mOCT1, mOCT2, mOCTN1, and mOCTN2<br />

were prepared as described previously [18]. Sequences of primers,<br />

appropriate annealing temperature, and length of products<br />

are summarized in l " Table 2.<br />

Statistical analysis<br />

All data were expressed as mean ± standard error of the mean<br />

(SEM). Statistical analysis was performed using a one-way analysis<br />

of variance (ANOVA) followed by Dunnettʼs multiple comparison<br />

tests to determine the level of significance. A value of p < 0.05<br />

was considered statistically significant. Figures were obtained by<br />

the Statistical Analysis System (GraphPad Prism 4, GraphPad<br />

Software, Inc.).<br />

Supporting information<br />

1 13 H-NMR, C-NMR, MS, and HPLC spectra of mulberroside A are<br />

available as Supporting Information.<br />

Results<br />

!<br />

Serum uric acid levels of oxonate-induced hyperuricemic mice<br />

were significantly decreased in a dose-dependent manner by a<br />

one-week treatment with mulberroside A compared with the hyperuricemia-vehicle<br />

group (l " Fig. 2A). Twenty or 40 mg/kg mulberroside<br />

A lowered the serum uric acid levels of hyperuricemic<br />

mice to a comparable value obtained from normal-vehicle mice.<br />

Treatment of hyperuricemic mice with 100 mg/kg probenecid, a<br />

widely used uricosuric drug, dramatically reduced serum uric acid<br />

levels compared with vehicle-treated hyperuricemic mice. Simultaneously,<br />

urine volume did not differ among all experimental<br />

groups (l " Fig. 2B). Mulberroside A at 10, 20, and 40 mg/kg significantly<br />

increased urinary urate excretion in 24 h (l " Fig. 2 C),<br />

resulting in a remarkable elevation of FEUA (20–40 mg/kg)<br />

(l " Fig. 2D), and the highest dose completely reversed FEUA alteration<br />

of hyperuricemic mice to normal. Probenecid at 50 and<br />

100 mg/kg also showed uricosuric effects in hyperuricemic mice.<br />

To evaluate renal function in hyperuricemic mice treated with<br />

mulberroside A, we detected serum creatinine and BUN levels,<br />

creatinine clearance, urinary NAG activity, β2-MG and albumin<br />

levels. Oxonate induced NAG hyperactivity as well as β2-MG and<br />

albumin level elevation in mice. Mulberroside A significantly decreased<br />

serum creatinine and BUN levels (10–40 mg/kg) (l " Fig. 3A<br />

and B), urinary NAG activity (10–40 mg/kg) (l " Fig. 3 D), β2-MG<br />

(10–40 mg/kg) and albumin (20–40 mg/kg) levels (l " Fig. 3 E and<br />

F), and increased creatinine clearance (10–40 mg/kg) (l " Fig. 3 C)<br />

in hyperuricemic mice compared with the hyperuricemia-vehicle<br />

group. Moreover, 40 mg/kg mulberroside A completely reversed<br />

abnormalities of these renal function indicators in hyperuricemic<br />

mice to normal. Five mg/kg mulberroside A had no effect on these<br />

changes in this model. Probenecid also decreased serum creatinine<br />

and BUN levels (100 mg/kg) (l " Fig. 3 A and B), urinary NAG ac-<br />

Table 2 Summary of gene-specific PCR primer sequences, length of production, and the appropriate annealing temperature used in the experiments.<br />

Description Genebank Sense primer (5′ → 3′) Antisense primer (5′ → 3′) Product Tm Numbers<br />

size (bp) (°C) of thermal<br />

cycle<br />

mGLUT9 NM_001012363 CTCATTGTGGGACGGTTCA GCTACTTCGTCCTCGGTA 316 56 30<br />

mURAT1 NM_009203 GCTACCAGAATCGGCACGCT CACCGGGAAGTCCACAATCC 342 58 30<br />

mOAT1 NM_008766 ACGGGAAACAAGAAGAGGG AAGAGAGGTATGGAGGGGTAG 580 56 29<br />

mOCT1 NM_009202 ACATCCATGTTGCTCTTTCG TTGCTCCATTATCCTTACCG 315 56 29<br />

mOCT2 NM_013667 ACAGGTTTGGGCGGAAGT CACCAGAAATAGAGCAGGAAG 331 56 29<br />

mOCTN1 NM_019687 TGTTCTTCGTAGGCGTTCT TGGAATAAACCACCACAGG 392 53.3 30<br />

mOCTN2 NM_011396 TCTACGAAGCCTCAGTTGC ATTCCTTTGACCCTTAGCAT 623 53.3 30<br />

mGAPDH NM_008084 TGAGGCCGGTGCTGAGTATGT CAGTCTTCTGGGTGGCAGTGAT 299 58 35<br />

Wang C-P et al. Mulberroside A Possesses… <strong>Planta</strong> Med 2011; 77: 786–794


Fig. 2 Effects of mulberroside A and probenecid on serum uric acid levels<br />

(A), urine volume (B), urinary urate excretion in 24 h (C), and fractional excretion<br />

of uric acid (FEUA) (D) in oxonate-induced hyperuricemia in mice. Experiments<br />

were performed as described in the Materials and Methods sec-<br />

tivity, and β2-MG levels (50–100 mg/kg) (l " Fig. 3D and E), and increased<br />

creatinine clearance (50–100 mg/kg) (l " Fig. 3C) but<br />

failed to affect urinary albumin levels (l " Fig. 3F) in hyperuricemic<br />

mice compared with the hyperuricemia-vehicle group. On<br />

the other hand, vacuolar and granular degeneration could be<br />

seen in renal tubular epithelial cells of hyperuricemic mice, and<br />

tubular lumen was dilated without visible urate crystals<br />

(l " Fig. 4B). Mulberroside A reduced vacuolar and granular degeneration<br />

in the same parts of hyperuricemic mice (l " Fig. 4C–F).<br />

The degeneration almost disappeared after the highest dose treatment<br />

of mulberroside A (l " Fig. 4 F). Probenecid had similar effects<br />

in this model (l " Fig. 4G, H).<br />

To elucidate whether mulberroside A promotes renal urate excretion<br />

in hyperuricemia mediated by regulating expression of renal<br />

mGLUT9, mURAT1, and mOAT1, we examined their protein and<br />

mRNA levels. Mulberroside A at 10, 20, and 40 mg/kg significantly<br />

suppressed the elevated protein and mRNA levels of renal<br />

mGLUT9 and mURAT1 in hyperuricemic mice compared with the<br />

hyperuricemia-vehicle group (l " Fig. 5 A,B and Fig. 6 A, B). It also<br />

remarkably upregulated the expression levels of renal mOAT1<br />

protein (10, 20, and 40 mg/kg) and mRNA (20 and 40 mg/kg)<br />

(l " Fig. 5C and Fig. 6 C) in this model. One hundred mg/kg probenecid<br />

reduced protein and mRNA levels of mURAT1 and upregulated<br />

mOAT1 expression, which was in agreement with other<br />

reports that the uricosuric activity of probenecid was associated<br />

with its regulatory roles on hURAT1 in Xenopus oocyte [5] and<br />

hURAT1 and hOAT1 in human proximal tubule cells [19]. In line<br />

Original Papers<br />

tion. Data are expressed as the mean ± SEM for 8 mice; ### p < 0.001 vs. normal-vehicle<br />

animals; * p < 0.05, ** p < 0.01, and *** p < 0.001 vs. hyperuricemia-vehicle<br />

animals.<br />

with the previous study in Xenopus laevis oocytes [4], probenecid<br />

treatment did not alter mGLUT9 in hyperuricemic mice<br />

(l " Fig. 5A and Fig. 6 A).<br />

Next, we examined the effects of mulberroside A on expression<br />

profiles of renal mOCT1, mOCT2, mOCTN1, and mOCTN2 to elucidate<br />

the potential mechanisms underlying renal function improvement<br />

in hyperuricemic mice. Downregulation of renal<br />

mOCT1, mOCT2, mOCTN1, and mOCTN2 protein levels in hyperuricemic<br />

mice was attenuated significantly by the treatment with<br />

10, 20, and 40 mg/kg mulberroside A compared with the hyperuricemia-vehicle<br />

group (l " Fig. 5D–G). Mulberroside A also remarkably<br />

increased mRNA levels of renal mOCT1 (5–40 mg/kg),<br />

mOCT2 (10–40 mg/kg), mOCTN1 (20–40 mg/kg), and mOCTN2<br />

(20–40 mg/kg) in this model (l " Fig. 6 D–G). Probenecid treatment<br />

elevated protein and mRNA levels of these renal transporters in<br />

almost all cases with the exception of the 50 mg/kg concentration<br />

of probenecid which failed to alter protein and mRNA levels of renal<br />

mOCT1 and mRNA levels of renal mOCTN1 in hyperuricemic<br />

mice.<br />

Discussion<br />

!<br />

The present study showed that mulberroside A, a major stilbene<br />

glycoside of M. alba, significantly reduced serum uric acid levels,<br />

increased urinary urate excretion, resulting in a significant FEUA<br />

elevation in oxonate-induced hyperuricemic mice. Hyperurice-<br />

Wang C-P et al. Mulberroside A Possesses … <strong>Planta</strong> Med 2011; 77: 786–794<br />

789


790<br />

Original Papers<br />

Fig. 3 Effects of mulberroside A and probenecid on serum creatinine (A)<br />

and urea nitrogen (BUN) (B) levels, creatinine clearance (Ccr) (C), urinary Nacetyl-β-D-glucosaminidase<br />

(NAG) activity (D), β2-microglobulin (β2-MG) (E)<br />

and albumin (F) levels in oxonate-induced hyperuricemia in mice. Data are<br />

mia is an increased risk for the progression of renal disease [20,<br />

21]. The altered urinary enzyme activity and protein levels are<br />

generally regarded as indicators of renal injury [22,23]. The<br />

present study found that hyperuricemic mice developed an elevation<br />

of urinary NAG activity, β 2-MG, and albumin levels, accompanied<br />

with decreased creatinine clearance and increased<br />

serum creatinine and BUN levels, confirming renal function impairment<br />

in hyperuricemic mice. Mulberroside A was found to attenuate<br />

oxonate-induced renal function impairment in mice. In<br />

line with biochemical markers of renal function, mulberroside A<br />

could reduce kidney histological changes induced by oxonate in<br />

Wang C-P et al. Mulberroside A Possesses… <strong>Planta</strong> Med 2011; 77: 786–794<br />

expressed as the mean ± SEM for 8 mice; ## p < 0.01, ### p < 0.001 vs. normalvehicle<br />

animals; * p < 0.05, ** p < 0.01, and *** p < 0.001 vs. hyperuricemiavehicle<br />

animals.<br />

mice. This is the first study demonstrating that mulberroside A<br />

has uricosuric and nephroprotective effects. These biochemical<br />

data may provide evidence for the clinical application of M. alba<br />

in the treatment of gout and hyperuricemia in traditional Chinese<br />

medicine.<br />

Uric acid level is a key factor for the prevention and treatment of<br />

hyperuricemia and gout. hGLUT9 has been recently found to reduce<br />

urate reabsorption, resulting in serum uric acid reduction<br />

and FEUA elevation [24, 25]. mGLUT9 at the apical and basolateral<br />

membranes of distal convoluted tubules may be related to renal<br />

urate reabsorption [26]. URAT1, as a mediator of urate reabsorp-


tion, plays a pivotal role in uric acid homeostasis [5]. Mutations in<br />

hURAT1 are associated with the pathogenesis of primary hyperuricemia<br />

and gout [27]. Renal mURAT1 is also involved in renal<br />

urate reabsorption [28]. Renal OAT1 is associated with renal urate<br />

secretion [3, 7]. Suppression of renal rOAT1 was observed in<br />

oxonic acid and uric acid-induced hyperuricemic rats [14].<br />

OAT1-knockout mice exhibit reduced secretion of urate in the<br />

kidney [29]. In addition, patients with renal diseases show low<br />

renal OAT1 expression [30]. Reduction of renal rOCT1 and rOCT2<br />

was observed in streptozotocin-induced diabetes of rats with<br />

kidney dysfunction [8]. Down-expression of rOCT2 was also observed<br />

in rats with chronic renal failure [9] and hyperuricemia<br />

[14]. Defective mutations in hOCTN2 may impair reabsorption<br />

and secretion of organic cations [11]. In the present study, we<br />

demonstrated that oxonate-induced hyperuricemic mice developed<br />

upregulation of renal mGLUT9 and mURAT1, and downregulation<br />

of renal mOAT1, mOCT1, mOCT2, mOCTN1, and<br />

mOCTN2. It has been noted that hOCT2 mediates the basolateral<br />

membrane uptake of creatinine into HEK293 cells [31] and human<br />

tubular cell monolayers [32]. Clearance of both creatinine<br />

and urate in mutant hURAT1 carriers is significantly higher than<br />

that in healthy subjects [33]. Thus, the distorted expression and<br />

function of renal organic ion transporters may cause alterations<br />

in serum and urine creatinine levels in oxonate-treated animals,<br />

resulting in hyperuricemia and renal dysfunction.<br />

Mulberroside A may elicit its uricosuric effect by inhibiting urate<br />

reabsorption via renal mGLUT9 and mURAT1 (l " Fig. 5 A,B and<br />

Fig. 6 A,B) and promoting renal urate secretion via mOAT1<br />

(l " Fig. 5C and Fig. 6 C) in hyperuricemic mice. In addition, mulberroside<br />

A also upregulated expression levels of renal mOCT1,<br />

mOCT2, mOCTN1, and mOCTN2 (l " Fig. 5D–G and Fig. 6 D–G).<br />

These results suggest that regulation of these renal organic ion<br />

transporters by mulberroside A may be responsible for its enhancement<br />

of urate excretion, resulting in the reduction of the<br />

serum uric acid level, and the improvement of renal function in<br />

hyperuricemia. The present study was consistent with other reports<br />

that hypouricemic agents attenuate abnormality of renal<br />

organic ion transporters in oxonic acid-induced hyperuricemic<br />

rats [14] and patients with chronic renal disease [34]. Although<br />

it is unknown how mulberroside A regulates expression of these<br />

renal organic ion transporters, recent studies have provided<br />

some insights. In addition, oxyresveratrol-2-O-beta-D-glucopyranoside,<br />

oxyresveratrol-3′-O-beta-D-glucopyranoside, and oxyresveratrol,<br />

three major metabolites of mulberroside A, have recently<br />

been isolated from the small intestinal contents of rats<br />

Original Papers<br />

Fig. 4 Histological studies of the kidney in oxonate-induced<br />

hyperuricemia in mice. The kidney of<br />

normal-vehicle mice (A), hyperuricemia-vehicle<br />

mice treated with water (B), mulberroside A at 5<br />

(C), 10 (D), 20 (E), and 40 (F) mg/kg, probenecid at<br />

50 (G) and 100 (H) mg/kg; (A–H) 200 ×.<br />

[35]. Oxyresveratrol is transported to tissues much faster than<br />

mulberroside A [36]. Whether these metabolites of mulberroside<br />

A confer the uricosuric activity remains unknown. Further investigation<br />

about the precise molecular mechanism of mulberroside<br />

A regulation on these renal organic ion transporters is necessary.<br />

Of note, mGLUT9 shares 85% homology with hGLUT9 [37]. The<br />

amino acid sequence of mURAT1 has a 74% identity with that of<br />

hURAT1 [28]. hOAT1, hOCT1, and hOCT2 also exhibit a high identity<br />

to mOAT1 (82.9%) [38], mOCT1 (77–95%), and mOCT2 (80%)<br />

[39], and mOCTN1 and mOCTN2 are 84 and 85.5% identical to<br />

hOCTN1 [40] and hOCTN2 [41]. If indeed the present findings in<br />

mice can be extrapolated to humans, mulberroside A may be a<br />

potential candidate for the treatment of hyperuricemia and gout.<br />

In addition, deregulation of these renal organic ion transporters<br />

by certain drugs may cause excessive accumulation of endogenous<br />

and exogenous noxious substances in the kidney, which<br />

may further promote renal dysfunction and elicit nephrotoxicity<br />

[19]. In this regard, it should be noted that disorders of these renal<br />

organic ion transporters may elevate the bodyʼs exposure to<br />

drugs, and induce unexpected side effects of drugs during the<br />

treatment of hyperuricemia and renal dysfunction in the clinic.<br />

In conclusion, the present study showed that mulberroside A<br />

treatment enhanced renal urate excretion, resulting in a reduction<br />

of serum uric acid levels in oxonate-induced hyperuricemic<br />

mice. It also improved renal function characterized by decreasing<br />

serum creatinine and BUN levels, and urinary NAG activity, β2-<br />

MG and albumin levels, and increasing creatinine clearance in<br />

this model. The uricosuric and nephroprotective effects of mulberroside<br />

A were elicited in part by its regulation of renal<br />

mGLUT9, mURAT1, mOAT1, mOCT1, mOCT2, mOCTN1, and<br />

mOCTN2 in hyperuricemic mice. Our findings provide a scientific<br />

basis for the use of mulberroside A as a new drug candidate for<br />

the treatment of hyperuricemia with renal dysfunction.<br />

Acknowledgements<br />

!<br />

This work was co-financed by grants from NCET-06-0442, NSFC<br />

(No. 30873413 and 81025025), JSNSF (BK2010365), and 07-C-<br />

016 to Ling-Dong Kong.<br />

Wang C-P et al. Mulberroside A Possesses … <strong>Planta</strong> Med 2011; 77: 786–794<br />

791


792<br />

Original Papers<br />

Fig. 5 Effects of mulberroside A and probenecid on protein levels of renal<br />

organic ion transporters in oxonate-induced hyperuricemia in mice. Kidney<br />

cortex tissues were used for Western blot analysis of mGLUT9 (A), mOAT1(C),<br />

mOCT1 (D), and mOCT2 (E). The brush boarder membranes of kidney cortex<br />

Wang C-P et al. Mulberroside A Possesses… <strong>Planta</strong> Med 2011; 77: 786–794<br />

were prepared for Western blot analysis of mURAT1 (B), mOCTN1 (F) and<br />

mOCTN2 (G). Data are expressed as the mean ± SEM for 4 mice; ### p < 0.001<br />

versus normal-vehicle animals; * p < 0.05, ** p < 0.01, and *** p < 0.001 vs.<br />

hyperuricemia-vehicle animals.


Fig. 6 Effects of mulberroside A and probenecid on mRNA levels of renal<br />

ion transporters in oxonate-induced hyperuricemia in mice. Kidney cortex<br />

RNA was extracted for RT-PCR analysis of mGLUT9 (A), mURAT1 (B), mOAT1<br />

(C), mOCT1 (D), mOCT2 (E), mOCTN1 (F), and mOCTN2 (G). Data are ex-<br />

Original Papers<br />

pressed as the mean ± SEM for 4 mice; ## p < 0.01, ### p < 0.001 vs. normalvehicle<br />

animals; * p < 0.05, ** p < 0.01 and *** p < 0.001 vs. hyperuricemiavehicle<br />

animals.<br />

Wang C-P et al. Mulberroside A Possesses … <strong>Planta</strong> Med 2011; 77: 786–794<br />

793


794<br />

Original Papers<br />

References<br />

1 Choi HK, Mount DB, Reginato AM. Pathogenesis of gout. Ann Intern Med<br />

2005; 143: 499–516<br />

2 Weiner DE, Tighiouart H, Elsayed EF, Griffith JL, Salem DN, Levey AS. Uric<br />

acid and incident kidney disease in the community. J Am Soc Nephrol<br />

2008; 19: 1204–1211<br />

3 Imaoka T, Kusuhara H, Adachi-Akahane S, Hasegawa M, Morita N, Endou<br />

H, Sugiyama Y. The renal-specific transporter mediates facilitative<br />

transport of organic anions at the brush border membrane of mouse<br />

renal tubules. J Am Soc Nephrol 2004; 15: 2012–2022<br />

4 Caulfield MJ, Munroe PB, OʼNeill D, Witkowska K, Charchar FJ, Doblado M,<br />

Evans S, Eyheramendy S, Onipinla A, Howard P, Shaw-Hawkins S, Dobson<br />

RJ, Wallace C, Newhouse SJ, Brown M, Connell JM, Dominiczak A, Farrall M,<br />

Lathrop GM, Samani NJ, Kumari M, Marmot M, Brunner E, Chambers J, Elliott<br />

P, Kooner J, Laan M, Org E, Veldre G, Viigimaa M, Cappuccio FP, Ji C, Iacone<br />

R, Strazzullo P, Moley KH, Cheeseman C. SLC2A9 is a high-capacity<br />

urate transporter in humans. PLoS Med 2008; 5: 1509–1523<br />

5 Enomoto A, Kimura H, Chairoungdua A, Shigeta Y, Jutabha P, Cha SH, Hosoyamada<br />

M, Takeda M, Sekine T, Igarashi T, Matsuo H, Kikuchi Y, Oda T,<br />

Ichida K, Hosoya T, Shimokata K, Niwa T, Kanai Y, Endou H. Molecular<br />

identification of a renal urate-anion exchanger that regulates blood urate<br />

levels. Nature 2002; 417: 447–452<br />

6 Ichida K, Hosoyamada M, Kimura H, Takeda M, Utsunomiya Y, Hosoya T,<br />

Endou H. Urate transport via human PAH transporter hOAT1 and its<br />

gene structure. Kidney Int 2003; 63: 143–155<br />

7 Eraly SA, Vallon V, Rieg T, Gangoiti JA, Wikoff WR, Siuzdak G, Barshop BA,<br />

Nigam SK. Multiple organic anion transporters contribute to net renal<br />

excretion of uric acid. Physiol Genomics 2008; 33: 180–192<br />

8 Grover B, Buckley D, Buckley AR, Cacini W. Reduced expression of organic<br />

cation transporters rOCT1 and rOCT2 in experimental diabetes.<br />

J Pharmacol Exp Ther 2004; 308: 949–956<br />

9 Ji L, Masuda S, Saito H, Inui K. Down-regulation of rat organic cation<br />

transporter rOCT2 by 5/6 nephrectomy. Kidney Int 2002; 62: 514–524<br />

10 Tokuhiro S, Yamada R, Chang XT, Suzuki A, Kochi Y, Sawada T, Suzuki M,<br />

Nagasaki M, Ohtsuki M, Ono M, Furukawa H, Nagashima M, Yoshino S,<br />

Mabuchi A, Sekine A, Saito S, Takahashi A, Tsunoda T, Nakamura Y, Yamamoto<br />

K. An intronic SNP in a RUNX1 binding site of SLC22A4, encoding<br />

an organic cation transporter, is associated with rheumatoid arthritis.<br />

Nat Genet 2003; 35: 341–348<br />

11 Glube N, Closs E, Langguth P. OCTN2-mediated carnitine uptake in a<br />

newly discovered human proximal tubule cell line (Caki-1). Mol Pharm<br />

2007; 4: 160–168<br />

12 Harris MD, Siegel LB, Alloway JA. Gout and hyperuricemia. Am Fam<br />

Physician 1999; 59: 925–934<br />

13 Nunome S, Kondo K, Terabayashi S, Sasaki H, Xiao C, Hao X, Okada M.<br />

Studies on the quality of mulberry bark (2). Botanical source and<br />

chemical components of mulberry cortex. Nat Med 2000; 54: 33–37<br />

14 Habu Y, Yano I, Okuda M, Fukatsu A, Inui K. Restored expression and activity<br />

of organic ion transporters rOAT1, rOAT3 and rOCT2 after hyperuricemia<br />

in the rat kidney. Biochem Pharmacol 2005; 69: 993–999<br />

15 Kang DH, Nakagawa T, Feng LL, Watanabe S, Han L, Mazzali M, Truong L,<br />

Harris R, Johnson RJ. A role for uric acid in the progression of renal disease.<br />

J Am Soc Nephrol 2002; 13: 2888–2897<br />

16 Hall IH, Scoville JP, Reynolds DJ, Simlot R, Duncan P. Substituted cyclic<br />

imides as potential antigout agents. Life Sci 1990; 46: 1923–1927<br />

17 Hu QH, Wang CA, Li JM, Zhang DM, Kong LD. Allopurinol, rutin, and<br />

quercetin attenuate hyperuricemia and renal dysfunction in rats induced<br />

by fructose intake: renal organic ion transporter involvement.<br />

Am J Physiol Renal Physiol 2009; 297: F1080–F1091<br />

18 Hu QH, Jiao RQ, Wang X, Lv YZ, Kong LD. Simiao pill ameliorates urate<br />

underexcretion and renal dysfunction in hyperuricemic mice.<br />

J Ethnopharmacol 2010; 128: 685–692<br />

19 Rizwan AN, Burckhardt G. Organic anion transporters of the SLC22 family:<br />

biopharmaceutical, physiological, and pathological roles. Pharm<br />

Res 2007; 24: 450–470<br />

20 Iseki K, Oshiro S, Tozawa M, Iseki C, Ikemiya Y, Takishita S. Significance of<br />

hyperuricemia on the early detection of renal failure in a cohort of<br />

screened subjects. Hypertens Res 2001; 24: 691–697<br />

21 Chonchol M, Shlipak MG, Katz R, Sarnak MJ, Newman AB, Siscovick DS,<br />

Kestenbaum B, Carney JK, Fried LF. Relationship of uric acid with progression<br />

of kidney disease. Am J Kidney Dis 2007; 50: 239–247<br />

22 Price RG. Urinalysis to exclude and monitor nephrotoxicity. Clin Chim<br />

Acta 2000; 297: 173–182<br />

Wang C-P et al. Mulberroside A Possesses… <strong>Planta</strong> Med 2011; 77: 786–794<br />

23 Lauwerys R, Bernard A. Preclinical detection of nephrotoxicity – description<br />

of the tests and appraisal of their health significance. Toxicol<br />

Lett 1989; 46: 13–29<br />

24 Matsuo H, Chiba T, Nagamori S, Nakayama A, Domoto H, Phetdee K, Wiriyasermkul<br />

P, Kikuchi Y, Oda T, Nishiyama J, Nakamura T, Morimoto Y,<br />

Kamakura K, Sakurai Y, Nonoyama S, Kanai Y, Shinomiya N. Mutations<br />

in glucose transporter 9 gene SLC2A9 cause renal hypouricemia. Am<br />

J Hum Genet 2008; 83: 744–751<br />

25 Vitart V, Rudan I, Hayward C, Gray NK, Floyd J, Palmer CN, Knott SA, Kolcic<br />

I, Polasek O, Graessler J, Wilson JF, Marinaki A, Riches PL, Shu X, Janicijevic<br />

B, Smolej-Narancic N, Gorgoni B, Morgan J, Campbell S, Biloglav Z,<br />

Barac-Lauc L, Pericic M, Klaric IM, Zgaga L, Skaric-Juric T, Wild SH, Richardson<br />

WA, Hohenstein P, Kimber CH, Tenesa A, Donnelly LA, Fairbanks<br />

LD, Aringer M, McKeigue PM, Ralston SH, Morris AD, Rudan P, Hastie<br />

ND, Campbell H, Wright AF. SLC2A9 is a newly identified urate transporter<br />

influencing serum urate concentration, urate excretion and<br />

gout. Nat Genet 2008; 40: 437–442<br />

26 Preitner F, Bonny O, Laverriere A, Rotman S, Firsov D, Da Costa A, Metref<br />

S, Thorens B. Glut9 is a major regulator of urate homeostasis and its genetic<br />

inactivation induces hyperuricosuria and urate nephropathy.<br />

Proc Natl Acad Sci USA 2009; 106: 15 501–15 506<br />

27 Vazquez-Mellado J, Jimenez-Vaca AL, Cuevas-Covarrubias S, Alvarado-<br />

Romano V, Pozo-Molina G, Burgos-Vargas R. Molecular analysis of the<br />

SLC22A12 (URAT1) gene in patients with primary gout. Rheumatology<br />

2007; 46: 215–219<br />

28 Hosoyamada M, Ichida K, Enomoto A, Hosoya T, Endou H. Function and<br />

localization of urate transporter 1 in mouse kidney. J Am Soc Nephrol<br />

2004; 15: 261–268<br />

29 Nigam SK, Bush KT, Bhatnagar V. Drug and toxicant handling by the OAT<br />

organic anion transporters in the kidney and other tissues. Nat Clin<br />

Pract Nephrol 2007; 3: 443–448<br />

30 Sakurai Y, Motohashi H, Ueo H, Masuda S, Saito H, Okuda M, Mori N,<br />

Matsuura M, Doi T, Fukatsu A, Ogawa O, Inui K. Expression levels of renal<br />

organic anion transporters (OATs) and their correlation with<br />

anionic drug excretion in patients with renal diseases. Pharm Res<br />

2004; 21: 61–67<br />

31 Urakami Y, Kimura N, Okuda M, Inui K. Creatinine transport by basolateral<br />

organic cation transporter hOCT2 in the human kidney. Pharm Res<br />

2004; 21: 976–981<br />

32 Brown CDA, Sayer R, Windass AS, Haslam IS, De Broe ME, DʼHaese PC,<br />

Verhulst A. Characterisation of human tubular cell monolayers as a<br />

model of proximal tubular xenobiotic handling. Toxicol Appl Pharmacol<br />

2008; 233: 428–438<br />

33 Ichida K, Hosoyamada M, Hisatome I, Enomoto A, Hikita M, Endou H, Hosoya<br />

T. Clinical and molecular analysis of patients with renal hypouricemia<br />

in Japan-influence of URAT1 gene on urinary urate excretion.<br />

J Am Soc Nephrol 2004; 15: 164–173<br />

34 Siu YP, Leung KT, Tong MKH, Kwan TH. Use of allopurinol in slowing the<br />

progression of renal disease through its ability to lower serum uric acid<br />

level. Am J Kidney Dis 2006; 47: 51–59<br />

35 Zhaxi MC, Chen LX, Li XG, Komatsu K, Yao XS, Qiu F. Three major metabolites<br />

of mulberroside A in rat intestinal contents and feces. <strong>Planta</strong> Med<br />

2010; 76: 362–364<br />

36 Qiu F, Komatsu K, Saito K, Kawasaki K, Yao XS, Kano Y. Pharmacological<br />

properties of traditional medicines. XXII. Pharmacokinetic study of<br />

mulberroside A and its metabolites in rat. Biol Pharm Bull 1996; 19:<br />

1463–1467<br />

37 Keembiyehetty C, Augustin R, Carayannopoulos MO, Steer S, Manolescu<br />

A, Cheeseman CI, Moley KH. Mouse glucose transporter 9 splice variants<br />

are expressed in adult liver and kidney and are up-regulated in diabetes.<br />

Mol Endocrinol 2006; 20: 686–697<br />

38 Hosoyamada M, Sekine T, Kanai Y, Endou H. Molecular cloning and functional<br />

expression of a multispecific organic anion transporter from human<br />

kidney. Am J Physiol Renal Physiol 1999; 276: F122–F128<br />

39 Kakehi M, Koyabu N, Nakamura T, Uchiumi T, Kuwano M, Ohtani H, Sawada<br />

Y. Functional characterization of mouse cation transporter<br />

mOCT2 compared with mOCT1. Biochem Biophys Res Commun 2002;<br />

296: 644–650<br />

40 Tamai I, Yabuuchi H, Nezu J, Sai Y, Oku A, Shimane M, Tsuji A. Cloning<br />

and characterization of a novel human pH-dependent organic cation<br />

transporter, OCTN1. FEBS Lett 1997; 419: 107–111<br />

41 Lu KM, Nishimori H, Nakamura Y, Shima K, Kuwajima M. A missense<br />

mutation of mouse OCTN2, a sodium-dependent carnitine cotransporter,<br />

in the juvenile visceral steatosis mouse. Biochem Biophys Res<br />

Commun 1998; 252: 590–594


Telemetry as a Tool to Measure Sedative Effects<br />

of a Valerian Root Extract and Its Single Constituents<br />

in Mice<br />

Authors Nicholas K. Chow 1 , Michael Fretz 2 , Matthias Hamburger 2 , Veronika Butterweck 1<br />

Affiliations<br />

Key words<br />

l " Valeriana officinalis L.<br />

l " Valerianaceae<br />

l " linarin<br />

l " valerenic acid<br />

l " apigenin<br />

l " telemetry<br />

received Sept. 8, 2010<br />

revised Nov. 3, 2010<br />

accepted Nov. 5, 2010<br />

Bibliography<br />

DOI http://dx.doi.org/<br />

10.1055/s-0030-1250589<br />

Published online December 10,<br />

2010<br />

<strong>Planta</strong> Med 2011; 77: 795–803<br />

© Georg Thieme Verlag KG<br />

Stuttgart · New York ·<br />

ISSN 0032‑0943<br />

Correspondence<br />

Dr. Veronika Butterweck<br />

Department of Pharmaceutics<br />

College of Pharmacy<br />

University of Florida<br />

PO Box 100494<br />

Gainesville FL 32610<br />

USA<br />

Phone: + 13 52273 78 59<br />

Fax: + 1352273 7854<br />

butterwk@cop.ufl.edu<br />

1 Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA<br />

2 Institute of Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland<br />

Abstract<br />

!<br />

Valeriana officinalis L. is a popular herbal treatment<br />

for mild sleep disorders. Clinical and nonclinical<br />

studies found contradictory results for valerian<br />

extracts and single constituents regarding<br />

the influence on sleep parameters. It was the aim<br />

of this study to investigate the sedative effects of a<br />

valerian root extract. Therefore, locomotor activity<br />

and core body temperature were recorded in<br />

male mice using radiotelemetry. A 70% ethanolic<br />

extract prepared from the roots of V. officinalis<br />

(s. l.) and some of its single constituents, valerenic<br />

acid, linarin, and apigenin, were tested for effects<br />

on locomotion and body temperature over 180<br />

minutes after oral administration. The extract<br />

was tested in a dose range of 250–1000 mg/kg,<br />

and only a dose of 1000 mg/kg valerian extract<br />

Introduction<br />

!<br />

An estimated 10 to 15% of the adult population<br />

suffers from chronic insomnia (≥ 1 month) and<br />

an additional 25 to 35% from transient or occasional<br />

insomnia (< 1 month) [1]. These high prevalence<br />

rates connected with pharmacological<br />

treatments cause significant economic and clinical<br />

costs. Traditionally, preparations obtained<br />

from the roots of Valeriana officinalis L. (Valerianaceae)<br />

are used as an herbal remedy because of<br />

their putative sleep promoting effects. In most<br />

countries, valerian preparations are a marketed<br />

over-the-counter product with remarkable success.<br />

One reason for this is that herbal products<br />

and dietary supplements are often considered as<br />

more natural and safer than prescription or other<br />

nonprescription drugs and therefore are very<br />

popular [2].<br />

Not only are extracts from Valeriana officinalis<br />

L. the best known, but also the most studied herbal<br />

treatment for insomnia. Clinical studies of its<br />

Original Papers<br />

showed a mild short-term sedative effect with reduced<br />

locomotor activity between 66–78 min<br />

minutes after administration. Paradoxically, an<br />

increased activity was observed after 150 minutes<br />

after gavage. A dose of 1 mg/kg valerenic acid<br />

produced an intermittent stimulation of activity.<br />

However, a mild short-term sedative effect was<br />

found for linarin at 12 mg/kg and apigenin at<br />

1.5 mg/kg. Considering the cumulative locomotor<br />

activity over the observation period of 180 min, it<br />

is concluded that neither the extract nor one of<br />

the compounds had considerable sedative effects.<br />

More precisely, the observed short-term changes<br />

in activity pattern indicate that valerian extract<br />

as well as the flavonoids linarin and apigenin are<br />

rather effective to reduce sleep latency than to act<br />

as a sleep-maintaining agent.<br />

sleep-inducing and sleep-promoting effects are<br />

contradictory [2–7]. Although sedative and anxiolytic<br />

effects have been observed in nonclinical<br />

studies [8, 9], literature provides insufficient evidence<br />

to imply beneficial effects on sleep.<br />

Several compounds have been isolated and identified<br />

from valerian root preparations, but it is<br />

still unclear which of them are responsible for<br />

the recorded activities. Some of the active compounds<br />

found in commonly used extracts are the<br />

sesquiterpenic acids, especially valerenic acid,<br />

which was recently identified as a GABAA receptor<br />

modulator [8, 10]. Recently, sedative or sleepenhancing<br />

properties were detected for the flavonoids:<br />

linarin (acacetin-7-O-rutinoside) [11],<br />

6-methylapigenin (4′,5,7-trihydroxy-6-methylflavone)<br />

[12,13], and hesperidin (2S(−)-7-rhamnoglucosyl-hesperetin)<br />

[12]. More recently, lignan<br />

derivatives, in particular 4′-O-beta-D-glucosyl-9-O-(6′′-deoxysaccharosyl)olivil<br />

found in V.<br />

officinalis, were shown to bind the adenosine A 1<br />

receptor, which is linked to sleep induction [14].<br />

Chow NK et al. Telemetry as a… <strong>Planta</strong> Med 2011; 77: 795–803<br />

795


796<br />

Original Papers<br />

In animal studies, sleep parameters are optimally evaluated<br />

under familiar and undisturbed conditions where usual routine<br />

is not interrupted. Therefore, the measurement of sleep-inducing<br />

effects in nonclinical studies is complicated by handling-related<br />

disturbances or unfamiliar measuring instruments. For this reason,<br />

changes in locomotor activity are used as a surrogate parameter<br />

to quantify the effect of valerian root, and experiments are<br />

conducted with methods such as open field [15], rotarod test<br />

[16], and elevated plus maze [8, 9]. But these approaches require<br />

special apparatuses, equipped with a particular measuring system<br />

(e.g., photocells and detectors, infrared beams and sensors,<br />

electromagnetic field, or video cameras). Moreover, long-term<br />

studies in animals are not possible using these methods, and<br />

therefore results are inadequate for the investigation of sleep-inducing<br />

effects (plus handling effects which cause stress). Sleepwake<br />

cycle and body temperature are strongly influenced by<br />

each other [17]. While core temperature in humans is rapidly rising<br />

in the morning and falling in the evening, activity and body<br />

temperature in mice are subject to an inversed circadian rhythm.<br />

It is known that individuals normally fall asleep when core body<br />

temperature is decreasing [18]. Thus, it is indicated to consult<br />

both, activity and core temperature, to evaluate possible effects<br />

on sleep.<br />

The present study represents an innovative, objective approach<br />

to measure sedative effects with minimized handling-related<br />

stress and remote data collection. The animals can be left undisturbed<br />

during the recording period and long-term data acquisition<br />

is possible. Most importantly, effects on locomotion can directly<br />

be quantified. For the first time, radiotelemetry was used<br />

to investigate a 70% ethanolic extract of Valeriana officinalis L. in<br />

mice. Additionally, selected constituents with reported positive<br />

pharmacological activity, namely valerenic acid, linarin, and apigenin<br />

[8, 10, 11,19], were tested. Locomotor activity and body<br />

temperature were recorded and analyzed for possible acute and<br />

medium-term sedative effects.<br />

Material and Methods<br />

!<br />

Extract and single compounds<br />

Valerian extract was manufactured by Finzelberg GmbH & Co.<br />

KG, with 70% ethanol as the solvent and a drug : extract ratio<br />

(DER) of 3–6: 1 (91% native extract, batch #: 08016674). Its content<br />

of sesquiterpenic acids, calculated as valerenic acid, was<br />

0.32% (w/w). Valerenic acid (purity ≥ 98.5%) was purchased from<br />

Phytolab GmbH & Co. Linarin (purity ≥ 98.5%) and apigenin (purity<br />

≥ 99%) were purchased from Indofine Chemical Company,<br />

Inc.<br />

Chemicals and drugs<br />

Midazolam hydrochloride 50 mg/10 mL solution was purchased<br />

from Bedford Laboratories. Zolpidem tablets (5 mg) were purchased<br />

from Henry Schein, Inc. and comminuted by mortar and<br />

pestle for preparation as a suspension for oral administration.<br />

Caffeine (purity ≥ 99%) was purchased from Sigma-Aldrich, Inc.<br />

Drug preparation<br />

All preparations for administration were made on the morning of<br />

the experimental run. Compounds for oral administration were<br />

dissolved, suspended or diluted in a vehicle of deionized water<br />

containing 0.5% propylene glycol (Fisher Scientific, Inc.). Compounds<br />

were administered by feeding needle in a volume of<br />

Chow NK et al. Telemetry as a … <strong>Planta</strong> Med 2011; 77: 795–803<br />

10 mL/kg in the following concentrations: midazolam 5 mg/kg,<br />

zolpidem 5 mg/kg, caffeine 5 mg/kg, valerian extract 250–<br />

1000 mg/kg, valerenic acid 0.5–5.0 mg/kg, linarin 2.0–12 mg/kg,<br />

and apigenin 1.5–6.0 mg/kg. The concentration of each compound<br />

was chosen based on published literature. Control animals<br />

only received vehicle of the same volume.<br />

Animals<br />

Non-fasted male C57BL/6J mice (supplied by Harlan Sprague<br />

Dawley, Inc.) weighing 22–30 g (between 8–12 weeks old) were<br />

singly caged in a temperature controlled (20 ± 2°C) quiet room<br />

and maintained on a nonreversed 12-h light/dark cycle (lights<br />

on at 6 a. m. and lights off at 6 p.m.). The animals had unlimited<br />

access to food (Harlan Teklad LM-485, standard diet) and water.<br />

From the day of the E-Mitter implantation, all mice were handled<br />

daily before the treatment in order to reduce potential handling<br />

stress. Each mouse was considered available for experimental<br />

runs after a minimum of 7 days post-transmitter implantation.<br />

Animals were randomly assigned to the different treatment<br />

groups (n = 8 per group). Experiments were conducted between<br />

9 a. m. and 1 p. m. All animal experiments were performed according<br />

to the policies and guidelines of the Institutional Animal<br />

Care and Use Committee (IACUC) of the University of Florida,<br />

Gainesville, USA (NIH publication #85-23), study protocol #<br />

200801768 (approved on 09/25/2008).<br />

G2 E-Mitter implantation<br />

The surgical implantation of the G2 E-Mitters was performed<br />

under aseptic conditions according to the University of Florida,<br />

Animal Care Services Guidelines for Telemetry Implants and<br />

Guidelines for Survival Surgery in Mice. Before surgery, meloxicam<br />

(Sigma-Aldrich, Inc.) 1 mg/kg was administered orally for analgesia,<br />

and anesthesia was maintained by 1–3% isoflurane delivered<br />

by nose cone. Twenty-four hours before implantation, the E-Mitters<br />

were sterilized with 2.4% activated glutaraldehyde (Cidex ® ,<br />

Johnson & Johnson, Inc.). The abdomen was opened by making a<br />

2-cm incision along the linea alba. The intestines and colon were<br />

gently reflected to allow insertion of the E-Mitter into the abdominal<br />

cavity along the sagittal plane, placing it in front of the<br />

caudal arteries and veins, but dorsal to the digestive organs. The<br />

positioning is important to acquire accurate temperature data. In<br />

order to prevent migration of the E-Mitter within the peritoneal<br />

cavity, transmitters were anchored to the abdominal cavity wall<br />

by the silastic suture sleeve. The peritoneal incision was closed<br />

using 4/0 caliber monofilament polypropylene nonabsorbable<br />

sutures with reverse cutting needle in a simple interrupted stitch<br />

pattern. The skin incision was closed with a series of 9-mm surgical<br />

staples which were removed 7 days after surgery. Rehydration<br />

at the end of the surgery was assured by subcutaneous administration<br />

of sterile isotonic saline (10 mL/kg body weight). Wound<br />

recovery was checked regularly.<br />

Telemetry system<br />

A data acquisition system, the VitalView ® from Mini Mitter, a<br />

Respironics Company (Series 4000), was used to acquire animal<br />

body temperature and activity data. VitalView ® is a software<br />

and hardware system specially designed for controlling data collection<br />

in laboratory monitoring of physiological parameters<br />

without the need for animal handling and prior habituation to<br />

the test environment or the presence of the experimenter. E-Mitters<br />

capture energy from the field of radio waves emitted by coils<br />

of the ER-4000 Energizer/Receiver, allowing the E-Mitter devices


to be free from battery requirements. The signal returned to the<br />

receiver consists of sequences of pulses whose period is dependent<br />

on temperature. The VitalView ® system records the average<br />

rate and the receiver converts this pulse frequency into a serial<br />

bit stream using calibration values specific to each device. For<br />

the quantification of locomotor activity, movement of the implanted<br />

E-Mitter results in fluctuations in signal strength, which<br />

are detected by the ER-4000 Receiver via telemetry and noted as<br />

activity counts. The total number of these counts during a sampling<br />

period interval is recorded. After a minimum 5-day washout<br />

period following administration of the test substance, each<br />

mouse was administered its matched control vehicle for a comparison<br />

run. At least two corresponding control values were<br />

matched with every test value. On the day of the experiment,<br />

the cages were placed on the receivers at least one hour before<br />

dosing. At 9:29 a. m. EST, dosing was started for the first four mice<br />

and at 9:35 a.m. EST for the second four mice. To assure and control<br />

constant environmental conditions, experiment runs were<br />

always accompanied by control animals.<br />

Original Papers<br />

Fig. 1 Changes in locomotor activity (A,C,E) and<br />

in body temperature (B,D, F) after oral administration<br />

of midazolam (5 mg/kg), zolpidem (5 mg/kg),<br />

caffeine (5 mg/kg), or vehicle. Values are expressed<br />

as mean ± SEM, n = 8 per group. Zero min represents<br />

time of dosing.<br />

Statistical analysis<br />

All statistical procedures were calculated with GraphPad Prism ®<br />

statistical software package, version 5.00 (GraphPad Software,<br />

Inc). Locomotor activity and body temperature data were collected<br />

in 6-minute intervals and are displayed as mean ± SEM.<br />

Comparative cumulative locomotor activity between groups was<br />

analyzed using two-way analysis of variance (ANOVA) followed<br />

by Bonferroniʼs post hoc test. Additionally, temperature and activity<br />

pattern differences between control and each treatment<br />

group were evaluated using the unpaired two-tailed t-test. A<br />

probability level of p < 0.05 was set as statistically significant.<br />

Results<br />

!<br />

l " Fig. 1A shows the depressant effect of midazolam (5 mg/kg,<br />

p.o.) and illustrates the rapid onset of action within the first minutes.<br />

Locomotor activity was significantly lower at 12 minutes<br />

(control: 104.86 ± 16.48, midazolam: 17.36 ± 4.55; p < 0.001,<br />

two-way ANOVA) and 18 minutes (control: 109.45 ± 20.85, midazolam:<br />

24.73 ± 11.33; p < 0.001, two-way ANOVA) after administration.<br />

As shown in l " Fig. 1B, body temperature showed a sig-<br />

Chow NK et al. Telemetry as a… <strong>Planta</strong> Med 2011; 77: 795–803<br />

797


798<br />

Original Papers<br />

nificant drop of 0.78 °C 12 minutes after dosing (control: 37.19 ±<br />

0.15 °C, midazolam: 36.41 ± 0.18 °C; p < 0.05, two-way ANOVA).<br />

The mean body temperature for the initial 30 minutes was significantly<br />

lower in the midazolam group compared to vehicle (control:<br />

37.13 ± 0.09 °C, midazolam: 36.66 ± 0.12 °C; p < 0.05, unpaired<br />

two-tailed t-test).<br />

Similarly to midazolam, zolpidem (5 mg/kg, p.o.) decreased the<br />

activity in the first 30 minutes (l " Fig. 1C). Mean body temperature<br />

at 12 minutes after administration was significantly lower<br />

in the zolpidem group (35.96 ± 0.13°C) than in the control group<br />

(36.43 ± 0.07 °C; p < 0.01, unpaired two-tailed t-test) (l " Fig. 1 D).<br />

The total activity for 180 minutes after administration of 5 mg/<br />

kg caffeine was increased to 126% compared to the control group<br />

(l " Fig. 1E). More precisely, locomotion was significantly stimulated<br />

only during the initial 30 minutes with a high significance<br />

at 12 minutes (control: 72.40 ± 10.86, caffeine: 176.00 ± 31.76;<br />

p < 0.001), 18 minutes (control: 42.71 ± 10.38, caffeine: 146.43 ±<br />

36.90; p < 0.001) and 24 minutes (control: 30.09 ± 7.23, caffeine:<br />

140.29 ± 32.96; p < 0.001) indicating a rapid onset of action. Body<br />

temperature was not considerably affected (l " Fig. 1F). The cumulative<br />

locomotor activity of midazolam, zolpidem, and caffeine<br />

are shown in l " Table 1.<br />

l " Fig. 2A and C show no major effect on locomotor activity after<br />

oral administration of 250 and 500 mg/kg valerian extract, re-<br />

Chow NK et al. Telemetry as a … <strong>Planta</strong> Med 2011; 77: 795–803<br />

Treatment Group Cumulative activity<br />

[0–180 min]<br />

Control 1242 ± 73<br />

Caffeine 5 mg/kg 1622 ± 97*<br />

Midazolam 5 mg/kg 829 ± 66**<br />

Zolpidem 5 mg/kg 951 ± 87*<br />

Control 1286 ± 75<br />

Valerian extract 250 mg/kg 1305 ± 112<br />

Valerian extract 500 mg/kg 1398 ± 158<br />

Valerian extract 1000 mg/kg 1367 ± 108<br />

Control 1405 ± 97<br />

Valerenic acid 0.5 mg/kg 1232 ± 175<br />

Valerenic acid 1.0 mg/kg 1221 ± 152<br />

Valerenic acid 2.0 mg/kg 1268 ± 97<br />

Valerenic acid 5.0 mg/kg 1282 ± 171<br />

Control 1303 ± 59<br />

Linarin 2.0 mg/kg 1254 ± 130<br />

Linarin 6.0 mg/kg 1335 ± 134<br />

Linarin 12.0 mg/kg 1197 ± 151<br />

Control 1173 ± 73<br />

Apigenin 1.5 mg/kg 1026 ± 194<br />

Apigenin 3.0 mg/kg 1185 ± 124<br />

Apigenin 6.0 mg/kg 1195 ± 126<br />

Values are expressed as mean ± SEM, two-way analysis of variance<br />

(ANOVA) followed by Bonferroniʼs post hoc test, * p < 0.05;<br />

** p < 0.01<br />

Table 1 Cumulativelocomotor<br />

activity<br />

after oral administration<br />

of<br />

test compounds.<br />

Fig. 2 Changes in locomotor activity (A,C,E)and<br />

in body temperature (B,D, F) after oral administration<br />

of valerian extract (250, 500, and 1000 mg/kg,<br />

respectively) or vehicle. Values are expressed as<br />

mean ± SEM, n = 8 per group. Zero min represents<br />

time of dosing.


spectively, and body temperature remained mostly unaffected in<br />

these concentrations (l " Fig. 2 B, D). In a higher concentration of<br />

1000 mg/kg,valerian extractdid not induceasignificantdifference<br />

in the overall locomotor activity over 180 minutes (l " Table 1), but<br />

altered the pattern of activity with nonsignificant short-term decreases<br />

between 60 and 90 minutes (l " Fig. 2E) and increases in<br />

the extract group between 132 and 150 minutes. Body temperature<br />

remained unchanged during the whole recording period except<br />

with a nonsignificant increase between 132 and 180 minutes<br />

(control: 35.68 ± 0.05 °C, extract: 35.96 ± 0.10 °C) (l " Fig. 2 F).<br />

No major effects were observed for valerenic acid in a low concentration<br />

(0.5 mg/kg) (l " Fig. 3 A, B). One mg/kg of valerenic acid<br />

considerably altered the locomotion pattern (l " Fig. 3C). After a<br />

prominent increase at 78 minutes, a subsequent decrease occurred.<br />

Between 138 and 180 minutes, activity was significantly<br />

lower in the valerenic acid group. Simultaneously, body temperature<br />

was significantly decreased by 0.34°C during the same period<br />

(l " Fig. 3D; control: 35.82 ± 0.09°C, valerenic acid: 35.48 ±<br />

0.10 °C; p < 0.05, unpaired two-tailed t-test). In higher concentra-<br />

Original Papers<br />

Fig. 3 Changes in locomotor activity (A, C,E, G)<br />

and in body temperature (B,D, F, H) after oral administration<br />

of valerenic acid (0.5, 1.0, 2.0, and<br />

5.0 mg/kg, respectively) or vehicle. Values are expressed<br />

as mean ± SEM, n = 8 per group. Zero min<br />

represents time of dosing.<br />

tions (2 mg/kg and 5 mg/kg, respectively), valerenic acid did not<br />

induce any statistically significant changes in locomotion<br />

(l " Fig. 3E, G) or body temperature (l " Fig. 3 F, H). Overall, valerenic<br />

acid had no effects on the cumulative activity from 0–180 min<br />

(l " Table 1).<br />

l " Fig. 4A and 4B illustrate that linarin at a concentration of 2 mg/<br />

kg did not considerably affect locomotion or body temperature.<br />

After oral administration of 6 mg/kg linarin, a significant increase<br />

in locomotor activity during the initial 30 minutes compared to<br />

vehicle was observed. l " Fig. 4C shows the significantly higher activity<br />

values at 12 minutes (control: 72.40 ± 10.86, linarin:<br />

150.20 ± 35.00; p < 0.05), 18 minutes (control: 42.71 ± 10.38, linarin:<br />

152.80 ± 41.04; p < 0.001) and 24 minutes (control: 30.09 ±<br />

7.23, linarin: 110.80 ± 40.42; p < 0.01). Body temperature was not<br />

affected over the whole observation period (l " Fig. 4B). In a dose<br />

of 12 mg/kg, linarin significantly reduced locomotor activity between<br />

60 and 132 minutes (l " Fig. 4 E) whereas no effect on body<br />

temperature was observed (l " Fig. 4 F). Considering the effects on<br />

Chow NK et al. Telemetry as a… <strong>Planta</strong> Med 2011; 77: 795–803<br />

799


800<br />

Original Papers<br />

the cumulative activity from 0–180 min, linarin had no significant<br />

effect (l " Table 1).<br />

Oral administration of 1.5 mg/kg apigenin induced a significant<br />

reduction in locomotor activity (l " Fig. 5 A). The evaluation indicates<br />

significantly lower activity between 90 and 120 minutes<br />

after gavage. The overall activity during 180 minutes did not significantly<br />

differ between the groups after oral administration of<br />

3 mg/kg apigenin (l " Fig. 5 C). However, locomotion was slightly<br />

lowered between 84 and 114 minutes. Locomotion pattern was<br />

also not considerably altered after administration of 6 mg/kg apigenin<br />

(l " Fig. 5 E). However, the total cumulative activity remained<br />

unaffected (l " Table 1). No considerable effect on body<br />

temperature was observed for all concentrations (l " Fig. 5 B, D,F).<br />

Discussion<br />

!<br />

It is generally acknowledged that the quality of physiological<br />

measurements collected from conscious unstressed animals possesses<br />

the highest validity and best reflects the normal state of<br />

the animal [20–22]. The radiotelemetry system therefore represents<br />

an adequate method and has proven its justification in<br />

modern pharmaceutical research thanks to a minimum of handling-related<br />

or restraint stress to the animal [17, 23,24]. In light<br />

of the above, the present study was interested in possible sedative<br />

effects of a 70% ethanolic valerian extract and some of its sin-<br />

Chow NK et al. Telemetry as a … <strong>Planta</strong> Med 2011; 77: 795–803<br />

Fig. 4 Changes in locomotor activity (A,C,E)and<br />

in body temperature (B,D, F) after oral administration<br />

of linarin (2.0, 6.0, and 12.0 mg/kg, respectively)<br />

or vehicle. Values are expressed as mean ±<br />

SEM, n = 8 per group. Zero min represents time of<br />

dosing.<br />

gle constituents (valerenic acid, linarin, and apigenin), which are<br />

assumed to be mainly responsible for valerianʼs activity.<br />

The outcomes of the positive (midazolam, zolpidem) and negative<br />

(caffeine) controls are supported by existing literature data<br />

and validate the used method [23, 25–29].<br />

The effects of valerian root on sleep parameters are only sparsely<br />

investigated in animals. Wagner et al. [30] observed a sedative action<br />

of a fresh valerian tincture at 1000 mg/kg p.o. in mice,<br />

whereas an aqueous extract induced a slight decrease in spontaneous<br />

motility at 10 mg/kg p. o. Similar findings are reported by<br />

Leuschner et al. [15] who found pronounced sedative properties<br />

after oral administration of 20 and 200 mg/kg of an aqueous alkaline<br />

extract. While assessing these results, it must be taken into<br />

consideration that statistical specification or significance is absent<br />

in these studies [15, 30]. Additionally, enhancing of barbiturate<br />

sleeping time is demonstrated in several studies [15, 31] and<br />

considered to be evidence for sedation. But its relevance for proving<br />

sedative properties remains unclear since barbiturates have<br />

been shown to interfere with CYP P450 enzymes [32], and therefore<br />

an increased sleeping time due to interactions of valerian<br />

with the barbiturate metabolism in mice cannot be excluded.<br />

Tokunaga et al. [33] reported significant shortening of sleep latency<br />

in rats after oral administration of 1000 mg/kg valerian extract<br />

without affecting total time of wakefulness, non-REM sleep,<br />

and REM sleep. These results are not supported by the present<br />

study, which did not find any significant effect on total cumula-


tive activity over 180 minutes. However, 1000 mg/kg altered the<br />

activity pattern of the mice and induced a nonsignificant intermittent<br />

drop in activity (66–78 min). This short-term depressant<br />

effect is not sufficient to ascribe positive effects on sleep, since a<br />

stimulating activity was observed thereafter. In contrast, 250 mg/<br />

kg and 500 mg/kg did not affect locomotor activity at all. Hattesohl<br />

et al. [9] drew a similar conclusion since they reported no effect<br />

on locomotion for several valerian extract preparations after<br />

acute and subacute (19 days) oral administration.<br />

Temperature data indicate that valerian extract produces a mild<br />

dose-dependent hyperthermic effect in higher doses. This finding<br />

is contradictory to Hendriks et al. [34] who reported a hypothermic<br />

effect for the essential oil of V. officinalis and several constituents,<br />

including valerenic acid. In contrast, Hiller and Zetler<br />

[31] did not observe any influence on body temperature after administration<br />

of 50 and 100 mg/kg ethanolic valerian extract (i. p.),<br />

whereas diazepam produced a significant hypothermic effect. No<br />

marked effects on temperature have been observed in the<br />

present study. In conclusion, considering our results on locomotor<br />

activity and body temperature obtained with telemetry, it<br />

seems unlikely that valerian has any significant sedative actions.<br />

None of the tested concentrations of valerenic acid did significantly<br />

lower the activity over 180 minutes. However, at a concentration<br />

of 1 mg/kg the activity pattern was considerably altered.<br />

A short-lasting increase was observed between 78 and 102 minutes<br />

after administration followed by a reduction in locomotion<br />

Original Papers<br />

Fig. 5 Changes in locomotor activity (A,C,E)and<br />

in body temperature (B,D, F) after oral administration<br />

of apigenin (1.5, 3.0, and 6.0 mg/kg, respectively)<br />

or vehicle. Values are expressed as mean ±<br />

SEM, n = 8 per group. Zero min represents time of<br />

dosing.<br />

between 138 and 180 minutes. These findings are in line with results<br />

from recent studies. Benke et al. [8] reported no indication<br />

for sedative activity for 10 mg/kg p.o. and 30 mg/kg i.p. (elevated<br />

plus-maze). A similar conclusion was drawn by Fernandez et al.<br />

[11]. They observed no in vivo effects in mice at low doses up to<br />

15 mg/kg i.p. (sodium thiopental-induced sleeping time and<br />

holeboard test). In contrast, Hendriks et al. [34] found valerenic<br />

acid to decrease motility of mice at a concentration of 50 mg/kg<br />

i. p. and higher. The same authors reported in a later study a decrease<br />

in rotarod performance in mice (100 mg/kg i. p.) and a<br />

dose-related increase in pentobarbital-induced sleeping time<br />

(50 and 100 mg/kg i. p.) [16]. Furthermore, a significant decrease<br />

of rectal temperature in mice at concentrations of 100, 200, and<br />

400 mg/kg was observed. Paradoxically, after administration of<br />

50 mg/kg, a mild increase in body temperature occurred [16].<br />

Overall, the authors concluded that this compound has central<br />

depressant properties. However, considering the extremely high<br />

and unphysiological doses, the relevance of these results has to<br />

be questioned.<br />

In the present study, linarin induced only at a higher concentration<br />

(12 mg/kg) a significant reduction of locomotor activity between<br />

60 and 132 minutes; however, when considering the cumulative<br />

activity, no significant changes were detected. Therefore,<br />

the findings of Fernandez et al. [11], could partly be confirmed.<br />

The authors observed depressant effects at doses of 4<br />

and 7 mg/kg i. p. in the holeboard test. Furthermore, 7 and<br />

Chow NK et al. Telemetry as a… <strong>Planta</strong> Med 2011; 77: 795–803<br />

801


802<br />

Original Papers<br />

14 mg/kg i. p. significantly prolonged the thiopental-induced<br />

sleeping time in mice. Hence it was concluded that linarin possesses<br />

sedative and sleep-enhancing activity. In contrast to the<br />

findings of Fernandez et al. [11] we found an initial stimulating<br />

effect after oral administration of 6 mg/kg linarin.<br />

While in the present study all substances were administered orally,<br />

Fernandez et al. [11] used intraperitoneal injection. It is<br />

likely that the different routes of administration caused different<br />

effects on locomotor activity. Consequentially, results obtained<br />

using different routes of administration should not be compared<br />

directly.<br />

In the present study, apigenin showed mild sedative effects at<br />

low concentrations. Locomotor activity was significantly reduced<br />

between 90 and 120 minutes (1.5 mg/kg) after gavage. The observed<br />

effect was indeed short-lasting since after 120 minutes activity<br />

began to rise to control levels. In higher doses, apigenin did<br />

not significantly reduce activity or body temperature. Viola et al.<br />

[35] suggested apigenin to act as a benzodiazepine partial agonist<br />

and to possess anxiolytic activity at low doses (3 mg/kg). In the<br />

same study, sedative effects were only observed after i. p. administration<br />

of high doses (30 and 100 mg/kg) in mice (holeboard<br />

and locomotor activity test), but not for doses up to 10 mg/kg<br />

i. p. Similarly, Avallone et al. [36] reported potent sedative properties<br />

for high doses (25 and 50 mg/kg i. p.) in rats as well. However,<br />

in most of the previously reported studies apigenin was tested for<br />

anxiolytic properties and not particularly for sedation. Therefore,<br />

the presented results reveal for the first time the potential beneficial<br />

effect of apigenin on sleep.<br />

In summary, the present findings indicate that the radiotelemetry<br />

system is suitable for the investigation of sleep-modifying effects.<br />

Considering all aspects, it is concluded that neither the extract<br />

nor one of the compounds had significant sedative activities<br />

over 180 min. However, positive short-term sedative effects were<br />

found for valerian extract 1000 mg/kg, linarin 12 mg/kg, and apigenin<br />

1.5 mg/kg whereas valerenic acid (0.5, 1, 2, and 5 mg/kg)<br />

had no significant effect on locomotor activity or body temperature.<br />

The fact that mild sedative effects of the extract and the single<br />

compounds lasted only for a very short period suggests an interesting<br />

pharmacokinetic profile probably pointing to a short halflife<br />

of all compounds. Since pharmacokinetic data about valerian<br />

compounds are only sparsely available, this assumption presently<br />

is speculative. However, pharmacokinetic data are an important<br />

issue to link data with pharmacological assays. Our findings, obtained<br />

with telemetric recordings of locomotor activity and body<br />

temperature, do not support the use of valerian root for the treatment<br />

of sleep disorders in general. However, the observed shortterm<br />

effects presented for the flavonoids linarin and apigenin indicate<br />

that these compounds could be effective to reduce sleep latency<br />

rather than to act as sleep-maintaining agents. Future studies<br />

therefore should focus on the investigation of other flavonoids<br />

from valerian as sleep-inducing agents.<br />

References<br />

1 Benca RM. Diagnosis and treatment of chronic insomnia: a review. Psychiatr<br />

Serv 2005; 56: 332–343<br />

2 Bent S, Padula A, Moore D, Patterson M, Mehling W. Valerian for sleep: a<br />

systematic review and meta-analysis. Am J Med 2006; 119: 1005–1012<br />

3 Balderer G, Borbely AA. Effect of valerian on human sleep. Psychopharmacology<br />

(Berl) 1985; 87: 406–409<br />

4 Brattstroem A. Scientific evidence for a fixed extract combination (ZE<br />

91019) from valerian and hops traditionally used as a sleep-inducing<br />

aid. Wien Med Wochenschr 2007; 157: 367–370<br />

Chow NK et al. Telemetry as a … <strong>Planta</strong> Med 2011; 77: 795–803<br />

5 Diaper A, Hindmarch I. A double-blind, placebo-controlled investigation<br />

of the effects of two doses of a valerian preparation on the sleep,<br />

cognitive and psychomotor function of sleep-disturbed older adults.<br />

Phytother Res 2004; 18: 831–836<br />

6 Leathwood PD, Chauffard F. Aqueous extract of valerian reduces latency<br />

to fall asleep in man. <strong>Planta</strong> Med 1985; 4: 144–148<br />

7 Taibi DM, Vitiello MV, Barsness S, Elmer GW, Anderson GD, Landis CA. A<br />

randomized clinical trial of valerian fails to improve self-reported,<br />

polysomnographic, and actigraphic sleep in older women with insomnia.<br />

Sleep Med 2009; 10: 319–328<br />

8 Benke D, Barberis A, Kopp S, Altmann KH, Schubiger M, Vogt KE, Rudolph<br />

U, Möhler H. GABA A receptors as in vivo substrate for the anxiolytic<br />

action of valerenic acid, a major constituent of valerian root extracts.<br />

Neuropharmacology 2009; 56: 174–181<br />

9 Hattesohl M, Feistel B, Sievers H, Lehnfeld R, Hegger M, Winterhoff H. Extracts<br />

of Valeriana officinalis L. s.l. show anxiolytic and antidepressant<br />

effects but neither sedative nor myorelaxant properties. Phytomedicine<br />

2008; 15: 2–15<br />

10 Trauner G, Khom S, Baburin I, Benedek B, Hering S, Kopp B. Modulation of<br />

GABAA receptors by valerian extracts is related to the content of valerenic<br />

acid. <strong>Planta</strong> Med 2008; 74: 19–24<br />

11 Fernandez S, Wasowski C, Paladini AC, Marder M. Sedative and sleepenhancing<br />

properties of linarin, a flavonoid-isolated from Valeriana officinalis.<br />

Pharmacol Biochem Behav 2004; 77: 399–404<br />

12 Marder M, Viola H, Wasowski C, Fernandez S, Medina JH, Paladini AC. 6methylapigenin<br />

and hesperidin: new valeriana flavonoids with activity<br />

on the CNS. Pharmacol Biochem Behav 2003; 75: 537–545<br />

13 Wasowski C, Marder M, Viola H, Medina JH, Paladini AC. Isolation and<br />

identification of 6-methylapigenin, a competitive ligand for the brain<br />

GABA(A) receptors, from Valeriana wallichii. <strong>Planta</strong> Med 2002; 68:<br />

934–936<br />

14 Schumacher B, Scholle S, Holzl J, Khudeir N, Hess S, Muller CE. Lignans<br />

isolated from valerian: identification and characterization of a new olivil<br />

derivative with partial agonistic activity at A(1) adenosine receptors.<br />

J Nat Prod 2002; 65: 1479–1485<br />

15 Leuschner J, Muller J, Rudmann M. Characterisation of the central nervous<br />

depressant activity of a commercially available valerian root extract.<br />

Arzneimittelforschung 1993; 43: 638–641<br />

16 Hendriks H, Bos R, Woerdenbag HJ, Koster AS. Central nervous depressant<br />

activity of valerenic acid in the mouse. <strong>Planta</strong> Med 1985; 51: 28–<br />

31<br />

17 Mailliet F, Galloux P, Poisson D. Comparative effects of melatonin, zolpidem<br />

and diazepam on sleep, body temperature, blood pressure and<br />

heart rate measured by radiotelemetry in wistar rats. Psychopharmacology<br />

(Berl) 2001; 156: 417–426<br />

18 Weinert D, Waterhouse J. The circadian rhythm of core temperature: effects<br />

of physical activity and aging. Physiol Behav 2007; 90: 246–256<br />

19 Campbell EL, Chebib M, Johnston GA. The dietary flavonoids apigenin<br />

and (−)-epigallocatechin gallate enhance the positive modulation by<br />

diazepam of the activation by GABA of recombinant GABA(A) receptors.<br />

Biochem Pharmacol 2004; 68: 1631–1638<br />

20 Berkey DL, Meeuwsen KW, Barney CC. Measurements of core temperature<br />

in spontaneously hypertensive rats by radiotelemetry. Am<br />

J Physiol 1990; 258: R743–R749<br />

21 Harkin A, Connor TJ, OʼDonnell JM, Kelly JP. Physiological and behavioral<br />

responses to stress: what does a rat find stressful? Lab Anim (NY)<br />

2002; 31: 42–50<br />

22 Taylor SC, Little HJ, Nutt DJ, Sellars N. A benzodiazepine agonist and contragonist<br />

have hypothermic effects in rodents. Neuropharmacology<br />

1985; 24: 69–73<br />

23 Elliot EE, White JM. The acute effects of zolpidem compared to diazepam<br />

and lorazepam using radiotelemetry. Neuropharmacology 2001;<br />

40: 717–721<br />

24 Matthew CB. Telemetry augments the validity of the rat as a model for<br />

heat acclimation. Ann NY Acad Sci 1997; 813: 233–238<br />

25 Davies MF, Onaivi ES, Chen SW, Maguire PA, Tsai NF, Loew GH. Evidence<br />

for central benzodiazepine receptor heterogeneity from behavior tests.<br />

Pharmacol Biochem Behav 1994; 49: 47–56<br />

26 Durcan MJ, Morgan PF. Hypothermic effects of alkylxanthines: evidence<br />

for a calcium-independent phosphodiesterase action. Eur J Pharmacol<br />

1991; 204: 15–20<br />

27 Pieri L. Preclinical pharmacology of midazolam. Br J Clin Pharmacol<br />

1983; 16 (Suppl. 1): 17S–27S


28 Schlosberg AJ. Temperature responses in rats after acute and chronic<br />

administrations of caffeine. Pharmacol Biochem Behav 1983; 18: 935–<br />

942<br />

29 Yang JN, Tiselius C, Dare E, Johansson B, Valen G, Fredholm BB. Sex differences<br />

in mouse heart rate and body temperature and in their regulation<br />

by adenosine A(1) receptors. Acta Physiol (Oxf) 2007; 190: 63–75<br />

30 Wagner H, Jurcic K, Schaette R. Comparative studies on the sedative action<br />

of Valeriana extracts, valepotriates and their degradation product.<br />

<strong>Planta</strong> Med 1980; 39: 358–365<br />

31 Hiller KO, Zetler G. Neuropharmacological studies on ethanol extracts<br />

of Valeriana officinalis l: behavioural and anticonvulsant properties.<br />

Phytother Res 1996; 10: 145–151<br />

32 Kakinohana M, Taira Y, Kakinohana O, Okuda Y. The inhibition of lidocaine<br />

metabolism by various barbiturates in rat hepatic microsome.<br />

Masui 1998; 47: 1302–1310<br />

Original Papers<br />

33 Tokunaga S, Takeda Y, Niimoto T, Nishida N, Kubo T, Ohno T, Matsuura Y,<br />

Kawahara Y, Shinomiya K, Kamei C. Effect of valerian extract preparation<br />

on the sleep-wake cycle in rats. Biol Pharm Bull 2007; 30: 363–366<br />

34 Hendriks H, Bos R, Allersma DP, Malingre TM, Koster AS. Pharmacological<br />

screening of valerenal and some other components of essential oil of<br />

Valeriana officinalis. <strong>Planta</strong> Med 1981; 42: 62–68<br />

35 Viola H, Wasowski C, Levi de Stein M, Wolfman C, Silveira R, Dajas F,<br />

Medina JH, Paladini AC. Apigenin, a component of Matricaria recutita<br />

flowers, is a central benzodiazepine receptors-ligand with anxiolytic<br />

effects. <strong>Planta</strong> Med 1995; 61: 213–216<br />

36 Avallone R, Zanoli P, Puia G, Kleinschnitz M, Schreier P, Baraldi M. Pharmacological<br />

profile of apigenin, a flavonoid isolated from Matricaria<br />

chamomilla. Biochem Pharmacol 2000; 59: 1387–1394<br />

Chow NK et al. Telemetry as a… <strong>Planta</strong> Med 2011; 77: 795–803<br />

803


804<br />

Original Papers<br />

Antihyperglycemic Activity of Bacosine,<br />

a Triterpene from Bacopa monnieri,<br />

in Alloxan-Induced Diabetic Rats<br />

Authors Tirtha Ghosh 1 , Tapan Kumar Maity 2 , Jagadish Singh 2<br />

Affiliations<br />

Key words<br />

l " Bacopa monnieri<br />

l " Scrophulariaceae<br />

l " bacosine<br />

l " alloxan<br />

l " antidiabetic activity<br />

l " antioxidant activity<br />

received August 15, 2010<br />

revised October 12, 2010<br />

accepted Nov. 11, 2010<br />

Bibliography<br />

DOI http://dx.doi.org/<br />

10.1055/s-0030-1250600<br />

Published online December 10,<br />

2010<br />

<strong>Planta</strong> Med 2011; 77: 804–808<br />

© Georg Thieme Verlag KG<br />

Stuttgart · New York ·<br />

ISSN 0032‑0943<br />

Correspondence<br />

Tirtha Ghosh<br />

Department of Pharmacy,<br />

Contai Polytechnic<br />

P. O.-Darua,<br />

Dist-Purba Medinipur<br />

721401 West Bengal<br />

India<br />

Phone: + 91 9475 2017 91<br />

Fax: +913220255462<br />

tghosh75@yahoo.co.in<br />

1 Department of Pharmacy, Contai Polytechnic, West Bengal, India<br />

2 Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India<br />

Abstract<br />

!<br />

This article describes the antihyperglycemic activity,<br />

in vivo antioxidant potential, effect on hemoglobin<br />

glycosylation, estimation of liver glycogen<br />

content, and in vitro peripheral glucose utilization<br />

of bacosine, a triterpene isolated from<br />

the ethyl acetate fraction (EAF) of the ethanolic<br />

extract of Bacopa monnieri. Bacosine produced a<br />

significant decrease in the blood glucose level<br />

when compared with the diabetic control rats<br />

both in the single administration as well as in the<br />

multiple administration study. It was observed<br />

that the compound reversed the weight loss of<br />

the diabetic rats, returning the values to near normal.<br />

Bacosine also prevented elevation of glycosylated<br />

hemoglobin in vitro with an IC50 value of<br />

7.44 µg/mL, comparable with the one for the<br />

reference drug α-tocopherol. Administration of<br />

Introduction<br />

!<br />

Diabetes mellitus, a metabolic disorder of carbohydrate,<br />

fat, and protein metabolisms, is affecting<br />

nearly 10% of the population every year [1]. Its<br />

treatment includes the use of oral hypoglycemic<br />

agents and insulin, the former being reported to<br />

have serious side effects [2]. This leads to increasing<br />

demand for herbal products with an antidiabetic<br />

factor and little side effects. A large number<br />

of plants have been recognized to be effective in<br />

the treatment of diabetes mellitus [3].<br />

Bacopa monnieri Linn. (Scrophulariaceae) is a<br />

creeping, glabrous, succulent herb, rooting at<br />

nodes, distributed throughout India in all plain<br />

districts [4]. The plant is reported to contain tetracyclic<br />

triterpenoid saponins, bacosides A and B,<br />

hersaponin, alkaloids viz. herpestine and brahmine,<br />

and flavonoids [5]. In Ayurveda, the plant<br />

has been used in the treatment of insanity, epilepsy,<br />

and hysteria [6]. The other reported activ-<br />

Ghosh T et al. Antihyperglycemic Activity of… <strong>Planta</strong> Med 2011; 77: 804–808<br />

bacosine and glibenclamide significantly decreased<br />

the levels of malondialdehyde (MDA),<br />

and increased the levels of reduced glutathione<br />

(GSH) and the activities of superoxide dismutase<br />

(SOD) and catalase (CAT) in the liver of diabetic<br />

rats. Bacosine increased glycogen content in the<br />

liver of diabetic rats and peripheral glucose utilization<br />

in the diaphragm of diabetic rats in vitro,<br />

which is comparable with the action of insulin.<br />

Thus, bacosine might have insulin-like activity<br />

and its antihyperglycemic effect might be due to<br />

an increase in peripheral glucose consumption as<br />

well as protection against oxidative damage in alloxanized<br />

diabetes.<br />

Supporting information available online at<br />

http://www.thieme-connect.de/ejournals/toc/<br />

plantamedica<br />

ities include sedative, antiepileptic, vasoconstrictor,<br />

and anti-inflammatory properties [4]. Antidiabetic<br />

activity of the ethanolic extract of the<br />

plant has already been reported by the authors<br />

[7]. Hence, further exploitation of the ethanolic<br />

extract was done to find out the active principle<br />

responsible for the antidiabetic activity. We report<br />

herein the fractionation of the ethanol soluble<br />

extract of Bacopa monnieri, which has led to<br />

the isolation of a triterpenoid, bacosine. As triterpenoids<br />

have been shown to possess antidiabetic<br />

property [8], it was thought worthwhile to investigate<br />

the antidiabetic activity of bacosine in a scientific<br />

manner.<br />

In the present paper we report the antihyperglycemic<br />

activity, in vivo antioxidant potential, effect<br />

on hemoglobin glycosylation, liver glycogen content,<br />

and in vitro peripheral glucose utilization of<br />

bacosine, isolated from Bacopa monnieri.


Materials and Methods<br />

!<br />

Plant material<br />

The plant was identified by the taxonomists of the Botanical Survey<br />

of India, Govt. of India, Howrah, bearing reference no. CNH/1-<br />

1(99)/2005/Tech.II/1615 dated 07.12.2005. A voucher specimen<br />

is kept in our department (CP/PH/TG/02/09) for further reference.<br />

Fresh aerial parts of the young and matured plants were collected<br />

in bulk from the rural belt of Salipur, Orissa, India during early<br />

summer, washed, shade dried and then milled in to coarse powder<br />

by a mechanical grinder.<br />

Extraction and isolation<br />

The powdered plant material (2 kg) was defatted with petroleum<br />

ether (60–80°C) and then extracted with 3 L of ethanol (95%) in a<br />

soxhlet apparatus. The solvent was removed under reduced pressure,<br />

which resulted in a greenish-black sticky residue (232 g).<br />

The residue was partitioned successively in an ethyl acetatewater<br />

system and then in an n-butanol-water system (3 × 1 L).<br />

The respective solvents were removed similarly under reduced<br />

pressure, which produced an ethyl acetate fraction (EAF) (112 g)<br />

and an n-butanol fraction (57.4 g). The EAF fraction (40 g × 2<br />

times) was subjected to column chromatography over silica gel<br />

(60–120 mesh, 5 × 60 cm, flow rate 5 mL/min) using CHCl3-MeOH<br />

(gradient) as an eluent [CHCl3-MeOH = 100 : 0 (500 mL) and 95:5<br />

(1 L)]. The fractions 3–6 (500–1500 mL, 4 g) collected at<br />

CHCl3:MeOH 95: 5 were further subjected to preparative thin<br />

layer chromatography (PTLC) using silica gel G as the stationary<br />

phase (thickness of plates 0.5 mm) and C6H6 : MeOH = 8.5 : 1.5 as<br />

the mobile phase. Three spots were obtained. They were<br />

scrapped off. The spot at Rf value 0.30 yielded a product which<br />

was then recrystallized with CHCl3 and MeOH to afford a compound<br />

(120 mg) as shining white crystalline needles, which was<br />

characterized as bacosine (BS; % purity = 95%) (l " Fig. 1) onthe<br />

basis of spectroscopic (IR, 1 H, 13 C NMR, and MS) data [9].<br />

Animals used<br />

Wistar albino rats of either sex, weighing 180–250 g were used.<br />

The selected animals were housed in acrylic cages in standard environmental<br />

conditions (25–30°C). They were allowed free access<br />

to standard dry pellet diet (Hindustan Lever) and water ad<br />

libitum. The recommendations of the Institutional Animal Ethics<br />

Committee (Regn. No. 0367/01/C/CPCSEA) for the care and use of<br />

laboratory animals were strictly followed throughout the study.<br />

Drugs and chemicals used<br />

Thiobarbituric acid, nitro blue tetrazolium chloride (NBT), hemoglobin<br />

(Loba Chemie), and 5,5′-dithio bis-2-nitrobenzoic acid<br />

(DTNB) were used. Glibenclamide (% purity ≥ 99%) and α-tocopherol<br />

(% purity ≥ 96%) were purchased from Sigma-Aldrich Co.<br />

All the solvents were of analytical grade.<br />

Screening for antidiabetic activity [10]<br />

The animals were considered diabetic when the blood glucose<br />

level was raised beyond 300 mg/dL of blood after 72 h of i.p. injection<br />

of 150 mg/kg of alloxan monohydrate in normal saline.<br />

Bacosine was then evaluated for its antidiabetic activity.<br />

Single administration study: Animals were segregated into seven<br />

groups of six rats in each. Group I, II, and III rats were randomly<br />

selected from normal rats which received only distilled water<br />

and bacosine, 10 and 25 mg/kg, p.o. respectively. Group IV to<br />

Group VII animals were selected from the alloxanized rats. Group<br />

Original Papers<br />

Fig. 1 Structure of<br />

bacosine.<br />

IV animals served as diabetic controls. Groups V, VI, and VII animals<br />

were treated with bacosine, 10 and 25 mg/kg, p. o. [11] and<br />

glibenclamide, 600 µg/kg, respectively, in a similar manner. Blood<br />

was collected from the tip of the tail of each rat under mild ether<br />

anesthesia at 0 h, 1 h, 2 h, and 4 h after the administration of test<br />

samples. Estimation of blood glucose was carried out with the<br />

hemoglucostrips (Lifescan, Inc.) with the help of a Johnson &<br />

Johnson One Touch blood glucometer. The hemoglucostrips were<br />

inserted into the glucometer (code matched with that of the<br />

hemoglucostrip vial), and a drop of blood was touched with the<br />

edge of the test strips. The strips automatically draw the blood<br />

and give a reading within 5 sec.<br />

Multi-administration study: Administration of test samples was<br />

continued for 10 days, once daily. Blood samples were collected<br />

and estimated as above on the 1, 3, 7, and 10th day of the drug<br />

administration. Body weights of all the animals were recorded<br />

just prior to and on the 10th day of the study to determine the<br />

change in the body weight if any.<br />

Effect on oral glucose tolerance in rats: Animals were segregated<br />

into different groups as stated in the single administration study.<br />

After overnight fasting, a 0-min blood glucose level was estimated<br />

in different groups and without delay a glucose solution<br />

(2 g/kg) was administered. Four more blood samples were taken<br />

at 30, 60, 90, and 120 min after glucose administration [7] and estimated<br />

for blood glucose level.<br />

Determination of in vivo antioxidant activity: On the 10th day following<br />

study, the animals were deprived of food overnight and<br />

sacrificed by cervical dislocation. The livers were dissected out,<br />

washed in ice-cold saline, patted dry and weighed. A 10% w/v of<br />

homogenate was prepared in 0.15 M Tris-HCl buffer and processed<br />

for the estimation of lipid peroxidation [12], GSH [13],<br />

SOD [14], and CAT [15].<br />

Determination of in vitro glycosylation of hemoglobin: The degree<br />

of glycosylation of hemoglobin in vitro was measured colorimetrically<br />

as suggested by Fluckiger et al., 1978 [16]. Hemoglobin,<br />

5 mg/mL in 0.01 M phosphate buffer (pH 7.4) was incubated for<br />

72 h in the presence of 2 g/100 mL concentration of glucose in order<br />

to find out the best condition for hemoglobin glycosylation.<br />

The assay was performed by adding 1 mL of glucose solution,<br />

1 mL of hemoglobin solution, and 1 mL of gentamycin (20 mg/<br />

100 mL) in 0.01 M phosphate buffer (pH 7.4). The mixture was incubated<br />

in the dark at room temperature. The degree of glycosylation<br />

of hemoglobin in presence of different concentration of bacosine<br />

and its absence was measured colorimetrically at 440 nm.<br />

α-Tocopherol was used as a standard.<br />

Determination of peripheral consumption of glucose in vitro: The<br />

method of Chattopadhyay et al., 1992 [17] was followed. Peripheral<br />

glucose consumption was studied in the rat diaphragm prep-<br />

Ghosh T et al. Antihyperglycemic Activity of … <strong>Planta</strong> Med 2011; 77: 804–808<br />

805


806<br />

Original Papers<br />

aration from animals fasted for 36 h prior to the experiment. The<br />

animals were sacrificed by cervical dislocation, and the diaphragms<br />

were quickly taken out; followed by dividing each diaphragm<br />

into four pieces. The pieces of diaphragms were incubated<br />

in the nutrient solution with constant oxygenation and<br />

shaking (90 cycles/min) at 37°C for 90 min in accordance with<br />

the procedure. The nutrient solution with the diaphragms was<br />

aerated for 10 min and used immediately. Glucose was added to<br />

a final concentration of 500 mg %. Each piece of diaphragm was<br />

incubated in 2.5 mL of glucose nutrient mixture. The results were<br />

expressed as glucose consumption per 10 mg of dry diaphragm<br />

(by subtracting glucose concentration after incubation from glucose<br />

concentration before incubation). The dry weight was determined<br />

after oven-drying the diaphragm at 105 °C for 2 hours.<br />

Determination of glycogen content of liver: Glycogen estimation of<br />

the liver was carried out by the use of anthrone reagent [18].<br />

2 mL of the liver extract was added to 2 mL of 10 N KOH solution<br />

and boiled for 1 h. After cooling, 1 mL of glacial acetic acid was<br />

added, and the fluid was made up to 20 mL with water. 2 mL of<br />

this solution was added to 4 mL of anthrone reagent (0.5 g of<br />

anthrone was dissolved in 250 mL of 95% (w/v) H2SO4, shaken<br />

and boiled for 10 min for color development. The optical density<br />

was read within 2 h at 650 nm against a blank solution.<br />

Statistical analysis<br />

Statistical significance was determined by one-way analysis of<br />

variance (ANOVA) followed by Dunnetʼs t-test. P < 0.05 indicates<br />

a significant difference between group means.<br />

Supporting information<br />

Original spectra for bacosine are available as Supporting Information.<br />

Results and Discussion<br />

!<br />

The ethyl acetate fraction on further purification led to the isolation<br />

of a triterpene. The compound melted at 284–285 °C, which<br />

is characteristic of bacosine. The physical and spectral data were<br />

in complete agreement to those reported in literature for bacosine<br />

[9] (l " Fig. 1).<br />

l " Table 1 shows the blood glucose level of normal and experimental<br />

animals after oral administration of glucose (2 g/kg). Bacosine<br />

at a dose of 25 mg/kg showed a more significant decrease<br />

in the peak blood glucose level after 1 h and tended to bring the<br />

values near normal values after 2 h.<br />

The results of l " Tables 2 and 3 reveal that bacosine, when applied<br />

to normal rats, did not show any significant change in the blood<br />

glucose levels with respect to the normal control rats but showed<br />

a significant reduction (p < 0.001) of the glucose level in diabetic<br />

rats throughout the experimental period (in both single and multi-administration<br />

studies) in a dose-dependent manner. This<br />

proves that bacosine has antihyperglycemic activity rather than<br />

hypoglycemic activity.<br />

Diabetes mellitus impairs the ability to use glucose for energy<br />

which leads to increased utilization and decreased storage of<br />

protein, and this is responsible for reduction of body weight primarily<br />

by depletion of the body proteins [19]. In the present<br />

study, it was observed that bacosine, in a dose-dependent manner,<br />

reversed the weight loss of the diabetic rats and they returned<br />

to near normal values (l " Table 3).<br />

The excess glucose present in the blood during diabetes reacts<br />

with hemoglobin to form glycosylated hemoglobin, and with its<br />

improvement glycosylated hemoglobin decreases [20]. It is a<br />

well-established fact that the estimation of glycosylation of hemoglobin<br />

is a useful parameter in the management and prog-<br />

Table 1 Effect of bacosine (10 and 25 mg/kg, p.o.) on oral glucose tolerance test (OGTT) in normal and alloxan-induced diabetic rats.<br />

Treatment Blood sugar level (mg/dL)<br />

Fasting 30 min 60 min 90 min 120 min<br />

Normal 76.15 ± 0.83 157.64 ± 3.29 181.58 ± 2.37 120.44 ± 1.96 82.59 ± 1.47<br />

Normal + BS (10 mg/kg) 76.73 ± 0.87a 152.83 ± 2.76a 183.19 ± 2.64a 123.07 ± 2.39a 85.32 ± 1.86a Normal + BS (25 mg/kg) 75.82 ± 0.66a 147.35 ± 3.38a 179.26 ± 2.07a 116.40 ± 2.28a 82.43 ± 1.75a Diabetic control 257.19 ± 2.49c 315.38 ± 5.58c 380.67 ± 4.92c 325.47 ± 3.84c 312.39 ± 3.08c Diabetic + BS (10 mg/kg) 78.54 ± 0.55f 155.63 ± 3.75f 192.69 ± 3.28f 138.56 ± 3.95f 91.74 ± 1.63f Diabetic + BS (25 mg/kg) 78.38 ± 0.62f 149.24 ± 2.75f 183.48 ± 2.98f 125.17 ± 2.36f 86.54 ± 1.23f Diabetic + glibenclamide 77.26 ± 0.48f 145.04 ± 2.73f 181.59 ± 2.40f 121.32 ± 2.87f 84.16 ± 1.94f Values are mean ± SEM for n = 6; a not significant when compared to normal group; c p < 0.001 when compared to normal group; e p < 0.01 and f p < 0.001 when compared to<br />

diabetic control group<br />

Table 2 Effect of single administration treatment of bacosine (10 and 25 mg/kg, p.o.) on blood glucose level in normal and alloxan-induced diabetic rats.<br />

Treatment Blood glucose level(mg/dL)<br />

Basal value 1 h 2 h 4 h<br />

Normal 78.59 ± 0.73 78.76 ± 0.83 78.55 ± 0.73 79.14 ± 0.64<br />

Normal + BS (10 mg/kg) 79.18 ± 0.68a 79.15 ± 0.76a 78.84 ± 0.81a 78.53 ± 0.59a Normal + BS (25 mg/kg) 78.54 ± 0.76a 78.37 ± 0.66a 78.12 ± 0.83a 77.89 ± 0.74a Diabetic control 314.76 ± 3.82c 316.53 ± 3.75c 317.18 ± 4.15c 317.93 ± 3.38c Diabetic + BS (10 mg/kg) 316.13 ± 4.08d 295.48 ± 3.55e 284.53 ± 4.92f 276.83 ± 4.15f Diabetic + BS (25 mg/kg) 312.89 ± 3.57d 286.39 ± 2.88f 268.73 ± 3.15f 257.26 ± 3.18f Diabetic + glibenclamide 317.06 ± 4.19d 283.62 ± 3.17f 257.18 ± 3.84f 242.48 ± 2.86f Values are mean ± SEM for n = 6; a not significant when compared to normal group; c p < 0.001 when compared to normal group; d not significant when compared to diabetic<br />

control group; e p < 0.01 and f p < 0.001 when compared to diabetic control group<br />

Ghosh T et al. Antihyperglycemic Activity of… <strong>Planta</strong> Med 2011; 77: 804–808


Table 3 Effect of multiple administration treatment of bacosine (10 and 25 mg/kg, p.o.) on blood glucose level and change in body weight after 15 days in<br />

normal and alloxan-induced diabetic rats.<br />

Treatment Blood glucose level (mg/dL) Change in<br />

Basal value Day 1 Day 3 Day 7 Day 10<br />

body weight (g)<br />

Normal 78.59 ± 0.73 78.44 ± 0.87 79.59 ± 0.69 78.55 ± 0.54 79.23 ± 0.62 (+) 9.85 ± 1.29<br />

Normal + BS (10 mg/kg) 79.18 ± 0.68a 78.66 ± 0.65a 78.12 ± 0.91a 79.38 ± 0.88a 78.92 ± 0.59a (+) 10.06 ± 1.57a Normal + BS (25 mg/kg) 78.54 ± 0.76a 78.92 ± 0.80a 79.38 ± 0.71a 80.03 ± 0.92a 79.16 ± 0.68a (+) 10.92 ± 1.22a Diabetic control 314.76 ± 3.82c 319.25 ± 4.16c 325.54 ± 3.83c 316.39 ± 3.66c 307.85 ± 5.03c (−) 8.87 ± 1.15c Diabetic + BS (10 mg/kg) 316.13 ± 4.08d 255.85 ± 2.56f 221.38 ± 2.74f 200.41 ± 2.44f 192.38 ± 1.42f (+) 9.12 ± 1.73f Diabetic + BS (25 mg/kg) 312.89 ± 3.57d 234.19 ± 3.73* 206.13 ± 3.04f 188.57 ± 1.95f 176.65 ± 1.57f (+) 9.92 ± 1.81f Diabetic + glibenclamide 317.06 ± 4.19d 225.37 ± 3.18* 194.48 ± 2.65f 176.37 ± 1.39f 163.52 ± 1.46f (+) 10.25 ± 0.98f Values are mean ± SEM for n = 6; a not significant when compared to normal group; c p < 0.001 when compared to normal group; d not significant when compared to diabetic<br />

control group; f p < 0.001 when compared to diabetic control group<br />

Table 4 Effect of bacosine on percent inhibition of hemoglobin glycosylation<br />

in vitro.<br />

Sample Concentration % inhibition IC50 R<br />

(µg/mL) ±SD<br />

(µg/mL)<br />

Bacosine 1 2.86 ± 0.42 7.44 0.9909<br />

2.5 13.52 ± 2.06<br />

5 34.11 ± 2.69<br />

10 45.76 ± 3.58<br />

15 70.38 ± 5.10<br />

α-Tocopherol 7.74 0.9985<br />

Values are mean ± SD for n = 3; R = regression coefficient<br />

nosis of the disease [21]. The present study showed that bacosine<br />

significantly prevented glycosylation of hemoglobin in vitro, with<br />

an IC50 value of 7.44 µg/mL, which is comparable to the one of the<br />

reference drug α-tocopherol (l " Table 4). Non-enzymatic glycosylation<br />

of hemoglobin is reported to be an oxidative reaction<br />

[22]. As bacosine prevented significantly the increase of hemoglobin<br />

glycosylation, it was thus expected to possess antioxidant<br />

activity, and this fact was further substantiated by our in vivo<br />

studies.<br />

One of the characteristic features of chronic diabetes is lipid peroxidation.<br />

Alloxan gives rise to dialuric acid, which generates O2,<br />

H2O2, and OH [23] and stimulates lipid peroxidation. Administration<br />

of bacosine significantly decreased the levels of MDA in diabetic<br />

rats at a dose of 10 mg/kg (p < 0.05) and 25 mg/kg (p < 0.01)<br />

in a dose-dependent manner (l " Table 5).<br />

GSH, a tripeptide present in all the cells, is an important antioxidant<br />

[24]. Decreased GSH levels in diabetes have been considered<br />

to be an indicator of increased oxidative stress [25]. Our study<br />

showed a decrease in GSH in the liver during diabetes. Administration<br />

of bacosine and glibenclamide increased the level of GSH in<br />

the liver of diabetic rats, though no significant difference was observed<br />

in both MDA and GSH levels when bacosine was administeredtonormal<br />

ratswith respect tonormal control rats(l " Table 5).<br />

The cellular radical scavenging systems include enzymes such as<br />

SOD, which scavenges the superoxide ions by catalyzing its dismutation,<br />

and CAT, a heme enzyme which removes hydrogen<br />

peroxide [26]. Administration of bacosine increased the activity<br />

of SOD and CAT in a dose-dependent manner in diabetic rats. Bacosine<br />

did not show any significant change in SOD and CAT activity<br />

in normal rats when compared to normal control (l " Table 5).<br />

Lupeol, a triterpenoid, has been shown to protect cells from oxidative<br />

stress-induced damage by increasing the transcriptional<br />

activity of NRF 2, which induces the formation of endogenous<br />

Table 5 Effect of bacosine (10 and 25 mg/kg, p.o.) on the levels of MDA,<br />

GSH, glycogen content, and activities of SOD and CAT in the liver of normal<br />

and diabetic rats.<br />

Treatment MDA i GSH j SOD k CAT l Glycogen<br />

Normal 0.31 ± 0.02 9.56 ±<br />

0.53<br />

Normal + BS<br />

(10 mg/kg)<br />

Normal + BS<br />

(25 mg/kg)<br />

Diabetic<br />

control<br />

Diabetic + BS<br />

(10 mg/kg)<br />

Diabetic + BS<br />

(25 mg/kg)<br />

Diabetic +<br />

glibenclamide<br />

0.31 ± 0.01 a 9.88 ±<br />

0.68 a<br />

0.30 ± 0.02 a 10.52 ±<br />

0.74 a<br />

0.72 ± 0.06 d 4.73 ±<br />

0.26 d<br />

0.57 ± 0.05 f 7.94 ±<br />

0.66 g<br />

0.42 ± 0.04 g 9.08 ±<br />

0.56 h<br />

0.37 ± 0.02 h 9.43 ±<br />

0.75 h<br />

5.76 ± 0.43 75.68 ±<br />

6.77<br />

5.85 ± 0.57a 78.23 ±<br />

8.36a 5.91 ± 0.49a 79.42 ±<br />

7.52a 3.55 ± 0.28c 37.51 ±<br />

4.42d 4.83 ± 0.37f 55.41 ±<br />

6.19f 5.18 ± 0.41g 63.72 ±<br />

6.11g 5.52 ± 0.53g 68.81 ±<br />

5.06h Original Papers<br />

content m<br />

43.78 ± 1.52<br />

44.19 ± 1.06 a<br />

45.72 ± 0.98 a<br />

8.08 ± 0.47 d<br />

31.58 ± 1.52 h<br />

39.43 ± 1.65 h<br />

40.65 ± 1.02 h<br />

Values are mean ± SEM for n = 6 animals; a not significant when compared to normal<br />

group; c p < 0.01, and d p < 0.001 when compared to normal group; f p < 0.05,<br />

g h p < 0.01, and p < 0.001 when compared to diabetic control group. MDA: malondialdehyde;<br />

GSH: reduced glutathione; SOD: superoxide dismutase; CAT: catalase;<br />

i j k nmole of MDA/mg of protein; µg/mg of protein; units/mg of protein; one unit of<br />

activity means enzyme reaction responsible for 50% inhibition of NBT per min;<br />

l µ mole of H2O2 consumed/min/mg of protein; m mg/100 g of liver<br />

antioxidant enzymes like CAT, SOD, and others [27]. Bacosine,<br />

being a lupeol-type triterpene, showed an increase in SOD and<br />

CAT activity probably by the same mechanism.<br />

Alloxan causes irreversible destruction of the β-cells of the pancreas<br />

[28]. Thus, the antihyperglycemic activity might be due to<br />

extra pancreatic mechanism. Hence, the effect of bacosine on<br />

peripheral consumption of glucose was investigated. The result<br />

suggests that bacosine produces an antidiabetic effect mediated<br />

by an increase in peripheral glucose consumption in the diaphragm<br />

of diabetic rats, especially at a concentration of 50 µg/<br />

mL (l " Table 6). Insulin increased the peripheral glucose consumption<br />

in normal and diabetic rats. Thus, bacosine might have<br />

insulin-like activity, and the antihyperglycemic effect might be<br />

due to an increase in peripheral glucose consumption.<br />

The above in vitro observation was further substantiated by observing<br />

the effect of bacosine on glycogen content in the liver of<br />

diabetic rats in vivo. It has been reported that insulin is a potent<br />

activator of glycogen synthase and hexokinase, responsible for<br />

the formation of glycogen, and inhibitor of glycogen phosphorylase,<br />

responsible for glycogenolysis in the liver [29]. Insulin defi-<br />

Ghosh T et al. Antihyperglycemic Activity of … <strong>Planta</strong> Med 2011; 77: 804–808<br />

807


808<br />

Original Papers<br />

Table 6 Effect of bacosine on in vitro peripheral glucose consumption by the<br />

diaphragm of normal and diabetic rats.<br />

Glucose consumption (mg/10 mg<br />

of diaphragm dry weight)<br />

Bacosine (µg/mL) Insulin<br />

Control 25 (µg/mL) 50 (µg/mL) 5 U/mL<br />

Normal rats 0.57 ± 0.05 0.60 ± 0.07a 0.62 ± 0.08a 0.86 ± 0.06c Diabetic rats 0.62 ± 0.06 0.81 ± 0.05e 0.89 ± 0.06f 0.95 ± 0.08f Values are mean ± SEM for n = 6; a not significant when compared to normal rats;<br />

c e f p < 0.01 when compared to normal group; p < 0.05 and p < 0.01 when compared<br />

to diabetic control group<br />

ciency in diabetes results in decreased concentrations of glycogen<br />

in the liver. Bacosine caused a significant increase (p < 0.001) in<br />

glycogen content in the liver of diabetic rats (l " Table 5) and thus<br />

further proved its insulin-like activity leading to an increased uptake<br />

of glucose.<br />

Thus, from the present study it is evident that bacosine, a triterpene<br />

isolated from Bacopa monnieri, possesses significant antihyperglycemic<br />

activity against alloxan-induced diabetic rats,<br />

although it did not show any activity on normal rats. The abovementioned<br />

activity might be attributed to the protective action of<br />

bacosine on lipid peroxidation and at the same time to the enhancing<br />

effects on cellular antioxidant defense contributing to<br />

the protection against oxidative damage in alloxanized diabetes<br />

leading to an improvement of tissues and a subsequent increase<br />

in uptake and utilization of blood glucose.<br />

Acknowledgements<br />

The authors are thankful to the authorities of the Jadavpur University,<br />

Kolkata, India for providing the necessary facilities to<br />

carry out the research work.<br />

References<br />

1 Vetrichelvan T, Jagadeesan M, Uma Devi BA. Antidiabetic activity of alcohol<br />

extract of Celosia argentea Linn. seeds in rats. Biol Pharm Bull<br />

2002; 25: 526–528<br />

2 Pickup J, Williams G. Text book of diabetes. Blackwell: Oxford; 1991:<br />

467–469<br />

3 Nagarajan S, Jain HC, Aulakh GS. Indigenous plants used in the control<br />

of diabetes. New Delhi: CSIR; 1987: 586–590<br />

4 Anonymous. Indian herbal pharmacopoeia, Vol. I. Mumbai: IDMA;<br />

1998: 3037<br />

5 Kiritikar KR, Basu BD. Indian Medicinal plants, Vol. I. Dehradun: Bishen<br />

Singh Mahendrapal Singh; 1994: 1816–1817<br />

6 Chopra RN, Nayar SL, Chopra IC. Glossary of Indian medicinal plants.<br />

New Delhi, India: National Institute of Science and Communication<br />

(CSIR); 1986: 32–34<br />

7 Ghosh T, Maity TK, Sengupta P, Dash DK, Bose A. Antidiabetic and in vivo<br />

antioxidant activity of ethanolic extract of Bacopa monnieri Linn. aerial<br />

parts: a possible mechanism of action. Iranian J Pharm Res 2008; 7:<br />

61–68<br />

Ghosh T et al. Antihyperglycemic Activity of… <strong>Planta</strong> Med 2011; 77: 804–808<br />

8 Tan MJ, Ye J, Turner N, Hohnen-Behrens C, Ke CQ, Tang CP, Chen T, Weiss<br />

HC, Gesing ER, Rowland A, James DE, Ye Y. Antidiabetic activities of triterpenoids<br />

isolated from bitter melon associated with activation of the<br />

AMPK pathway. Chem Biol 2008; 15: 263–273<br />

9 Ahmed B, Rahman A. Bacosterol, a new 13,14-seco-steroid and Bacosine,<br />

a new triterpene from Bacopa monniera. Indian J Chem 2000;<br />

39B: 620–625<br />

10 Joy KL, Kuttan R. Antidiabetic activity of Picrorrhiza kurroa extract.<br />

J Ethnopharmacol 1999; 67: 143–148<br />

11 Vohra SB, Khanna T, Athar M. Analgesic activity of bacosine a new triterpene<br />

isolated from B. monniera. Fitoterapia 1997; 68: 361–365<br />

12 Fraga CG, Leibovitz BE, Toppel AL. Lipid peroxidation measured as thiobarbituric<br />

acid-reactive substances in tissue slices: characterization<br />

and comparison with homogenates and microsomes. Free Radic Biol<br />

Med 1988; 4: 155–161<br />

13 Ellman GL. Tissue sulfhydryl groups. Arch Biochem Biophys 1959; 82:<br />

70–77<br />

14 Kakkar P, Das B, Viswanathan PN. A modified spectroscopic assay of<br />

superoxide dismutase. Indian J Biochem Biophys 1984; 21: 130–132<br />

15 Maehly AC, Chance B. The assay of catalases and peroxidases. In: Glick<br />

D, editor. Methods of biochemical analysis, Vol I. New York: Interscience;<br />

1954: 357–358<br />

16 Fluckiger R, Winterhalter KH. Biochemical and clinical aspects of hemoglobin<br />

abnormalities. New York: Academic Press; 1978: 208–209<br />

17 Chattopadhyay RR, Sarkar SK, Ganguly S, Banerjee RN, Basu TK. Effect of<br />

extract of leaves of Vinca rosea Linn. on glucose utilization and glycogen<br />

deposition by isolated rat hemidiaphragm. Indian J Physiol Pharmacol<br />

1992; 36: 137–138<br />

18 Carroll NV, Longley RW, Roe JH. The determination of glycogen in liver<br />

and muscle by use of anthrone reagent. J Biol Chem 1956; 220: 583–<br />

596<br />

19 Guyton AC, Hall JE. Text book of medical physiology, 10th edition. New<br />

Delhi: Elsevier; 2000: 894–897<br />

20 Monnier VK, Cerami A. Non-enzymatic glycosylation and browning in<br />

diabetes and aging. Diabetes 1982; 31: 57–66<br />

21 Chang AT, Nobel J. Estimation of HbA1c like glycosylated proteins in<br />

kidneys of streptozotocin diabetes and controlled rats. Diabetes 1979;<br />

28: 408–415<br />

22 Yadav SB, Tripathi V, Singh RK, Pandey HP. Antioxidant activity of Cusuta<br />

reflexa stem. Indian J Pharm Sci 2000; 62: 477–478<br />

23 Winterbourn CC, Munday R. Glutathion-mediated redox cycling of<br />

alloxan. Biochem Pharmacol 1989; 38: 271–274<br />

24 Lu SC. Regulation of hepatic glutathion synthesis: current concepts and<br />

controversies. FASEB J 1999; 13: 1169–1183<br />

25 Mc Lennan SV, Heffernan S, Wright L, Rae C, Fisher E, Yue DK, Turtle JR.<br />

Changes in hepatic glutathion metabolism in diabetes. Diabetes 1991;<br />

40: 344–349<br />

26 Lee SE, Ju EM, Kim JH. Free radical scavenging and antioxidant enzyme<br />

fortifying activities of extracts from smilax china roots. Exp Mol Med<br />

2001; 34: 263–268<br />

27 Liby KT, Yore MM, Sporn MB. Triterpenoids and rexinoids as multifunctional<br />

agents for the prevention and treatment of cancer. Nat Rev Cancer<br />

2007; 7: 357–369<br />

28 Pari L, Uma Maheswari J. Hypoglycemic activity of Musa sapientum L.<br />

in alloxan-induced diabetic rats. J Ethnopharmacol 1999; 68: 321–325<br />

29 Bollen M, Keppens S, Stalmans W. Specific features of glycogen metabolism<br />

in the liver. Biochem J 1998; 336: 19–31


Effects of Ligustilide on Lipopolysaccharide-Induced<br />

Endotoxic Shock in Rabbits<br />

Authors Meng Shao 1 *, Kai Qu 2 *, Ke Liu 1 , Yuqin Zhang 2 , Ling Zhang 2 , Zeqin Lian 2 , Tingting Chen 2 ,<br />

Junfeng Liu 4 , Aili Wu 3 , Yue Tang 3 , Haibo Zhu 2<br />

Affiliations The affiliations are listed at the end of the article<br />

Key words<br />

l " ligustilide<br />

l " Angelica sinensis (Oliv.) Diels<br />

l " Umbelliferae<br />

l " septic shock<br />

l " tumor necrosis factor‑α<br />

l " interleukin‑1β<br />

received May 23, 2010<br />

revised Sept. 3, 2010<br />

accepted October 28, 2010<br />

Bibliography<br />

DOI http://dx.doi.org/<br />

10.1055/s-0030-1250573<br />

Published online November 23,<br />

2010<br />

<strong>Planta</strong> Med 2011; 77: 809–816<br />

© Georg Thieme Verlag KG<br />

Stuttgart · New York ·<br />

ISSN 0032‑0943<br />

Correspondence<br />

Professor Haibo Zhu, Ph.D.<br />

Institute of Materia <strong>Medica</strong><br />

Chinese Academy of Medicine<br />

Science & Peking Union <strong>Medica</strong>l<br />

College<br />

1 Xian Nong Tan Street<br />

Beijing 100050<br />

P. R. China<br />

Phone: + 86 1063 1881 06<br />

Fax: +861063017757<br />

zhuhaibo@imm.ac.cn<br />

Abstract<br />

!<br />

In this study, we investigated the protective effects<br />

of ligustilide against lipopolysaccharide<br />

(LPS)-induced endotoxic shock in Japanese white<br />

rabbits and attempted to elucidate the possible<br />

mechanism underlying these effects. Forty-two<br />

rabbits were randomized into 6 groups: normal<br />

group, LPS group, dexamethasone group (5 mg/<br />

kg), and 3 ligustilide groups (20, 40, and 80 mg/<br />

kg). After the rabbits had received a LPS infusion<br />

(0.3 mg/kg), dexamethasone and ligustilide were<br />

intravenously injected at the above-mentioned<br />

dosages. Heart rate (HR), mean arterial pressure<br />

(MAP), and rectal temperature (RT) were recorded<br />

throughout the experiment. Tumor necrosis<br />

factor-α (TNF-α), interleukin-1β (IL-1β),<br />

and nitric oxide (NO) levels were measured by radioimmunoassay<br />

every 30 minutes for the first<br />

hour and every 60 minutes thereafter until the<br />

end of the experiment. The serum levels of alanine<br />

transaminase (ALT), aspartate transaminase<br />

(AST), alkaline phosphatase (ALP), γ-glutamyl<br />

transpeptidase (GGT), creatinine kinase (CK), lactate<br />

dehydrogenase (LDH), total protein (TP), creatinine<br />

(Scr), blood urea nitrogen (BUN), total bilirubin<br />

(T. BIL), and counts of formed elements of<br />

Introduction<br />

!<br />

Septic shock is a serious clinical condition associated<br />

with significant morbidity and mortality [1].<br />

It is a systemic inflammatory disorder characterized<br />

by progressive failure of circulation, systemic<br />

hypotension, hyperreactivity to vasoconstrictors,<br />

and subsequent organ dysfunction, which ultimately<br />

leads to multiorgan failure. Septic shock<br />

is mostly caused by gram-negative bacterial infec-<br />

* These authors contributed equally to this work.<br />

blood were measured at 0, 120, and 300 minutes<br />

after the administration of LPS. Hemorheology<br />

was assayed 300 minutes after the LPS injection.<br />

The vital organs were collected and weighed before<br />

histopathologic examination. A comparison<br />

between the LPS group and the ligustilide groups<br />

showed that ligustilide significantly inhibited the<br />

decline in MAP and RT and decreased the levels of<br />

TNF-α, IL-1β, and NO, but had no apparent effect<br />

on HR. Ligustilide also inhibited the increase in<br />

the levels of biochemical markers, such as ALT,<br />

AST, ALP, GGT, LDH, CK, BUN, and Scr, but showed<br />

no apparent effect on T. BIL and TP. Furthermore,<br />

ligustilide partly restored the function of injured<br />

vital organs, including the heart, liver, lungs, and<br />

kidneys. These results suggest that ligustilide protected<br />

the rabbits against LPS-induced endotoxic<br />

shock.<br />

Abbreviation<br />

!<br />

TLC: thin layer chromatography<br />

Original Papers<br />

Supporting information available online at<br />

http://www.thieme-connect.de/ejournals/toc/<br />

plantamedica<br />

tion. Lipopolysaccharide (LPS) has been implicated<br />

as the major factor in the pathogenesis of<br />

septic shock caused by gram-negative bacteria<br />

[2–4]. In experimental animals, LPS challenge<br />

causes pathophysiologic changes that are similar<br />

to those observed in human patients with septic<br />

shock [5].<br />

LPS exerts most of its effects via endogenous mediators<br />

such as cytokines. Among these cytokines,<br />

tumor necrosis factor-α (TNF-α) appears to be<br />

particularly important for the endotoxic effects<br />

of LPS; studies have shown that it induces the<br />

most characteristic changes accompanying endo-<br />

Shao M et al. Effects of Ligustilide… <strong>Planta</strong> Med 2011; 77: 809–816<br />

809<br />

Downloaded by: University of British Columbia. Copyrighted material.


810<br />

Original Papers<br />

toxic shock [6, 7]. Interleukin-1β (IL-1β) has also been shown to<br />

contribute to the development of sepsis by inducing the production<br />

of other inflammatory mediators and cytokines involved in<br />

sepsis [8]. The excessive generation of nitric oxide (NO) via the<br />

induction of inducible nitric oxide synthase (iNOS) has been suggested<br />

to cause hypotension, vascular hyporeactivity, and death<br />

induced by LPS; these findings indicate that the overproduction<br />

of NO plays an important role in septic shock [9–11].<br />

Angelica sinensis (Oliv.) Diels, documented in the pharmacopoeia<br />

of China, is known to be an effective medicinal agent for the nourishment<br />

of blood, regulation of menstruation, alleviation of pain,<br />

and sliding of bowels [12]. A very recent publication showed that<br />

an ethyl acetate extract of A. sinensis significantly improved the<br />

survival of LPS-challenged mice with the reduction of TNF-α in<br />

vivo. This protective effect may be associated with the downregulation<br />

of NF-κB by ligustilide [13]. Ligustilide (l " Fig. 1), one of the<br />

major active components of the essential oil extracted from<br />

Angelica sinensis (Oliv.) Diels [14], has been identified as the inhibitor<br />

of LPS-induced TNF-α production in monocytes. This<br />

property of ligustilide is related to its inhibitory effect on the<br />

transcription of TNF-α mRNA. Furthermore, ligustilide exhibits<br />

significant suppressive effects on TNF-α-mediated nuclear factor-κB(NF-κB)<br />

activation in gene assays [15, 16].<br />

Therefore, in this study, we attempted to investigate the protective<br />

effects of ligustilide in a rabbit model of LPS-induced endotoxicity<br />

and to elucidate the mechanism underlying these protective<br />

effects.<br />

Materials and Methods<br />

!<br />

Plant material and preparation<br />

of ligustilide emulsion injection<br />

Roots of Angelica sinensis (Oliv.) Diels were collected from the<br />

northwest of China and identified by Dr. Ke Liu, Jilin University.<br />

A voucher specimen (No. 510073) was deposited at the herbarium<br />

of the College of Life Sciences, Jilin University.<br />

The dried and pulverized roots of Angelica sinensis (Oliv.) Diels<br />

were extracted with supercritical carbon dioxide twice at<br />

25 mPa and 45 °C, for 3 hours each time. Ligustilide (purity > 95%,<br />

Fig. 1S) was isolated and purified from the extract by silica gel<br />

chromatography performed according to a previously described<br />

method [17]. Considering its poor water solubility and stability,<br />

ligustilide was quickly prepared as a water-in-oil (W/O) emulsion<br />

by using a high-pressure homogenizer.<br />

Animals and reagents<br />

Japanese white rabbits (weight, 2.5–3.0 kg; age, 5–6 months; half<br />

male and half female) were purchased from Changchun Gaoxin<br />

Study Center of <strong>Medica</strong>l Animal (certificate number: SCXK-[Ji]-<br />

2003–0004). Ligustilide emulsion injection (ivory-white fluid;<br />

10 mg/mL) was obtained from Shandong Target-drug Research<br />

Co., Ltd. Dexamethasone injection (5 mg/mL) was purchased<br />

from Tianjing Pharmaceutical Group. Just before use, the abovementioned<br />

drugs were dissolved in sodium chloride solution to<br />

achieve the required concentrations. LPS (extracted from Escherichia<br />

coli serotype 0111 : B4) was obtained from Sigma-Aldrich<br />

Biotechnology. Radioimmunoassay kits for IL-1β and TNF-α were<br />

purchased from Scientific and Technological Development Center<br />

of PLA General Hospital. NO detection kit was obtained from<br />

Nanjing Jiancheng Bioengineering Institute. Assay kits for alanine<br />

aminotransferase (ALT), aspartate aminotransferase (AST), alka-<br />

Shao M et al. Effects of Ligustilide… <strong>Planta</strong> Med 2011; 77: 809–816<br />

Fig. 1 Chemical structure<br />

formula of ligustilide.<br />

line phosphatase (ALP), γ-glutamyltransferase (GGT), creatinine<br />

kinase (CK), lactate dehydrogenase (LDH), blood urea nitrogen<br />

(BUN), serum creatinine (Scr), total protein (TP), and total bilirubin<br />

(T. BIL) were purchased from Zhongsheng Beikong Bioscience<br />

and Technology, Ltd.<br />

Experimentally induced endotoxic shock<br />

All the animals were allowed to acclimatize tothe conditions atour<br />

laboratory facility for at least 7 days before being used for the<br />

study. The rabbits were housed under standard laboratory conditions<br />

(25 ± 2°C, 50–55% relative humidity, and 12-h light-dark<br />

cycle) and were provided ad libitum access to food and water. The<br />

experiments were conducted in accordance with the ethical<br />

guidelines for investigations on laboratory animals and were approved<br />

by the Ethics Committee for Animal Experimentation.<br />

The rabbits were administered intraperitoneal injections of<br />

40 mg/kg (5 mL/kg) pentobarbital and maintained under anesthesia<br />

throughout the experiment. A venous cannula linked to a<br />

3-way joint was inserted into the arteria carotis communis of<br />

the rabbits; this cannula was used for blood sampling and was<br />

connected to an 8-channel physiological recorder to measure<br />

mean arterial blood pressure (MAP) via a pressure capsule. Heparin<br />

(0.3%) was used as an anticoagulant in this closed system.<br />

Heart rate (HR) and rectal temperature (RT) were also recorded<br />

during the experiments. The rabbits in the LPS group were injected<br />

with 300 µg/kg (1 mL/kg) of LPS via the ear vein. Additionally,<br />

within 20 minutes of the LPS injection, the rabbits in the<br />

treatment groups received ligustilide injection at 80, 40, or<br />

20 mg/kg (8 mL/kg), via the ear vein; the dexamethasone group<br />

received dexamethasone (5 mg/kg, 8 mL/kg) after the administration<br />

of LPS. The normal group was not administered LPS and<br />

received only an equal volume of sodium chloride solution at<br />

the same time the other groups were injected with LPS.<br />

Assays for vital parameters<br />

The vital parameters, namely MAP level, HR, and RT, were recorded<br />

before and 30, 60, 120, 180, 240, and 300 minutes after<br />

the injection.<br />

TNF-α, IL-1β, and NO assay<br />

Blood samples (1 mL) were separately collected from the arteria<br />

carotis communis before and at 30, 60, 120, 180, 240, and 300<br />

minutes after the injection. The blood samples were then centrifuged<br />

(2500 rpm) to obtain the serum. TNF-α and IL-1β levels in<br />

the serum were measured using radioimmunoassay kits, and the<br />

NO level was determined by the nitrate reductase (NR) method,<br />

according to the manufacturerʼs instruction.<br />

Downloaded by: University of British Columbia. Copyrighted material.


ALT, AST, ALP, GGT, CK, LDH, TP, Scr, T. BIL, and BUN assays<br />

Blood samples (2 mL) were collected from the arteria carotis<br />

communis before and 120 and 300 minutes after the administration<br />

of LPS. The blood samples were then centrifuged (2500 rpm)<br />

to obtain the serum. The activities of ALT, AST, ALP, GGT, CK, and<br />

LDH in the serum were measured. The levels of TP, Scr, BUN, and<br />

T. BIL were measured using the biuret method, trinitrophenol<br />

method, 2-points dynamics method, and diazo method, respectively,<br />

according to the respective manufacturersʼ instructions.<br />

Assay for formed elements of blood<br />

Blood samples were collected from the arteria carotis communis<br />

before and 120 and 300 minutes after the administration of LPS.<br />

A blood-cell counter was used to measure the hemoglobin level<br />

(HGB), red blood cell (RBC) count, hematocrit (HCT) value, white<br />

blood cell (WBC) count, leukocyte differential count (LDC), and<br />

platelet (PLT) count.<br />

Hemorheology assay<br />

Blood samples (4 mL) from the arteria carotis communis were<br />

collected in heparinized tubes 300 minutes after the administration<br />

of LPS. The samples were used to measure the specific viscosity<br />

of whole blood (VWB), specific viscosity of hematoplasma,<br />

and blood sedimentation rate.<br />

Correlated organs weight and histopathology<br />

Rabbits were killed by bloodletting from the arteria carotis communis<br />

300 minutes after the administration of LPS. Subsequently,<br />

the vital organs, including the heart, liver, lungs, and kidneys, of<br />

the rabbits of all the 6 groups were dissected and weighed. The<br />

ponderal indices were expressed in terms of grams per kilogram<br />

body weight. These organs were fixed with 10% formaldehyde in<br />

phosphate buffer saline for more than 8 hours, dehydrated in<br />

ethanol, and embedded in paraffin. Next, 4-µm-thick sections<br />

were cut, and the sections were deparaffinized in xylene. Subsequently,<br />

the tissue sections were stained with hematoxylin and<br />

eosin (HE) for light microscopy.<br />

Statistical analysis<br />

Data shown represent the mean and standard error of the mean<br />

(SEM). Group comparison was performed using one-way analysis<br />

of variance (ANOVA) with the post hoc Tukeyʼs test. Differences<br />

were considered statistically significant if p < 0.05.<br />

Supporting information<br />

Detailed information on isolation and purification of ligustilide<br />

and microscopic sections of organs are available as Supporting<br />

Information.<br />

Results<br />

!<br />

At all time points after 60 minutes, the MAP level in the LPS group<br />

was markedly less than that in the normal group (p < 0.01;<br />

l " Fig. 2A). Treatment with dexamethasone and ligustilide led to<br />

significant inhibition of the decline in the MAP level (p < 0.01 and<br />

p < 0.05, respectively). There was no change in the HR in any<br />

group at all time points (l " Fig. 2 B). RT in the LPS group was significantly<br />

less (p < 0.01) than that in the normal group and<br />

tended to decrease continuously after the rabbits were administered<br />

LPS (l " Fig. 2 C). For all time points, compared to the LPS<br />

group, the groups treated with dexamethasone (5 mg/kg) and 40<br />

and 80 mg/kg of ligustilide showed a significant decrease in RT<br />

(p < 0.01 and p < 0.05, respectively), while there was no conspicuous<br />

effect in the group treated with 20 mg/kg of ligustilide.<br />

The levels of TNF-α, IL-1β, and NO in the LPS group at each time<br />

point after the LPS injection was significantly (p < 0.01) higher<br />

than the corresponding levels in the normal group (l " Fig. 3). The<br />

levels of TNF-α and IL-1β reached the peak value at 120 minutes,<br />

while that of NO continued to increase even after 120 minutes.<br />

After dexamethasone (5 mg/kg) administration, an obvious decrease<br />

was noted in the level of TNF-α, IL-1β, and NO (p < 0.05<br />

and p < 0.01, respectively). Compared to the LPS group, the group<br />

treated with 80 mg/kg lingustilide showed significant suppression<br />

(p < 0.05 and p < 0.01, respectively) of the levels of TNF-α,<br />

IL-1β, and NO, while 40 mg/kg lingustilide showed a marked decrease<br />

in the level of NO (p < 0.05 or p < 0.01), but no apparent effect<br />

on the levels of TNF-α and IL-1β. Moreover, ligustilide<br />

(20 mg/kg) did not have any apparent effect on the levels of the<br />

above-mentioned cytokines.<br />

After the administration of the LPS injection, the HGB level and<br />

WBC and PLT counts in the LPS group were less than those in the<br />

normal group (l " Table 1). Dexamethasone (5 mg/kg) induced a<br />

remarkable increase in the WBC and PLT counts (p < 0.05), while<br />

ligustilide had no apparent effect on the count of formed elements<br />

of blood.<br />

Compared to the normal group, the LPS group showed markedly<br />

high serum levels of ALT, AST, ALP, GGT, LDH, CK, BUN, and Scr,<br />

but no change in the levels of T. BIL and TP, after the rabbits were<br />

administered LPS. Dexamethasone and ligustilide (20, 40, and<br />

80 mg/kg) treatment had an obvious inhibitory effect on the increase<br />

in the levels of ALT, AST, ALP, GGT, LDH, CK, BUN, and Scr,<br />

which was seen in the LPS group (l " Table 2).<br />

Compared to the normal group, the LPS group showed a markedly<br />

higher value of viscosity of whole blood (VWB), but plasma viscosity<br />

and blood sedimentation rate showed no obvious change<br />

after the rabbits were administered LPS (l " Fig. 4). The value of<br />

VWB after the administration of dexamethasone (5 mg/kg) and<br />

ligustilide (80, 40 mg/kg) was markedly lower than that in the<br />

LPS group.<br />

Compared to the normal group, the LPS group showed significantly<br />

increased (p < 0.05) ponderal indices of the lungs and kidneys,<br />

and slightly increased ponderal indices of the heart and liver.<br />

There was no difference between the 4 drug-treated groups<br />

and the LPS group in terms of the ponderal indices of the abovementioned<br />

tissues (l " Fig. 5).<br />

Compared to the normal group, the LPS group showed a considerably<br />

greater extent of injury to the heart, liver, lungs, and kidneys,<br />

while dexamethasone and ligustilide treatment brought<br />

about a marked reduction in the extent of LPS-induced injury<br />

(Figs. 2S‑5S, Supporting Information).<br />

Discussion<br />

!<br />

Original Papers<br />

Septic shock, which is an acute and severe clinical condition, is a<br />

therapeutic challenge for clinicians. Septic shock leading to multiorgan<br />

failure has remained one of the main reasons for postsurgical<br />

complications and death of patients [18, 19]. LPS is known to<br />

play a central role in the pathogenesis of septic shock. Although it<br />

does not directly cause septic shock, LPS triggers a set of inflammatory<br />

responses that mediate a series of pathophysiologic<br />

changes, such as hypoplasia, fever, tissue necrosis, vascular endothelium<br />

injury, and multiorgan dysfunction, by stimulating some<br />

Shao M et al. Effects of Ligustilide… <strong>Planta</strong> Med 2011; 77: 809–816<br />

811<br />

Downloaded by: University of British Columbia. Copyrighted material.


812<br />

Original Papers<br />

endogenous mediators of inflammation, and particularly by inducing<br />

excessive secretion of cytokines [8, 20,21].<br />

In the present study, the rabbits injected with LPS showed an<br />

obvious decrease in the MAP level at all time points and a continuous<br />

decrease in RT. These changes are characteristic of severe<br />

shock. According to the published clinical statistical data, in approximately<br />

50% of the fatal cases of infectious shock, the cause<br />

of death is hypotension with hyporesponsiveness to vasoconstrictor<br />

agents and multiple organ dysfunction and failure [18]. We<br />

observed that at all doses, ligustilide could inhibit the decrease<br />

in the MAP level; notably, the effect of high-dose (80 mg/kg)<br />

ligustilide was similar to that of dexamethasone (5 mg/kg). Furthermore,<br />

treatment with ligustilide (80 and 40 mg/kg) restored<br />

the RT of the animals in the 2 therapeutic groups compared with<br />

that in the LPS group.<br />

In LPS-induced endotoxic shock, TNF-α is one of the crucial inflammatory<br />

cytokines that induces rapid production of other in-<br />

Shao M et al. Effects of Ligustilide… <strong>Planta</strong> Med 2011; 77: 809–816<br />

Fig. 2 Effect of ligustilide on mean arterial pressure,<br />

heart rate, and rectal temperature in the LPSinjected<br />

rabbits. Data were collected at the indicated<br />

time points and analyzed (A, B,andC). Data<br />

are represented as means ± SEM (n = 7 for each<br />

group). # P


mortality in septic shock [9, 30]. Therefore, we also investigated<br />

the effect of ligustilide on the production of NO. Our results<br />

showed that ligustilide can inhibit NO production. Taken together,<br />

these results indicate that the anti-endotoxic shock effect of<br />

ligustilide may be attributed to its inhibitory effect on proinflammatory<br />

mediators.<br />

Ligustilide can also regulate the alterations in serum biochemistry<br />

parameters induced by injury to the vital organs. In the LPS<br />

group, the levels of serum AST, ALT, and GGT, which are clinical<br />

marker enzymes for liver damage, showed a rapid increase, suggesting<br />

acute hepatic tissue damage (l " Table 2). However, the<br />

levels of AST, ALT, and GGT in the ligustilide treatment groups<br />

are markedly lower than those in the LPS group. Similarly, the<br />

levels of serum BUN and Scr, which are clinical marker enzymes<br />

for kidney damage, were markedly lower in the ligustilide treatment<br />

groups than in the LPS group. The serum levels of LDH and<br />

CK are often used as quantitative indices of inflammation and or-<br />

Original Papers<br />

Fig. 3 Effect of ligustilide on TNF-α,IL-1β,andNO<br />

levels in the lipopolysaccharide-injected rabbits.<br />

After the rabbits received a lipopolysaccharide injection,<br />

some of them were administered ligustilide<br />

emulsion. Serum was collected at the indicated<br />

time points, and the levels of TNF-α,IL-1β, and NO<br />

were analyzed separately (A, B,andC). Data are<br />

represented as means ± SEM (n = 7 for each group).<br />

## P < 0.01 compared to the normal group,<br />

* p < 0.05, ** p < 0.01 compared to the LPS group.<br />

gan injury, especially injury to the heart. In the rabbits of the LPS<br />

group, the levels of LDH and CK increased conspicuously. However,<br />

ligustilide treatment (80, 40, or 20 mg/kg) suppressed the<br />

increase in the levels of LDH and CK. These results are consistent<br />

with the histologic findings (Fig. 2S–5S, Supporting Information).<br />

Moreover, a decrease in the specific viscosity of whole blood and<br />

count of formed elements of blood induced by ligustilide also<br />

confirms its protective effect in the rabbit model of LPS-induced<br />

shock.<br />

In summary, the findings of the present study clearly demonstrate<br />

that ligustilide, the main active component of Angelica<br />

sinensis roots, has a potent effect on LPS-induced endotoxic shock<br />

in rabbits. The protective effects of ligustilide could partly be attributed<br />

to its ability to inhibit proinflammatory mediators.<br />

Shao M et al. Effects of Ligustilide… <strong>Planta</strong> Med 2011; 77: 809–816<br />

813<br />

Downloaded by: University of British Columbia. Copyrighted material.


814<br />

Original Papers<br />

Table 1 Effects of ligustilide on hemoglobin level and complete blood count in rabbit models of endotoxic shock.<br />

Testing Testing Normal LPS Dexamethasone Ligustilide Ligustilide Ligustilide<br />

item time<br />

5 mg/kg<br />

80 mg/kg<br />

40 mg/kg<br />

20 mg/kg<br />

HGB Before 134.9 ± 1.9 130.9 ± 2.0 130.4 ± 1.5 127.0 ± 2.2 131.9 ± 1.9 130.3 ± 1.3<br />

(g/L) 120 min 130.4 ± 2.3 117.3 ± 4.2 # 125.4 ± 3.6 112.3 ± 7.7 124.0 ± 4.6 117.4 ± 4.8<br />

300 min 122.4 ± 5.5 110.9 ± 5.5 114.6 ± 7.9 108.3 ± 8.7 115.0 ± 6.6 107.7 ± 4.8<br />

RBC Before 6.1 ± 0.1 5.9 ± 0.1 6.0 ± 0.2 5.6 ± 0.2 5.8 ± 0.2 5.8 ± 0.1<br />

(× 1012 /L) 120 min 5.8 ± 0.1 5.2 ± 0.2 5.8 ± 0.2 4.9 ± 0.4 5.6 ± 0.2 5.2 ± 0.3<br />

300 min 5.4 ± 0.2 4.9 ± 0.4 5.1 ± 0.4 4.7 ± 0.4 5.2 ± 0.3 4.7 ± 0.3<br />

HCT Before 39.2 ± 0.5 38.8 ± 0.6 39.2 ± 0.5 38.2 ± 0.7 38.5 ± 0.7 38.7 ± 0.7<br />

(%) 120 min 38.2 ± 0.5 34.6 ± 1.7 37.9 ± 1.3 33.1 ± 2.6 37.5 ± 1.4 34.6 ± 1.8<br />

300 min 35.7 ± 0.9 32.2 ± 2.2 34.0 ± 2.5 31.9 ± 2.8 34.6 ± 2.1 31.3 ± 1.5<br />

WBC Before 7.0 ± 1.0 6.3 ± 0.8 6.2 ± 0.5 5.7 ± 0.5 6.0 ± 0.5 7.1 ± 0.8<br />

(× 109 /L) 120 min 5.6 ± 1.2 2.8 ± 0.5 3.4 ± 0.5 2.9 ± 0.5 2.4 ± 0.4 2.3 ± 0.4<br />

300 min 5.2 ± 1.0 2.1 ± 0.6 # 4.2 ± 0.6* 3.0 ± 0.6 3.0 ± 0.4 3.0 ± 0.5<br />

LDC Before 31.3 ± 0.8 30.8 ± 0.7 31.2 ± 0.9 30.1 ± 0.8 30.8 ± 0.8 29.6 ± 0.6<br />

(%) 120 min 31.4 ± 1.2 31.9 ± 1.3 33.0 ± 0.4 32.8 ± 0.6 33.6 ± 1.1 33.8 ± 0.5<br />

300 min 31.5 ± 1.0 33.0 ± 1.3 31.7 ± 0.9 32.6 ± 0.5 33.9 ± 1.2 33.8 ± 1.1<br />

PLT Before 367.3 ± 13.2 374.7 ± 11.0 388.4 ± 11.1 358.0 ± 4.6 372.0 ± 9.1 381.9 ± 11.1<br />

(× 109 /L) 120 min 357.6 ± 11.2 325.0 ± 7.5 # 358.7 ± 13.1* 325.3 ± 17.5 357.1 ± 14.8 332.9 ± 18.8<br />

300 min 322.0 ± 11.0 326.4 ± 15.4 347.0 ± 14.1 316.7 ± 17.8 323.9 ± 15.6 319.1 ± 12.8<br />

Data are represented as means ± SEM (n = 7 for each group). # P < 0.05 compared to the normal group, * p < 0.05 compared to the LPS group<br />

Table 2 Effects of ligustilide on biochemical parameters in the rabbit models of endotoxic shock.<br />

Testing Testing Normal LPS Dexamethasone Ligustilide Ligustilide Ligustilide<br />

item time<br />

5 mg/kg<br />

80 mg/kg<br />

40 mg/kg<br />

20 mg/kg<br />

ALT Before 53.7 ± 6.3 56.5 ± 6.1 57.6 ± 7.4 56.8 ± 3.7 56.6 ± 6.7 60.9 ± 8.9<br />

(U/L) 120 min 55.1 ± 6.0 81.6 ± 7.1 # 61.6 ± 5.1** 62.0 ± 4.2* 58.3 ± 3.9** 64.1 ± 5.4<br />

300 min 57.9 ± 4.6 84.6 ± 6.1 ## 63.5 ± 5.2** 65.5 ± 5.1* 56.4 ± 3.8** 61.7 ± 5.7*<br />

AST Before 41.6 ± 2.5 41.5 ± 2.1 42.0 ± 2.6 41.4 ± 4.3 40.8 ± 3.3 41.0 ± 3.8<br />

(U/L) 120 min 44.8 ± 2.9 106.5 ± 7.2 ## 76.2 ± 7.5* 70.8 ± 7.8** 69.1 ± 6.7** 83.7 ± 6.7*<br />

300 min 42.1 ± 4.1 106.2 ± 8.5 ## 91.2 ± 7.1 76.2 ± 7.5* 76.2 ± 4.5** 86.6 ± 4.0<br />

ALP Before 157.1 ± 7.7 152.5 ± 7.3 155.0 ± 10.1 156.4 ± 9.0 154.2 ± 8.4 154.0 ± 9.4<br />

(U/L) 120 min 150.9 ± 10.0 207.4 ± 10.3 ## 161.0 ± 10.5** 166.7 ± 9.9* 164.3 ± 9.4** 174.5 ± 8.8*<br />

300 min 156.6 ± 6.2 249.8 ± 10.2 ## 172.1 ± 12.8** 170.3 ± 9.4** 173.6 ± 9.7** 187.2 ± 7.7**<br />

GGT Before 14.1 ± 0.4 13.4 ± 0.9 12.6 ± 0.5 12.9 ± 0.7 13.7 ± 0.6 14.0 ± 0.5<br />

(U/L) 120 min 13.6 ± 0.4 16.5 ± 0.9 # 14.2 ± 0.6* 13.9 ± 0.6* 14.9 ± 0.7 15.1 ± 0.5<br />

300 min 13.4 ± 0.5 22.0 ± 1.1 ## 15.8 ± 0.5** 14.2 ± 0.6** 17.5 ± 0.9** 17.2 ± 0.7**<br />

T. BIL Before 7.2 ± 0.3 7.0 ± 0.3 7.2 ± 0.3 7.0 ± 0.3 7.0 ± 0.4 7.2 ± 0.2<br />

(µM) 120 min 6.8 ± 0.5 6.8 ± 0.3 7.3 ± 0.4 7.4 ± 0.3 7.2 ± 0.2 6.8 ± 0.3<br />

300 min 7.4 ± 0.4 7.4 ± 0.2 6.8 ± 0.3 6.7 ± 0.3 7.0 ± 0.4 7.0 ± 0.3<br />

LDH Before 160.3 ± 10.7 165.7 ± 11.0 163.6 ± 8.8 165.6 ± 13.8 158.6 ± 10.0 161.0 ± 7.1<br />

(U/L) 120 min 166.0 ± 10.0 232.1 ± 12.9 ## 187.5 ± 10.2* 176.9 ± 14.2* 177.9 ± 8.7** 189.6 ± 7.7*<br />

300 min 171.2 ± 8.1 256.1 ± 12.8 ## 210.8 ± 12.7* 215.2 ± 11.5* 207.8 ± 13.3* 226.4 ± 13.3<br />

CK Before 2052.0 ± 63.1 2201.6 ± 189.4 1937.0 ± 312.8 2016.6 ± 239.7 2027.8 ± 89.9 1950.6 ± 240.5<br />

(U/L) 120 min 2127.3 ± 70.1 4804.7 ± 302.6 ## 3391.8 ± 395.1* 3212.5 ± 285.8** 3442.3 ± 195.2** 3758.1 ± 248.2*<br />

300 min 2131.7 ± 53.0 6294.8 ± 440.3 ## 4798.4 ± 406.3* 4146.6 ± 254.2** 4271.2 ± 232.0** 5075.5 ± 431.5<br />

TP Before 63.8 ± 3.1 61.8 ± 4.3 63.4 ± 4.2 60.5 ± 3.2 64.6 ± 3.0 62.4 ± 2.8<br />

(g/L) 120 min 61.9 ± 3.4 59.8 ± 3.5 60.8 ± 4.5 60.6 ± 4.2 60.9 ± 1.1 59.4 ± 3.4<br />

300 min 60.2 ± 3.8 57.0 ± 3.4 55.1 ± 2.7 56.1 ± 2.3 56.0 ± 5.2 57.6 ± 2.3<br />

Urea Before 7.0 ± 0.3 6.8 ± 0.4 7.2 ± 0.6 6.9 ± 0.6 7.1 ± 0.5 7.1 ± 0.6<br />

(mM) 120 min 6.9 ± 0.2 9.9 ± 0.6 ## 8.3 ± 0.8 7.3 ± 0.8* 9.0 ± 0.5 9.3 ± 0.9<br />

300 min 7.1 ± 0.2 11.8 ± 1.0 ## 8.3 ± 0.7* 8.1 ± 0.8* 4.5 ± 0.7 11.0 ± 0.8<br />

Scr Before 85.1 ± 2.6 88.4 ± 4.3 84.8 ± 6.4 87.4 ± 2.7 84.2 ± 3.4 84.8 ± 4.6<br />

(µM) 120 min 87.5 ± 2.7 124.0 ± 8.9 ## 101.3 ± 5.9 91.5 ± 3.1** 92.2 ± 5.1** 94.8 ± 3.4**<br />

300 min 86.9 ± 2.2 144.5 ± 11.1 ## 105.0 ± 5.1** 103.1 ± 7.5** 105.0 ± 6.2** 107.9 ± 2.8**<br />

Data are represented as means ± SEM (n = 7 for each group). # P < 0.05, ## p < 0.01 compared to the normal group, * p < 0.05, ** p < 0.01 compared to the LPS group<br />

Shao M et al. Effects of Ligustilide… <strong>Planta</strong> Med 2011; 77: 809–816<br />

Downloaded by: University of British Columbia. Copyrighted material.


Acknowledgements<br />

!<br />

This study was supported by grants from the National Science &<br />

Technology Major Project (2009ZX 09303-003;2009ZX09103-<br />

034), the National Natural Sciences Foundation of China<br />

(30873063; 30873455), the National 973 Fundamental Project<br />

of China (2009CB523004), the Natural Sciences Foundation of<br />

Beijing (7092068), the Key Project of Youth Foundation of Institute<br />

of Materia <strong>Medica</strong>, Chinese Academy of <strong>Medica</strong>l Sciences<br />

and Peking Union <strong>Medica</strong>l College (2006QZH01), and the Program<br />

for Changjiang Scholars and Innovative Research Team in<br />

University (PCSIRT, contract grant sponsor: IRT0514).<br />

Affiliations<br />

1 College of Life Sciences, Jilin University, Jilin, P.R. China<br />

2 Key Laboratory of Natural Drugs Biosynthesis, Ministry of Health &<br />

Key Laboratory of Bioactive Substances and Resources Utilization of Chinese<br />

Herbal Medicine, Ministry of Education; Institute of Materia <strong>Medica</strong>,<br />

Chinese Academy of <strong>Medica</strong>l Science & Peking Union <strong>Medica</strong>l College,<br />

Beijing, P.R. China<br />

3 Chinese Academy of <strong>Medica</strong>l Science & Peking Union <strong>Medica</strong>l College,<br />

Fuwai Hospital & Cardiovascular Institute, Beijing, P.R. China<br />

4 Shandong Target-Drug Research Co., Ltd., Shandong, P.R. China<br />

References<br />

1 Bone RC, Grodzin CJ, Balk RA. Sepsis: a new hypothesis for pathogenesis<br />

of the disease process. Chest 1997; 112: 235–243<br />

2 Friedman G, Silva E, Vincent JL. Has the mortality of septic shock<br />

changed with time. Crit Care Med 1998; 26: 2078–2086<br />

3 Wheeler AP, Bernard GR. Treating patients with severe sepsis. N Engl<br />

J Med 1999; 340: 207–214<br />

Original Papers<br />

Fig. 4 Effect of ligustilide on whole blood viscosity<br />

(mPa/s) at high shear (200/s), middle shear (30/s),<br />

and low shear (3/s); plasma viscosity (mPa/s); and<br />

blood sedimentation rate (Mm/H) in the LPS-injected<br />

rabbits. After the rabbits received a lipopolysaccharide<br />

injection, some of them were administered<br />

ligustilide emulsion. Data are represented as<br />

means ± SEM (n = 7 for each group). # P


816<br />

Original Papers<br />

16 Liu L, Ning ZQ, Shan S, Zhang K, Deng T, Lu XP, Cheng YY. Phthalide lactones<br />

from Ligusticum chuanxiong inhibit lipopolysaccharide-induced<br />

TNF-α production and TNF-α mediated NF-κB activation. <strong>Planta</strong> Med<br />

2005; 71: 808–813<br />

17 Ma JL, Li HY, Dai RJ, Deng YL. Isolation and purification of ligustilide in<br />

the Radix Angelicae Sinensis. Yaopin Pingjia 2005; 2: 214–215<br />

18 Rackow EC, Astiz ME. Pathophysiology and treatment of septic shock.<br />

JAMA 1991; 266: 548–554<br />

19 Bone RC. Gram-positive organisms and sepsis. Arch Intern Med 1994;<br />

154: 26–34<br />

20 Karima R, Matsumoto S. The molecular pathogenesis of endotoxic<br />

shock and organ failure. Mol Med Today 1999; 5: 123–132<br />

21 Brealey D, Brand M, Hargreaves I, Heales S, Land J, Smolenski R. Association<br />

between mitochondrial dysfunction and severity and outcome of<br />

septic shock. Lancet 2002; 360: 219–223<br />

22 Heinzel FP, Perko RM, Ling P, Hakimi J, Schoenhaut DS. Interleukin-12 is<br />

produced in vivo during endotoxemia and stimulates synthesis of interferon.<br />

Infect Immun 1994; 62: 4244–4249<br />

23 Suzuki K, Tateda K, Matsumoto T, Gondaira F, Tsujimoto S, Yamaguchi K.<br />

Effects of interaction between Escherichia coli verotoxin and lipopolysaccharide<br />

on cytokine induction and lethality in mice. J Med Microbiol<br />

2000; 49: 905–910<br />

Shao M et al. Effects of Ligustilide… <strong>Planta</strong> Med 2011; 77: 809–816<br />

24 Enomoto N, Takei Y, Hirose M, Kitamura T, Ikejima K, Sato N. Protective<br />

effect of thalidomide on endotoxin-induced liver injury. Alcohol Clin<br />

Exp Res 2003; 27: 2S–6S<br />

25 Shirakawa F, Saito K, Bonagura CA, Galson DL, Fenton MJ, Webb AC,<br />

Auron PE. The human prointerleukin 1 beta genen requires DNA sequences<br />

both proximal and distal to the transcription start site for tissue-specific<br />

induction. Mol Cell Biol 1993; 13: 1332–1344<br />

26 Dinarello CA. Proinflammatory and anti-inflammatory cytokines as<br />

mediators in the pathogenesis of septic shock. Chest 1997; 112:<br />

321S–329S<br />

27 Cobb J, Danner R. Nitric oxide and septic shock. JAMA 1996; 275: 1192–<br />

1196<br />

28 Wolkow PP. Involvement and dual effects of nitric oxide in septic shock.<br />

Inflamm Res 1998; 47: 152–166<br />

29 Llorens S, Nava E. Cardiovascular diseases and the nitric oxide pathway.<br />

Curr Vasc Pharmacol 2003; 1: 335–346<br />

30 Novelli GP. Role of free radicals in septic shock. J Physiol Pharmacol<br />

1997; 48: 517–527<br />

Downloaded by: University of British Columbia. Copyrighted material.


The Known Immunologically Active Components<br />

of Astragalus Account for Only a Small Proportion<br />

of the Immunological Adjuvant Activity When<br />

Combined with Conjugate Vaccines<br />

Authors Feng Hong 1 , Weilie Xiao 2 , Govind Ragupathi 3 , Clara B. S. Lau 4 , Ping Chung Leung 4 , K. Simon Yeung 5 ,<br />

Constantine George 1 , Barrie Cassileth 5 , Edward Kennelly 2 , Philip O. Livingston 3<br />

Affiliations The affiliations are listed at the end of the article<br />

Key words<br />

l " Astragalus membranaceus<br />

l " Leguminosae<br />

l " botanicals<br />

l " conjugate vaccine<br />

l " cancer vaccine<br />

l " astragalosides<br />

l " saponin<br />

received July 2, 2010<br />

revised October 20, 2010<br />

accepted October 28, 2010<br />

Bibliography<br />

DOI http://dx.doi.org/<br />

10.1055/s-0030-1250574<br />

Published online December 2,<br />

2010<br />

<strong>Planta</strong> Med 2011; 77: 817–824<br />

© Georg Thieme Verlag KG<br />

Stuttgart · New York ·<br />

ISSN 0032‑0943<br />

Correspondence<br />

Philip O. Livingston, MD<br />

Melanoma and Sarcoma Service<br />

Department of Medicine<br />

Memorial Sloan-Kettering<br />

Cancer Center<br />

1275 York Avenue<br />

New York, NY 10065<br />

USA<br />

Phone: + 16 46888 23 76<br />

Fax: + 1646422 0453<br />

livingsp@mskcc.org<br />

Abstract<br />

!<br />

The 95% ethanol extract of Astragalus has been<br />

demonstrated to have potent activity as an immunological<br />

adjuvant when administered with vaccines<br />

of various types. We endeavor here to identify<br />

the components of this extract that are responsible<br />

for this adjuvant activity. Mice were immunized<br />

with KLH conjugated to cancer carbohydrate<br />

antigens globo H and GD3 and cancer peptide<br />

antigen MUC1 combined with different Astragalus<br />

fractions or with commercially available<br />

Astragalus saponins and flavonoids. The antibody<br />

responses against cancer antigens and KLH were<br />

quantitated in ELISA assays, and toxicity was calculated<br />

by weight loss. Astragalosides II and IV<br />

were the most active components, but the toxicity<br />

of these two differed dramatically. Astragaloside<br />

II was the most toxic Astragalus component<br />

with 5–10% weight loss at a dose of 500 µg while<br />

astragaloside IV showed no weight loss at all at<br />

Introduction<br />

!<br />

Many widely used botanicals are claimed to have<br />

immunostimulant effects, but clear evidence that<br />

they are able to augment immunological responses<br />

against defined antigens is lacking.<br />

Screening botanicals for immunomodulatory activity<br />

after oral ingestion is difficult due to unknowns<br />

such as bioavailability (depending in part<br />

on issues such as formulation and concomitant<br />

food intake), the amount of active ingredient in<br />

the botanical selected, optimal dose, and appropriate<br />

assay. As a prelude to testing popular botanical<br />

medicines as immune modulators after<br />

oral ingestion, we have chosen to test their immunological<br />

activity as immunological adjuvants,<br />

mixed with antigens and injected subcutaneously,<br />

using resulting antibody titers as a read-out.<br />

We hypothesized that this would permit us to<br />

Original Papers<br />

this dose, suggesting that astragaloside IV might<br />

be utilized as an immunological adjuvant in future<br />

studies. Several flavonoids also had significant<br />

adjuvant activity. However, when the activities<br />

of these known immunologically active components<br />

of Astragalus (and of endotoxin) are calculated<br />

based on the extent of their presence in<br />

the 95% ethanol extract, they provide only a small<br />

proportion of the immunological activity. This<br />

raises the possibility that additional uniquely active<br />

components of Astragalus may contribute to<br />

adjuvant activity, or that the adjuvant activity of<br />

Astragalus is greater than the activity of the sum<br />

of its parts.<br />

Abbreviations<br />

!<br />

BSA: bovine serum albumin<br />

HSA: human serum albumin<br />

KLH: keyhole limpet hemocyanin<br />

identify active botanicals and botanical fractions<br />

and to identify the active ingredient(s). Once<br />

identified, these ingredients could then be administered<br />

orally in bioavailability and immunogenicity<br />

studies.<br />

When the breast cancer carbohydrate antigen<br />

globo H [1] or glycoprotein antigen MUC1, or neuroectodermal<br />

cancer ganglioside GD3 are conjugated<br />

to KLH, mixed with an immunological adjuvant<br />

and administered subcutaneously (s. c.), the<br />

magnitude of the antibody responses against globo<br />

H, MUC1, and GD3 and against KLH depend<br />

largely on the potency of the adjuvant [2]. This<br />

mix of carbohydrate autoantigens, peptide xenoantigen,<br />

and protein xenoantigen, weak, moderate,<br />

and strong immunogens, reflects the range<br />

of antigens targeted in vaccines against cancer<br />

and infectious diseases, and the range of antigens<br />

encountered in life. We have previously used this<br />

Hong F et al. The Known Immunologically… <strong>Planta</strong> Med 2011; 77: 817–824<br />

817


818<br />

Original Papers<br />

s. c. immunization model in mice to screen the immunomodulatory<br />

activity of a wide range of immunological adjuvants [2] and<br />

seven widely used botanicals selected based on prevalence of use<br />

and reports describing immunomodulatory activity [3]. The seven<br />

botanicals selected for that initial study were Astragalus [4],<br />

Coriolus [5, 6], Echinacea [6,7], H-48 (a formula consisting of ten<br />

herbs) [8, 9], maitake [7, 10], β-glucan of yeast origin [11], and<br />

turmeric [7, 12]. We found that only the 95% ethanolic extract of<br />

Astragalus (but not the water extract), Coriolus extracts, and yeast<br />

β-glucan had potent adjuvant activity. We focus here on identifying<br />

the active components in the 95% ethanolic extract of Astragalus.<br />

Yang and colleagues have tested saponins extracted from a variety<br />

of botanical sources for hemolytic effect (a surrogate for toxicity)<br />

and have constructed vaccines containing ovalbumin as an<br />

antigen to measure their potency as immunological adjuvants<br />

when mixed with ovalbumin and injected subcutaneously [21].<br />

They have identified the saponins from Astragalus as having a<br />

particularly high activity/toxicity ratio. While these reports are<br />

suggestive (they are the reason we included Astragalus in our initial<br />

screen), they do not guide us in the identification of the<br />

Astragalus components most responsible for the potent adjuvant<br />

activity identified in these or our previous study. Based on the<br />

Yang study, clearly the saponin fraction of Astragalus has adjuvant<br />

activity. Our goal here was to determine whether all of the<br />

Astragalus adjuvant activity resides in the saponin fraction and<br />

if so, which of the multiple saponins in this fraction are most active.<br />

Since activity of most adjuvants is dose related and the dose<br />

that can be administered is determined by local and systemic toxicity,<br />

our study includes toxicity determinations as well.<br />

Materials and Methods<br />

!<br />

Botanicals (see l " Table 1)<br />

The root of Astragalus membranaceus (Fisch.) Bge. (purchased<br />

from an herbal store in Mainland China) was provided by the Institute<br />

of Chinese Medicine (ICM), Chinese University of Hong Kong.<br />

A voucher specimen (no. HK 40399) of the Astragalus membranaceus<br />

studied here was deposited at the Herbarium of Agriculture,<br />

Fisheries and Conservation Department in Hong Kong. For the<br />

preparation of Astragalus extracts, the raw herb was cut into<br />

small pieces and refluxed with 95% ethanol for an hour twice.<br />

The supernatants obtained were combined and dried using a<br />

rotary evaporator to give the 95% ethanolic extract. Three subfractions<br />

of the 95% ethanolic extract of Astragalus were further<br />

prepared by passing through a D101 resin column and successively<br />

eluted with water, 30% ethanol, and 95% ethanol to give<br />

the corresponding water, 30%, and 95% ethanolic subfractions.<br />

Yeast β-glucan (SBG) was provided by Biotec Pharmacon and<br />

was > 95% pure.<br />

Analysis of botanical composition<br />

HPLC PDA and LC‑MS technologies were used for the quantitative<br />

analysis. Standards isoquercitrin, isorhamnetin-3-O-glucoside,<br />

ononin, and calycosin were purchased from ChromaDex. Standards<br />

astragalosides I, II, and IV, and formononetin were obtained<br />

from Zhongxin Innova Laboratories. The purities of the standards<br />

are listed in l " Table 1. Standard calycosin-7-O-β-D-glucoside was<br />

isolated from 95% EtOH Astragalus extract using the following<br />

procedures. Dried Radix Astragali samples (5.0 g) were extracted<br />

with 95% aqueous ethanol at room temperature (3 × 100 mL).<br />

Hong F et al. The Known Immunologically … <strong>Planta</strong> Med 2011; 77: 817–824<br />

After the EtOH was removed in vacuo, the residue (1.52 g) was<br />

separated over reversed-phase C18 eluting with MeOH−H2O<br />

(1 : 4, 2 :3, 1:1, 3:1, and 0 : 1) to give five fractions (I–V). Fraction<br />

II (250.0 mg) was further separated by Sephadex LH-20 eluting<br />

with methanol to give four subfractions (A–D). Calycosin-7-O-β-<br />

D-glucoside (1) (10.2 mg) was obtained from subfraction B by recrystallization.<br />

Its structure was determined by 1D and 2D NMR<br />

spectra, and the purity was more than 98% determined by<br />

HPLC‑PDA technology.<br />

Standard calycosin-7-O-β-D-glucoside was isolated from 95%<br />

EtOH Astragalus extract using reversed-phase chromatography<br />

as previously described [13]. Its structure was determined by 1D<br />

and 2D NMR spectra, and the purity was more than 98% determined<br />

by HPLC‑PDA technology.<br />

HPLC grade acetonitrile was from J.T. Baker; HPLC grade methanol<br />

was from E. Merck; distilled water was further purified by<br />

Milli-Q system (Millipore). Polyvinylidene difluoride (PVDF) syringe<br />

filters with a pore size of 0.45 µm were procured from National<br />

Scientific Co.<br />

Stock solutions (1 mg/mL) for the nine standards were prepared<br />

by dissolving individual standards in HPLC grade MeOH. Working<br />

standard solutions containing each of the nine compounds were<br />

prepared by diluting the stock solutions with methanol to a series<br />

of proper concentrations. The solutions were brought to<br />

room temperature, and an aliquot of 10 µL was injected into HPLC<br />

or LC‑MS for analysis. Calibration curves were obtained by plotting<br />

the peak area vs. the concentration of the standard.<br />

Analytes including 95% ethanol crude extract, water subfraction<br />

F1, 30% ethanolic subfraction F2, and 95% ethanolic subfraction<br />

F3 were dissolved in HPLC grade MeOH and prepared into<br />

20 mg/mL, which were filtered through a 0.45 µm syringe filters<br />

before injection. 10 µL of each sample was injected to HPLC and<br />

LC‑MS for quantitative analysis of flavonoids and astragalosides,<br />

respectively. The mobile phase, both flavonoids and astragalosides,<br />

consisted of water (A) and MeCN (B) with a flow rate of<br />

1 mL/min. The mobile phase composition began with 0% B, followed<br />

by a linear increase to 10% B in 5 min, 10–55% B in<br />

30 min, 55–100% B in 10 min. The column for separation is Phenomenex<br />

Hydro C 18 column (4.6 × 250 mm, 5 mm) at 25°C. The<br />

peaks for the nine standards were baseline separated. For the six<br />

flavonoids, separations were carried out on a Waters 2695 HPLC<br />

equipped with a PDA dectector. For astragalosides I, II, and IV,<br />

LC‑MS was performed on a Thermo Finnigan LCQ mass spectrometer<br />

in the positive mode with a Waters 2690 separations module.<br />

The instrument was equipped with an atmospheric pressure<br />

chemical ionization (APCI) source and controlled by Xcalibur<br />

software. The discharge current was set to 5 µA. The vaporizer<br />

and capillary temperatures were set to 450 and 200 °C, respectively.<br />

Nitrogen was used as the sheath gas and auxiliary gas at<br />

flow rates of 80 and 10 units, respectively. A mass range of 200–<br />

2000 amu was scanned.<br />

Vaccine production<br />

Globo H hexasaccharide (molecular weight 1055 Da) was synthesized<br />

and conjugated to KLH (8 × 10 6 Da) essentially as previously<br />

described [12]. Globo H/KLH molar ratios in the conjugate ranging<br />

between 500/1 and 800/1 were used in these studies. The globo<br />

H‑KLH used here was purchased from Optimer Pharmaceuticals<br />

who synthesized it under contract. It was provided as globo<br />

H ceramide for use as a target in ELISA assays and as globo H‑KLH<br />

conjugate for vaccine production. GD3 was extracted from bovine<br />

buttermilk, purchased from Matreya, Inc. and conjugated to KLH


Table 1 Botanicals tested with vaccines: source, dose, and active ingredients.<br />

as previously described [14, 15]. The GD3/KLH molar ratio in the<br />

conjugate was 950/1. MUC1 peptide containing 1 ½ tandem repeats<br />

(32 amino acids) was conjugated to KLH using the heterobifunctional<br />

linker MBS as previously described [16]. KLH for vaccine<br />

production and serological target was purchased from Sigma.<br />

OPT-821 (> 95% pure QS-21) at a dose of 20 µg and provided<br />

by Optimer Pharmaceuticals or yeast β-glucan [3] at a dose of<br />

250 µg were used as positive controls. The Astragalus fractions<br />

or purified components were used at doses between 4 µg and<br />

2 mg in individual experiments as indicated.<br />

Vaccine administration<br />

Six-week-old female C57Bl/6 mice were obtained from the Jackson<br />

Laboratory. Groups of 5 mice were immunized s. c. three<br />

times at 1-week intervals with globo H‑KLH containing 3–5µg<br />

of globo H, GD3-KLH containing 5 µg GD3, or MUC1-KLH containing<br />

1 µg MUC1 mixed with either Astragalus fractions or purified<br />

components in 0.1 mL saline, 20 µg OPT-821 as a positive control.<br />

The Memorial Sloan-Kettering Cancer Centerʼs (MSKCC) Animal<br />

Care and Use Program operates in compliance with all applicable<br />

federal, state, and local regulations. Specifically, the program<br />

complies with the Public Health Service Policy on Humane Care<br />

and Use of Laboratory Animals and has an assurance, # A3311-<br />

01, on file with the Office of Laboratory Animal Welfare. MSKCC<br />

is a registered research institution with the United States Department<br />

of Agriculture (registrant # 21-R-0072) and is fully accredited<br />

by the Association for the Accreditation of Laboratory Animal<br />

Care International.<br />

Original Papers<br />

Botanical extract Source Lot # Purity<br />

Dose Probable active<br />

by HPLC<br />

ingredients<br />

Astragalus (A. membranaceus)<br />

water crude extract<br />

ICM 500 µg No activity<br />

Astragalus (A. membranaceus)<br />

50% ethanol crude extract<br />

ICM 500 µg Saponins<br />

Astragalus (A. membranaceus)<br />

95% ethanol crude extract<br />

ICM 0.5–2 mg Saponins<br />

Astragalus 95% ethanol crude extract<br />

– water subfraction F1<br />

ICM 2 mg Saponins<br />

Astragalus 95% ethanol crude extract<br />

– 30% ethanolic subfraction F2<br />

ICM 2 mg Saponins<br />

Astragalus 95% ethanol crude extract<br />

– 95% ethanolic subfraction F3<br />

ICM 100 µg – 2 mg Saponins<br />

Standards Source Lot # Purity Dose Probable active<br />

ingredients<br />

Calycosin-7-O-β-D-glucoside Weilie Xiao 98% 20–100 µg Flavonoid<br />

Astragalosides I Chromadex 111195-(607) 99.0% 20–500 µg Saponin<br />

Astragalosides II Chromadex 11196-(607) 99.5% 4–500 µg Saponin<br />

Astragalosides IV Chromadex 11197-(203) 99.9% 20–500 µg Saponin<br />

Calycosin Chromadex 3 071-(109) 93.1% 20–100 µg Flavonoid<br />

Formononetin Chromadex 6 190-(971) 91.0% 20–500 µg Flavonoid<br />

Isoquercitrin Chromadex 9 505-(311) 91.2% 500 µg Flavonoid<br />

Isorhamnetin-3-b-glucoside Chromadex 9 535-(509) 500 µg Flavonoid<br />

Ononide ononin Chromadex 15371-(206) 99.9% 500 µg Flavonoid<br />

Yeast β-glucan Biotec Pharmacon 100 µg – 2mg β-1,3 backbone<br />

Positive controls<br />

β-glucan<br />

β-1,6 side chains<br />

OPT-821 Quillaja saponaria, fraction 21 Optimer Pharmaceuticals > 99% 20 µg Saponin<br />

Serological assays<br />

Mice were bled from the retro-orbital sinus under general anesthesia<br />

seven days after the third immunization for ELISA, and sera<br />

was frozen for future testing.<br />

ELISA: Enzyme linked immunosorbent assays were performed as<br />

described previously [14, 15]. The target antigens are globo H ceramide,<br />

GD3, MUC1 peptide, or KLH. To determine the titers of<br />

antibodies, ELISA plates were coated with antigen, generally at<br />

0.1 µg/well. Serially diluted sera in 1% HSA in PBS were added to<br />

wells of the coated plate and incubated for 1 hour at room temperature.<br />

Goat anti-mouse IgM or IgG conjugated with alkaline<br />

phosphatase (Southern Biotechnology) serve as second antibodies.<br />

The antibody titer is defined as the highest serum dilution<br />

showing an absorbance of 0.1 or greater over that of normal sera.<br />

A response is considered positive by ELISA if the titer of reactivity<br />

increased from undetectable pretreatment to at least 1 : 40 after<br />

vaccination, or in case of detectable pretreatment, by 8-fold.<br />

Endotoxin assay<br />

Endotoxin levels in the botanical samples were tested at Charles<br />

River Laboratories, using their Endosafe ® PTS system.<br />

Statistical analysis<br />

For each experimental run, results in serological assays for treated<br />

mice were compared to the no-adjuvant controls using the<br />

Mann-Whitney test or Studentʼs t-test.<br />

Hong F et al. The Known Immunologically… <strong>Planta</strong> Med 2011; 77: 817–824<br />

819


820<br />

Original Papers<br />

Results<br />

!<br />

The adjuvant activity of Astragalus fractions was determined.<br />

After vaccination with globo H‑KLH plus or minus crude Astragalus<br />

extracts, we found that the entire potency of Astragalus as immunological<br />

adjuvant with globo H‑KLH was present in the 95%<br />

ethanol crude extract portion as indicated in l " Table 2. This<br />

strongly suggested that activity was concentrated in the saponin<br />

or possibly flavonoid components. Significant augmentation of<br />

anti-KLH antibody titers using the 95% ethanol crude extract of<br />

Astragalus was confirmed in the 500 µg-2000 µg dose range in 9<br />

of 11 subsequent experiments, though augmentation of antibody<br />

titers against globo H or GD3 was not seen. This 95% ethanol<br />

crude extract of Astragalus was further fractionated into three<br />

subfractions (eluted by water, 30% ethanol, or 95% ethanol), and<br />

the individual fractions tested. Each of these 3 fractions significantly<br />

augmented antibody titers against KLH in 2 of 3 experiments<br />

but in no case were antibody titers higher than with the<br />

same dose of the initial 95% ethanol “crude” Astragalus extract.<br />

The saponin (astragaloside) and flavonoid content of these fractions<br />

was quantified using HPLC and available known astragaloside<br />

and flavonoid positive controls, as summarized in l " Table 3.<br />

While neither could be detected in subfraction F1, subfraction F3<br />

contained the astragalosides and most of the flavonoids, and subfraction<br />

F2 contained a high concentration of the flavonoid calycosin-7-O-β-D-glucoside.<br />

The commercially available purified saponin components of Astragalus<br />

are astragaloside I, astragaloside II, and astragaloside IV.<br />

Seven experiments were conducted with these astragalosides.<br />

l " Table 4 demonstrates that all 3 astragalosides are able to augment<br />

the antibody response against both MUC1 and KLH when<br />

injected with MUC1-KLH. Overall, astragaloside I significantly<br />

augmented KLH antibody titers in 2 of 4 experiments and astragaloside<br />

IV in 3 of 5 experiments. In most cases, significant reactivity<br />

was limited to the 500 µg dose level. Astragaloside II demonstrated<br />

significant elevations in titers against KLH in 6 of 6 experiments,<br />

4 of 6 experiments, and 3 of 6 experiments at the<br />

500 µg, 100 µg, and 20 µg dose levels, respectively. In 2 of 6 experiments,<br />

the anti-KLH titers induced with astragaloside II at<br />

the 500 µg dose level approached the titers of (were not significantly<br />

lower than) those obtained with 20 µg of OPT-821. Despite<br />

the high anti-KLH antibody titers induced and the consistent induction<br />

of anti-GD3 titers between 1/80 and 1/320 when 20 µg of<br />

Table 3 Quantity of astragalosides and flavonoids in dried Astragalus root extracts.<br />

Table 2 ELISA antibody titers after vaccination with globo H‑KLH plus or<br />

minus crude Astragalus extracts.<br />

Globo H‑KLH<br />

vaccine plus<br />

Globo H IgM KLH IgG<br />

No adjuvant 800(3), 1600, 400, 800(2), 1 600,<br />

3200<br />

3200<br />

Water crude extract 400, 800, 1 600(2), 0, 400, 800(2), 3200<br />

500 µg<br />

3200<br />

50% ethanol crude 400(2), 800(2), 1600, 3200(2), 6400,<br />

extract 500 µg<br />

1600<br />

12800<br />

95% ethanol crude 800, 1600(2), 25 600(3), 51 200(2)<br />

extract 500 µg<br />

3200, 6400<br />

Yeast β-glucan 250 µg 800, 1600(2), 102 400, 819 200(2),<br />

3200, 12800 1 638 400(2)<br />

* Reciprocal antibody titer. Number in parentheses is the number of mice with that<br />

titer, if more than 1. ** Groups in bold are significantly (p ≤ 0.05) higher than the<br />

no adjuvant control group<br />

OPT-821 was used as adjuvant with GD3-KLH, in none of the experiments<br />

were astragalosides able to induce detectable antibodies<br />

against GD3.<br />

The commercially available purified flavonoid components of Astragalus<br />

are isoquercitrin, isorhamnetin-3-b-glucoside, ononin,<br />

calycosin, and formononetin. The adjuvant activities of these<br />

were tested in a series of experiments with MUC1-KLH as the<br />

antigen. MUC1 was highly immunogenic, as immunogenic as<br />

KLH, and a more sensitive indicator of adjuvant activity. The results<br />

of one experiment are summarized in l " Table 5. Astragalosides<br />

II and IV at a dose of 100 µg, and Astragalus 95% EtOH extract<br />

at a dose of 500 µg or 2 mg were the most active, though<br />

the flavonoids were also active in this experiment. Overall, ononin<br />

and isorhamnetin-3-O-glucoside were tested in a single experiment<br />

at a dose of 500 µg and failed to significantly elevate<br />

antibody titers against KLH while formononetin and isoquercitrin<br />

were positive at a dose of 500 µg against KLH in 1 of 2 experiments<br />

and at a dose of 100 µg induced antibodies against<br />

MUC1, but not KLH, in 1 experiment.<br />

To address the possibility that endotoxin contamination of the<br />

various extracts was responsible for some or all reactivity, several<br />

of the relevant extracts were sent to Charles River Laboratories<br />

for determination of endotoxin levels. All levels were less than<br />

0.04 EU per dose (see l " Table 5). When the 2 EU dose of endotox-<br />

Qi, 2006 [26] Yu, 2007 [27]<br />

Astragalus 95% Ethanol crude Water subfraction 30% Ethanolic 95% Ethanolic<br />

extract (µg/mg)<br />

F1 (µg/mg)<br />

subfraction F2 subfraction F3<br />

Astragalosides<br />

I 0.61 1.15 1.86 ± 0.06* ND** ND 21.15 ± 0.36*<br />

II 0.24 0.14 0.63 ± 0.06 ND ND 7.27 ± 0.23<br />

IV<br />

Flavonoids<br />

0.14 0.042 0.44 ± 0.03 ND ND 3.76 ± 0.02<br />

Calycosin 0.09 0.03 1.92 ± 0.05 ND ND 23.11 ± 0.14<br />

Calycosin-7-O-β-D-glucoside 0.35 0.43 1.34 ± 0.02 ND 16.41 ± 0.07* 9.03 ± 0.26<br />

Formononetin 0.059 0.022 1.59 ± 0.05 ND ND 12.03 ± 0.05<br />

Isoquercitrin 0.051 ND ND ND ND ND<br />

Isorhamnetin-3-O-β-glucoside 0.13 0.042 ND ND ND ND<br />

Ononin 0.092 0.101 1.14 ± 0.01 ND ND 6.69 ± 0.17<br />

* Standard deviation of triplicates. ** ND: not detectable < 0.005 µg/mg<br />

Hong F et al. The Known Immunologically … <strong>Planta</strong> Med 2011; 77: 817–824


Table 4 Median ELISA antibody titers after vaccination with MUC1-KLH plus<br />

or minus Astragalus extract and astragalosides.<br />

GD3-KLH vaccine plus: MUC1 IgG KLH IgG<br />

Pretreatment < 40(5) < 100(5)<br />

No adjuvant < 40(3), 40(2), 100(2), 400(6),<br />

80(2), 320, 640,<br />

1280<br />

1600, 6400<br />

95% Ethanol crude extract 2 mg 640(3), 1 280(2) 1600(5)<br />

95% Ethanol crude extract 500 µg 40, 320, 640(2),<br />

1280<br />

400(3), 1600(2)<br />

Astragaloside I 500 µg 160, 1 280, 400, 1 600(2),<br />

2560(2), 5120 6400(2)<br />

Astragaloside I 100 µg 320(4), 5 120 400(2), 1600<br />

(2), 6400<br />

Astragaloside II 100 µg 1280(3), 2560, 6400, 25600(3),<br />

5120<br />

102 400<br />

Astragaloside II 20 µg 320, 640,<br />

1280(2), 5120<br />

400, 1 600(4)<br />

Astragaloside IV 100 µg 1280, 2 560(2),<br />

5120(2)<br />

1600(2), 6400(3)<br />

Astragaloside IV 20 µg 40, 80, 320(2), 400, 1 600(2),<br />

1280<br />

6400(2)<br />

OPT-821 20 µg 640, 1 280(2),<br />

2560, 5 120<br />

102, 400(5)<br />

* Reciprocal antibody titer of each mouse (number of mice at that titer). ** Groups in<br />

bold are significantly (p ≤ 0.05) higher than the 10 no adjuvant control mice<br />

in demonstrated significant adjuvant activity (l " Table 5), we performed<br />

an additional experiment to gage the impact of lower<br />

doses of endotoxin. Doses of 0.5 EU or lower had no significant<br />

adjuvant activity (l " Table 6).<br />

Weight loss after immunizations was used as a way to gauge toxicity.<br />

Significant weight loss (between 5% and 10% of body<br />

weight) was consistently seen with the 500 µg dose of astragaloside<br />

II in the 4 experiments where this was evaluated (see<br />

l " Fig. 1). The same applies to OPT-821 at the 20 µg dose level,<br />

but not to any of the other extracts tested in the experiments described<br />

here. This is an indication that astragaloside II and OPT-<br />

821 doses cannot be further escalated to gain immunogenicity.<br />

Doses of the other preparations could be further escalated safely,<br />

but doses here were limited by expense (all of the commercially<br />

available astragalosides and flavonoids) or volume (Astragalus<br />

extracts).<br />

Compared to the quantities of astragalosides and flavonoids described<br />

by Qi and Yu in whole Astragalus root preparations, the<br />

ethanol extraction procedure that we utilized did purify the astragalosides<br />

by several fold and most of the flavonoids by 4–30<br />

fold [26, 27]. Consistent adjuvant activity was demonstrated in<br />

our experiments with the 500 µg and 2 mg doses of this 95%<br />

EtOH extract of Astragalus. At the 1000 µg dose level, there are<br />

less than 2 µg of any of the major individual astragalosides or<br />

flavonoids known to be in Astragalus (see l " Table 3 under 95%<br />

EtOH extract). A mixture of the 6 astragalosides and flavonoids<br />

most highly expressed in the 95% EtOH extract of astragalus was<br />

tested at the 20 and 100 µg dose levels. Significantly increased<br />

antibody titers resulted against MUC1 at the lower mixture dose<br />

and against MUC1 and KLH at the 100 µg dose level (l " Table 5).<br />

The 100 µg dose level contained 23 µg of calycosin, formononetin,<br />

and astragaloside I, 15 µg of calycosin-7-O-β-D-glucoside, and<br />

8 µg of astragaloside II and astragaloside IV. None of these individual<br />

doses resulted in demonstrable adjuvant activity, so cer-<br />

Original Papers<br />

Table 5 Median ELISA antibody titers after vaccination with MUC1-KLH plus<br />

Astragalus extract, subfractions, astragalosides, flavonoids, or a mixture of<br />

astragalosides and flavonoids.<br />

MUC1-KLH MUC1 Mann- KLH Mann- Endotoxin<br />

vaccine plus IgG Whitney IgG Whitney EU/dose<br />

Calycosin 20 µg 320 0.214 400 0.608<br />

Calycosin 100 µg 640 0.048 1600 0.075 < 0.06 EU<br />

Calycosin-7-O-β-Dglucoside<br />

20 µg<br />

160 0.901 400 0.295<br />

Calycosin-7-O-β-Dglucoside<br />

100 µg<br />

160 0.901 400 0.366<br />

Formononetin 20 µg 320 0.138 400 0.366<br />

Formononetin<br />

100 µg<br />

1 280 0.016 1600 0.179 < 0.06 EU<br />

Astragaloside I<br />

100 µg<br />

320 0.069 1600 0.142<br />

Astragaloside I<br />

500 µg<br />

2 560 0.009 1600 0.058<br />

Astragaloside II 20 µg 1 280 0.016 1600 0.075<br />

Astragaloside II<br />

100 µg<br />

1 280 0.004 25600 0.002 0.018 EU<br />

Astragaloside IV<br />

20 µg<br />

320 0.268 1600 0.143<br />

Astragaloside IV<br />

100 µg<br />

2 560 0.003 6400 0.011 < 0.005 EU<br />

Astragalus 95% EtOH<br />

extract crude<br />

500 µg<br />

640 0.122 400 0.366 < 0.009 EU<br />

Astragalus 95% EtOH<br />

extract crude 2 mg<br />

640 0.026 1600 0.031 0.035 EU<br />

Astragalus 95% EtOH<br />

subfraction F1<br />

100 µg<br />

640 0.048 400 0.366<br />

Astragalus 95% EtOH<br />

subfraction F1<br />

500 µg<br />

320 0.048 400 0.366<br />

Astragalus 95% EtOH<br />

subfraction F2<br />

100 µg<br />

640 0.019 1600 0.075<br />

Astragalus 95% EtOH<br />

subfraction F2<br />

500 µg<br />

2 560 0.012 6400 0.011<br />

Astragalus 95% EtOH<br />

subfraction F3<br />

100 µg<br />

800 0.122 1600 0.432<br />

Astragalus 95% EtOH<br />

subfraction F3<br />

500 µg<br />

640 0.031 1600 0.020<br />

Mixture 20 µg 640 0.036 400 0.661<br />

Mixture 100 µg 1 280 0.026 1600 0.015<br />

OPT-821 20 µg 1 280 0.007 102400 0.002 < 0.005 EU<br />

Endotoxin 2 EU 2 560 0.004 6400 0.007<br />

No adjuvant<br />

(10 mice)<br />

60 400<br />

* Median antibody titer of 5 mice. ** Mixture is Calycosin, Calycosin-7-O-β-D-glucoside,<br />

formononetin, astragaloside I, astragaloside II, and astragaloside IV in weight of<br />

3, 2, 3, 3, 1, and 1<br />

tainly the 20 µg mixture dose and probably the 100 µg dose demonstrated<br />

more than additive reactivity.<br />

Hong F et al. The Known Immunologically… <strong>Planta</strong> Med 2011; 77: 817–824<br />

821


822<br />

Original Papers<br />

Table 6 Median ELISA antibody titers after vaccination with MUC1-KLH plus<br />

or minus Astragalus extract, astragaloside IV, OPT-821, and endotoxin.<br />

MUC1-KLH vaccine plus: KLH IgG Mann-Whitney<br />

Astragalus 95% EtOH crude extract 2 mg 25600 0.011<br />

Astragaloside IV 100 µg 25600 0.050<br />

OPT-821 20 µg 102400 < 0.001<br />

Endotoxin 0.5 EU 6400 0.259<br />

Endotoxin 0.1 EU 6400 0.125<br />

Endotoxin 0.02 EU 6400 0.178<br />

No adjuvant (10 mice) 6400<br />

* Median antibody titer of 5 mice<br />

Discussion<br />

!<br />

We have identified saponins and in particular the saponin fraction<br />

QS-21 and the semisynthetic saponin mix GPI-0100 as<br />

uniquely potent immunological adjuvants when mixed with conjugate<br />

vaccines containing glycolipids or peptides chemically<br />

conjugated to keyhole limpet hemocyanin (KLH) [2,17]. The maximal<br />

doses of QS-21 used in mice (20 µg) and patients (100 µg)<br />

were selected based on toxicity, acceptable weight loss in mice,<br />

and acceptable local erythema/induration and systemic flu-like<br />

symptoms in patients [18]. The quest continues for immunological<br />

adjuvants with more potent adjuvant activity and more limited<br />

local and systemic toxicities. While most studies have been<br />

focused on Quillaja saponaria saponins, there are many additional<br />

botanicals expressing other saponins. A possible case in point is<br />

Astragalus membranaceus which we have demonstrated previously<br />

[3] and demonstrate here to have significant adjuvant activity<br />

in the 95% ethanolic subfraction.<br />

A. membranaceus is a well-known traditional Chinese medicinal<br />

plant used widely for a variety of indications. The applicable part<br />

of Astragalus is the root. Astragalus root preparations contain a<br />

variety of constituents thought to be immunologically active including<br />

isoflavonoids and triterpene saponins (primarily multiple<br />

astragalosides), as well as a variety of other components including<br />

polysaccharides, multiple trace minerals, amino acids,<br />

and coumarins [19–23]. We have attempted to identify the components<br />

with greatest adjuvant activity in our model by further<br />

fractionating the 95% ethanolic Astragalus fraction into 3 fractions<br />

containing saponins, flavonoids, and the remainder, and<br />

Hong F et al. The Known Immunologically … <strong>Planta</strong> Med 2011; 77: 817–824<br />

by testing the commercially available components which include<br />

the 3 major saponins and most of the major flavonoids. We demonstrate<br />

here that the 95% ethanolic subfraction of A. membranaceus<br />

(the saponin rich fraction) augments antibody responses<br />

not only against strong xenoantigens such as KLH but also against<br />

moderately immunogenic peptides such as MUC1 and weak glycolipid<br />

autoantigens such as globo H by 4- to 20-fold in different<br />

experiments. At least 15 separate saponins have been identified<br />

among A. membranaceus saponins and their chemical structures<br />

defined [21]. Four of these (astragalosides I–IV), the most heavily<br />

expressed, are commercially available. While purifying the other<br />

11 individual saponins from extracted saponins for use as adjuvants<br />

would be difficult, the recent description of the total<br />

chemical synthesis of QS-21 [24] raises the possibility that these<br />

remaining individual A. membranaceus saponins could be synthesized.<br />

The described low hemolytic activity of these saponins<br />

and their potent adjuvant activity suggest that this might be a<br />

fruitful approach to identification of new adjuvants. It is expected<br />

that even among this family of saponins, some would have greater<br />

or lesser toxicity and that this might be distinct from their adjuvant<br />

activity.<br />

We have now conducted a quantitative analysis of the saponins<br />

and flavonoids present in the 95% ethanol fraction. Most of the<br />

major peaks have been identified and quantitated previously<br />

and are available commercially. Consequently, we were able to<br />

explore the immunological adjuvant potency of these components<br />

in the context of antibody response against our KLH conjugate<br />

vaccines. We confirmed the safety and adjuvant potency of<br />

astragalosides II and IV which are clearly the most immunologically<br />

active of the various components present in the 95% ethanol<br />

extract of Astragalus but also identified a variety of flavonoids<br />

with significant adjuvant activity. Each of these components<br />

were well tolerated at immunologically active doses with only astragaloside<br />

II at a dose of 500 µg inducing significant weight loss<br />

after each immunization.<br />

None of the flavonoids demonstrated detectable immunological<br />

activity in our model at doses of 100 µg or lower, and even astragalosides<br />

II and IV, the most active Astragalus components, demonstrated<br />

only low level activity at a dose of 20 µg and no detectable<br />

activity at a dose of 4 µg. The 500 µg dose of 95% ethanolic<br />

fraction of Astragalus contains less than 1.1 µg of astragalosides<br />

II and IV combined and less than 10 µg of the major flavonoids<br />

combined. When the eight active Astragalus components were<br />

Fig. 1 Toxicity study in C57BL/6J female mice injected<br />

s. c. with different doses of astragaloside<br />

compared to 20 µg OPT 821.


mixed together at the ratios detected in the 95% ethanolic extract<br />

and administered at different doses, activity was seen only at the<br />

100 µg total dose but not at the 20 µg dose. This represents 10fold<br />

the amount of this mixture in 500 µg of the 95% EtOH extract<br />

of Astragalus. These results suggest that the immunologic activity<br />

demonstrated by the 95% ethanolic extract of Astragalus is not<br />

determined by any one or two components but by the entire<br />

mix including components not yet detected or at least not identified<br />

here.<br />

The contribution of even low levels of contaminating endotoxin is<br />

always a concern when studying adjuvant activity. The low levels<br />

of endotoxin (0.035 and 0.018 EU) present in the Astragalus 95%<br />

EtOH and astragaloside II extracts, but not in astragaloside IV,<br />

were sufficient to contribute to adjuvant activity. While these<br />

levels of endotoxin were not high enough to explain all or even<br />

most of the adjuvant activity seen with these extracts, they may<br />

have been responsible for a small amount of the activity. Given<br />

the low levels of the known adjuvant active components in the<br />

95% ethanol Astragalus extract, it may be that there are additional<br />

active components yet to be defined. The recent report from<br />

Liu et al. [25] demonstrating that part of the immunological activity<br />

of Radix Astragali and Radix Hedysari resides in their polysaccharides<br />

is consistent with this possibility. It is also possible<br />

that the components are synergistic and more active than the<br />

sum of the immunologic activities of the component parts (including<br />

endotoxin), although this is less likely since the mixture<br />

of known components was not uniquely active.<br />

The consequences of these findings are that there will be no easy<br />

short cuts to determining batch-to-batch immunologic consistency<br />

for different batches of the same Astragalus preparations<br />

or batches obtained from different sources. It also suggests that<br />

studies aimed at determining the relevant bioavailability from<br />

an immunologic point of view of Astragalus after oral ingestion<br />

will be exceedingly difficult given the many individual components<br />

which contribute to activity. Finally, they suggest that astragaloside<br />

IV has a uniquely favorable adjuvant potency/toxicity<br />

ratio and might be an excellent immunological adjuvant for further<br />

study if it could be obtained more economically.<br />

Acknowledgements<br />

!<br />

This project was supported by Grant Number 1 P50 AT002779-01<br />

from the National Center for Complementary and Alternative<br />

Medicine (NCCAM) and the Office of Dietary Supplements<br />

(ODS). Its contents are solely the responsibility of the authors<br />

and do not necessarily represent the official views of the NCCAM,<br />

ODS, or the National Institute of Health.<br />

The authors would like to thank Professor Cao Hui (National Engineering<br />

Research Centre for Modernization of Traditional Chinese<br />

Medicine, Guangdong, China) for the morphological identification<br />

of the root of Astragalus membranaceus.<br />

Affiliations<br />

1 Department of Medicine, Laboratory of Tumor Vaccinology,<br />

Memorial Sloan-Kettering Cancer Center, New York, New York, United States<br />

2 Department of Biological Sciences, Lehman College and<br />

The Graduate Center, City University of New York, Bronx, New York,<br />

United States<br />

3 Department of Medicine, Melanoma Sarcoma Service,<br />

Memorial Sloan-Kettering Cancer Center, New York, New York, United States<br />

4 Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin,<br />

Hong Kong<br />

5 Department of Medicine, Integrative Medicine Service,<br />

Memorial Sloan-Kettering Cancer Center, New York, New York,<br />

United States<br />

Original Papers<br />

References<br />

1 Ragupathi G, Park TK, Zhang S, Kim IJ, Graeber K, Adluri S, Lloyd KO, Danishefsky<br />

SJ, Livingston PO. Immunization of mice with the synthetic<br />

hexasaccharide Globo H results in antibodies against human cancer<br />

cells. Angew Chem Int Ed Engl 1997; 36: 125–128<br />

2 Kim SK, Ragupathi G, Cappello S, Kagan E, Livingston PO. Effect of immunological<br />

adjuvant combinations on the antibody and T-cell response<br />

to vaccination with MUC1-KLH and GD3-KLH conjugates. Vaccine<br />

2000; 19: 530–537<br />

3 Ragupathi G, Yeung KS, Leung PC, Lee M, Lau CB, Vickers A, Hood C, Deng<br />

G, Cheung NK, Cassileth B, Livingston P. Evaluation of widely consumed<br />

botanicals as immunological adjuvants. Vaccine 2008; 26: 4860–4865<br />

4 Ross GD, Vetvicka V, Yan J, Xia Y, Vetvickova J. Therapeutic intervention<br />

with complement and beta-glucan in cancer. Immunopharmacology<br />

1999; 42: 61–74<br />

5 [No authors listed]. Astragalus membranaceus. Monograph. Altern Med<br />

Rev 2003; 8: 72–77<br />

6 Chu KK, Ho SS, Chow AH. Coriolus versicolor: a medicinal mushroom<br />

with promising immunotherapeutic values. J Clin Pharmacol 2002;<br />

42: 976–984<br />

7 Spellman K, Burns J, Nichols D, Winters N, Ottersberg S, Tenborg M. Modulation<br />

of cytokine expression by traditional medicines: a review of<br />

herbal immunomodulators. Altern Med Rev 2006; 11: 128–150<br />

8 Kamiyama H, Takano S, Ishikawa E, Tsuboi K, Matsumura A. Anti-angiogenic<br />

and immunomodulatory effect of the herbal medicine “Juzentaiho-to”<br />

on malignant glioma. Biol Pharm Bull 2005; 28: 2111–2116<br />

9 Matsumoto T, Sakurai MH, Kiyohara H, Yamada H. Orally administered<br />

decoction of Kampo (Japanese herbal) medicine, “Juzen-Taiho-To”<br />

modulates cytokine secretion and induces NKT cells in mouse liver. Immunopharmacology<br />

2000; 46: 149–161<br />

10 Kodama N, Komuta K, Nanba H. Can maitake MD-fraction aid cancer<br />

patients? Altern Med Rev 2002; 7: 236–239<br />

11 Smith JESR. Immunomodulatory activities of mushroom glucans and<br />

polysaccharide-protein complexes in animals and humans, Chapter 6.<br />

London: Cancer Res UK 2002; 106<br />

12 Maheshwari RK, Singh AK, Gaddipati J, Srimal RC. Multiple biological activities<br />

of curcumin: a short review. Life Sci 2006; 78: 2081–2087<br />

13 Xiao W, Wang Q, Unachukwu U, Lau CB, Jiang B, Hong F, Leung PC, Motley<br />

T, Livingston P, Cassileth B, Kennelly E. Chemical and genetic assessment<br />

of commercial Radix Astragali (Astragalus spp.) by ion trap LC‑MS<br />

and nuclear ribosomal DNA sequence analyses. J Agric Food Chem, in<br />

press<br />

14 Ragupathi G, Cappello S, Yi SS, Canter D, Spassova M, Bornmann WG,<br />

Danishefsky SJ, Livingston PO. Comparison of antibody titers after immunization<br />

with monovalent or tetravalent KLH conjugate vaccines.<br />

Vaccine 2002; 20: 1030–1038<br />

15 Ragupathi G, Meyers M, Adluri S, Howard L, Musselli C, Livingston PO. Induction<br />

of antibodies against GD3 ganglioside in melanoma patients<br />

by vaccination with GD3-lactone-KLH conjugate plus immunological<br />

adjuvant QS‑21. Int J Cancer 2000; 85: 659–666<br />

16 Gilewski T, Adluri S, Ragupathi G, Zhang S, Yao TJ, Panageas K, Moynahan<br />

M, Houghton A, Norton L, Livingston PO. Vaccination of high-risk breast<br />

cancer patients with mucin-1 (MUC1) keyhole limpet hemocyanin<br />

conjugate plus QS‑21. Clin Cancer Res 2000; 6: 1693–1701<br />

17 Kim SK, Ragupathi G, Musselli C, Choi SJ, Park YS, Livingston PO. Comparison<br />

of the effect of different immunological adjuvants on the antibody<br />

and T-cell response to immunization with MUC1-KLH and GD3-KLH<br />

conjugate cancer vaccines. Vaccine 1999; 18: 597–603<br />

18 Livingston PO, Adluri S, Helling F, Yao TJ, Kensil CR, Newman MJ, Marciani<br />

D. Phase 1 trial of immunological adjuvant QS‑21 with a GM2 ganglioside-keyhole<br />

limpet haemocyanin conjugate vaccine in patients with<br />

malignant melanoma. Vaccine 1994; 12: 1275–1280<br />

19 Chu DT, Wong WL, Mavligit GM. Immunotherapy with Chinese medicinal<br />

herbs. II. Reversal of cyclophosphamide-induced immune suppression<br />

by administration of fractionated Astragalus membranaceus in<br />

vivo. J Clin Lab Immunol 1988; 25: 125–129<br />

20 Sun HX, Qin F, Ye YP. Relationship between haemolytic and adjuvant activity<br />

and structure of protopanaxadiol-type saponins from the roots<br />

of Panax notoginseng. Vaccine 2005; 23: 5533–5542<br />

Hong F et al. The Known Immunologically… <strong>Planta</strong> Med 2011; 77: 817–824<br />

823


824<br />

Original Papers<br />

21 Yang ZG, Sun HX, Fang WH. Haemolytic activities and adjuvant effect of<br />

Astragalus membranaceus saponins (AMS) on the immune responses to<br />

ovalbumin in mice. Vaccine 2005; 23: 5196–5203<br />

22 Leung AY, Foster S. Encyclopedia of common natural ingredients used in<br />

food, drugs and cosmetics, 2nd edition. New York, NY: John Wiley &<br />

Sons; 1996<br />

23 Upton RSC. Astragalus root: Analytical, quality control, and therapeutic<br />

monograph. Scotts Valley: American Herbal Pharmacopoeia; 1999:<br />

1–25<br />

24 Kim YJ, Wang P, Navarro-Villalobos M, Rohde BD, Derryberry J, Gin DY.<br />

Synthetic studies of complex immunostimulants from Quillaja saponaria:<br />

synthesis of the potent clinical immunoadjuvant QS-21Aapi. J Am<br />

Chem Soc 2006; 128: 11 906–11 915<br />

Hong F et al. The Known Immunologically … <strong>Planta</strong> Med 2011; 77: 817–824<br />

25 Liu J, Hu X, Yang Q, Yu Z, Zhao Z, Yi T, Chen H. Comparison of the immunoregulatory<br />

function of different constituents in radix astragali and<br />

radix hedysari. J Biomed Biotechnol 2010; 2010: 479426<br />

26 Qi LW, Yu QT, Li P, Li SL, Wang YX, Sheng LH, Yi L. Quality evaluation of<br />

Radix Astragali through a simultaneous determination of six major active<br />

isoflavonoids and four main saponins by high-performance liquid<br />

chromatography coupled with diode array and evaporative light scattering<br />

detectors. J Chromatogr A 2006; 1134: 162–169<br />

27 Yu QT, Qi LW, Li P, Yi L, Zhao J, Bi Z. Determination of seventeen main<br />

flavonoids and saponins in the medicinal plant Huang-qi (Radix astragali)<br />

by HPLC‑DAD-ELSD. J Sep Sci 2007; 30: 1292–1299


Chemical Composition, Acute Toxicity,<br />

and Antinociceptive Activity of the Essential Oil<br />

of a Plant Breeding Cultivar of Basil<br />

(Ocimum basilicum L.)<br />

Authors Antônio Medeiros Venâncio 1,2 , Alexandre Sherlley Casimiro Onofre 2 , Amintas Figueiredo Lira 2 ,<br />

Péricles Barreto Alves 3 , Arie Fitzgerald Blank 4 , Ângelo Roberto Antoniolli 2 , Murilo Marchioro 2 ,<br />

Charles dos Santos Estevam 2 , Brancilene Santos de Araujo 2<br />

Affiliations The affiliations are listed at the end of the article<br />

Key words<br />

l " Ocimum basilicum L.<br />

l " Lamiaceae<br />

l " lethal dose<br />

l " central nervous system<br />

l " central antinociception<br />

l " peripheral antinociception<br />

received June 7, 2010<br />

revised Nov. 7, 2010<br />

accepted Nov. 15, 2010<br />

Bibliography<br />

DOI http://dx.doi.org/<br />

10.1055/s-0030-1250607<br />

Published online December 14,<br />

2010<br />

<strong>Planta</strong> Med 2011; 77: 825–829<br />

© Georg Thieme Verlag KG<br />

Stuttgart · New York ·<br />

ISSN 0032‑0943<br />

Correspondence<br />

Brancilene Santos de Araujo<br />

Departamento de Fisiologia<br />

Universidade Federal de Sergipe<br />

Laboratório de Bioquímica<br />

de Produtos Naturais<br />

Campus São Cristovão<br />

CEP 49100-000<br />

Aracaju, SE<br />

Brasil<br />

Phone: + 55 7921 0566 47<br />

Fax: +557921056647<br />

bsa@ufs.br<br />

Abstract<br />

!<br />

Ocimum basilicum L. is an aromatic herb used in<br />

Brazil to treat illnesses such as respiratory and<br />

rheumatic problems, vomiting, and pain. In the<br />

present study, the chemical composition, acute<br />

toxicity, and antinociceptive effects of the essential<br />

oil (EO) of the cultivar “Maria Bonita” obtained<br />

from O. basilicum L. PI 197442 genotype<br />

were evaluated in Swiss mice (20–35 g each). Lethal<br />

dose to cause 50% death (LD50) was calculated<br />

from a dose-response curve (100–5000 mg/<br />

kg body wt.; n = 6) as 532 mg/kg body wt. In the<br />

acetic acid-induced writhing test (0.6% i. p.), EO<br />

(50, 100, and 200 mg/kg body wt., n = 8, s. c.) was<br />

Introduction<br />

!<br />

According to the World Health Organization, 80%<br />

of the world population use plants to treat illnesses,<br />

such as microbial and viral diseases, inflammation,<br />

and pain [1]. In this context, culinary<br />

herbs and their essential oils bring benefits to human<br />

health and can be consumed in natura or<br />

added to improve flavor, taste, and the therapeutic<br />

potential of food. Plants can produce several<br />

medicinal effects in the organism, such as antinociception.<br />

They are active against inflammation<br />

and pain by modulating genes, receptors, and enzymes<br />

involved in the inflammatory cascade.<br />

They can be used associated with nonsteroidal<br />

anti-inflammatory drugs (NSAIDs) to attenuate<br />

side effects during the treatment of mucosal erosion<br />

and ulcers in the intestine, perforation and<br />

hemorrhage, kidney damage, and increased blood<br />

pressure [2, 3].<br />

Ocimum (Lamiaceae) is a genus that comprises<br />

more than 150 species, which are distributed<br />

from tropical to subtropical regions [4–6]. The essential<br />

oil of many Ocimum species is used to treat<br />

headaches, diarrhea, helminthes infestation, in-<br />

Original Papers<br />

effective in reducing the abdominal contractions<br />

at all doses (48–78%). In the hot-plate test, EO significantly<br />

increased the latency at 50 mg/kg body<br />

wt. at all times (37–52%, n = 8, s.c.). However, the<br />

effects of morphine and EO at 50 mg/kg were reverted<br />

in the presence of naloxone, an opioid antagonist.<br />

In the formalin test, EO significantly reduced<br />

paw licking time in the first and second<br />

phases of pain at 200 mg/kg body wt. (38 and<br />

75%, respectively, n = 8, s. c.). The results suggested<br />

that the peripheral and central antinociceptive<br />

effects of EO are related to the inhibition<br />

of the biosynthesis of pain mediators, such as<br />

prostaglandins and prostacyclins, and its ability<br />

to interact with opioid receptors.<br />

flammation, and pain [1, 7,8]. It is also used in<br />

the pharmacy, perfume, and cosmetics industries<br />

because of its odorant, bactericide, fungicide, and<br />

insect repellent properties. A chromatographic<br />

analysis of the essential oils showed that plants<br />

from this genus are rich in volatile constituents<br />

such as linalool, geraniol, citral, alcanfor, eugenol,<br />

thymol, 1,8-cineole, and neryl acetate [1, 4–6].<br />

Ocimum basilicum L. is a perennial plant with yellow,<br />

white, and violet flowers, and oval leaves<br />

which have a sharp tip. This plant is cultivated<br />

and sold in Brazil as a culinary herb [9,10]. Extracts<br />

of its aerial parts and essential oil have repellent,<br />

antispasmodic, sedative, antioxidant,<br />

antimicrobial, antiviral, and cardiotonic activities,<br />

which are attributed to the presence of apigenin,<br />

linalool, and ursolic acid. It is active against respiratory<br />

and rheumatic problems, vomiting, and<br />

stomach aches [4, 9–12]. Although it is used to<br />

treat inflammation and pain, little is known about<br />

its antinociceptive mechanisms. In addition, there<br />

is little information about the acute toxicity of the<br />

plant, mainly its essential oil, despite the fact that<br />

its leaves and stems are used in the popular cuisine<br />

as an ingredient in salads and stuffing. A<br />

Medeiros Venâncio A et al. Chemical Composition, Acute … <strong>Planta</strong> Med 2011; 77: 825–829<br />

825


826<br />

Original Papers<br />

plant breeding study with a genotype of O. basilicum was performed<br />

in the Department of Agronomical Engineering of the<br />

Federal University of Sergipe in Brazil, resulting in one cultivar,<br />

named “Maria Bonita”, which was adapted for use in the northeast<br />

of Brazil [13]. In this study, we investigated the chemical<br />

composition, toxicity, and antinociceptive activity of the essential<br />

oil extracted from the leaves of “Maria Bonita”.<br />

Materials and Methods<br />

!<br />

Drugs and reagents<br />

Morphine sulfate (Dimorf ® , 99%) and naloxone hydrochloride<br />

(Narcan ® , 99%) were purchased from Cristália do Brasil S/A. Propylene<br />

glycol, formaldehyde, and acetic acid were from Vetec. All<br />

other drugs and reagents used were of analytical grade.<br />

Plant material<br />

Voucher specimens of the cultivar used in the present study were<br />

deposited in the Herbarium of the Federal University of Sergipe<br />

under the number 13162. They resulted of a plant breeding study<br />

with the O. basilicum PI 197442 genotype obtained from the<br />

Germplasm Bank at the North Central Regional Plant Introduction<br />

Station, Iowa State University, USA [13]. Briefly, the genotype<br />

was cultivated in the Experimental Field of the Federal University<br />

of Sergipe (EF‑FUS), Brazil, and plants of the first and second generations<br />

were selected based on their essential oil yield and linalool<br />

content to be introduced in cages without pollinating<br />

agents so that they could autopollinate. “Maria Bonita” (PI<br />

197442-S3-bulk 5) was one of the cultivars resulting of the autopollination<br />

and was chosen for the present study due to its highest<br />

essential oil yield and linalool content. Fresh leaves of the cultivar<br />

were obtained from the Medicinal Plant Garden at the<br />

EF‑FUS and were dried (45 °C) for use in the present study.<br />

Essential oil extraction and analysis<br />

The essential oil (EO) of the leaves (600 g) was extracted by hydrodistillation<br />

in a Klavenger apparatus to give 28.5 mL of EO<br />

with a yield of 4.8% (v/w) and density of 0.87 g/mL. The components<br />

of EO were determined by gas chromatography coupled to<br />

mass spectrometry (GC‑MS) using a Shimadzu DC17A instrument.<br />

The GC separation was performed on a column DB-5 ms<br />

(30 m × 0.25 mm i. d., film thickness 0.25 µm) with the following<br />

oven program: (a) 80 °C, (b) increase at a rate of 3°C/min up to<br />

180 °C, and (c) increase at a rate of 10 °C/min up to 300 °C. Sample<br />

injections (2 µL) were performed in the split mode (1 : 100) at<br />

250 °C using helium as the carrier gas (1 mL/min). The mass spectrometer<br />

ionization energy was 70 eV with a scan time of 1 s and<br />

temperatures of 200 °C (ion source) and 280 °C (analyzer).<br />

Animals<br />

Swiss male and female mice (20–35 g) were used in the present<br />

study. The animals were kept in plastic boxes at standard environmental<br />

temperature (25 ± 1 °C) and fed with food and water<br />

ad libitum. Feeding was interrupted 12 h before the antinociceptive<br />

assays. All mice were treated in accordance with the guidelines<br />

approved by the Ethics Committee for Research on Animals<br />

of the Federal University of Sergipe.<br />

Medeiros Venâncio A et al. Chemical Composition, Acute … <strong>Planta</strong> Med 2011; 77: 825–829<br />

Acute toxicity<br />

The 50% lethal dose (LD50) of EO was determined to find out the<br />

acute toxicity. A prior study was performed by intraperitoneal<br />

administration of aqueous solutions of EO to mice at 1000, 3000,<br />

and 5000 mg/kg body weight (n = 6 each group). Subsequently,<br />

groups of six mice received doses of 100, 250, 500, 1000, and<br />

1500 mg/kg body weight and were surveyed. On the first days,<br />

the animals were observed every 10 min followed by 24, 48, and<br />

72 h observations for any changes in spontaneous motor activity,<br />

gate, respiration, writhing, piloerection, etc., and up to 30 days for<br />

mortality. The LD 50 was determined by logarithmic regression<br />

analysis of the death numbers using the Probit method. Concentrations<br />

used in the pharmacological tests were below the LD50 to<br />

reduce the interferences of EO in the results due to its toxicity<br />

whose signs can be confused with the ones for antinociceptive<br />

behavior.<br />

Acetic acid-induced abdominal writhings in mice<br />

The antinociceptive effect was evaluated according to a method<br />

previously described [14]. Briefly, three experimental groups received<br />

EO at 50, 100, and 200 mg/kg (s. c., diluted in the vehicle<br />

50% propylene glycol in saline), while another group received indomethacin<br />

as the positive control (10 mg/kg body weight, s. c.).<br />

All animals received intraperitoneal injections of 0.6% acetic acid<br />

(0.1 mL/10 g body weight, i. p.) to induce abdominal muscle contractions<br />

and/or stretching of the legs 30 min after the treatment.<br />

The negative control groups received the vehicle (s. c.). After<br />

10 min of the acetic acid injection, the number of writhings was<br />

registered for 20 min for the mice in each group (n = 8).<br />

Hot plate test<br />

The antinociceptive assay was performed according to the classical<br />

hot-plate technique [15]. Briefly, the animals were treated<br />

with vehicle 50% propylene glycol in saline (s. c.), morphine<br />

(10 mg/kg body weight, s. c.), morphine (10 mg/kg body weight,<br />

s. c.) plus naloxone (5 mg/kg body weight, i. p.), EO extract (50,<br />

100, and 200 mg/kg body weight, s. c.), and EO (50 mg/kg, s. c.)<br />

plus naloxone (5 mg/kg, i.p.) 30 min before the beginning of the<br />

experiment. In the groups treated with naloxone, it was administered<br />

15 min before the treatment with morphine or EO. The latency<br />

time for the mice to lick their paws in each group (n = 8)<br />

was registered during the maximum period of 30 s at intervals<br />

of 0, 15, 30, and 60 min.<br />

Formalin test<br />

Animals (n = 8) were preadapted for 10 days to the surrounding<br />

environment before the formalin test took place [16]. They were<br />

treated with vehicle 50% propylene glycol in saline (s. c.), morphine<br />

(10 mg/kg body weight, s. c.), and EO extract (50, 100, and<br />

200 mg/kg body weight, s. c.) 30 min before the injection of 1%<br />

formalin (20 µL/animal, s. c.) in the hind paw. The negative control<br />

group received only vehicle. The pain reaction observations<br />

were registered immediately after the formalin injection utilizing<br />

a stopwatch for registering the time that the animals remained<br />

paw licking or biting during the first phase (0–5 min) and the second<br />

phase (20–25 min) of the test.<br />

Statistical analysis<br />

Results were expressed as mean ± SEM. Data were analyzed by<br />

ANOVA followed by Dunnettʼs test for comparison. Differences<br />

were considered significant when p < 0.05.


Results and Discussion<br />

!<br />

“Maria Bonita”, a cultivar of O. basilicum obtained in a plant<br />

breeding study, was not characterized beyond the increase in EO<br />

yield (from 2.5 to 4.8%) and linalool content (61.6 to 69.5%) [13].<br />

Therefore, the complete chemical composition, acute toxicity,<br />

and antinociceptive potential of EO from its leaves were investigated<br />

in the present study.<br />

Twenty-seven compounds were identified out of thirty-eight<br />

peaks in the EO GC‑MS chromatograms (l " Table 1). The essential<br />

oil consisted mainly of linalool, geraniol, 1,8-cineole, neryl acetate,<br />

and α-trans-bergamotene, representing 94.3%. The components<br />

were separated into aliphatic monoterpenes, cyclic monoterpenes,<br />

bycyclic monoterpenes, oxygenated monoterpenes, cyclic<br />

sesquiterpenes, bycyclic sesquiterpenes, oxygenated sesquiterpenes,<br />

and aliphatic secondary alcohols.<br />

The EO acute toxicity was investigated before the antinociceptive<br />

study. Our results revealed no signs of toxicity or mortality under<br />

250 mg/kg, while animal mortality rates ranged from 16.6 to<br />

100% with this and other concentrations. The LD50 was 532 mg/<br />

kg and may be related to the higher content of linalool because<br />

its reported mice intraperitoneal LD50 is well documented in the<br />

literature ranging from 200 to 1200 mg/kg [17]. Geraniol and 1,8cineole,<br />

also known for their toxicity, may be contributing to the<br />

total toxicity of EO. Therefore, EO in the therapeutic doses used in<br />

our study was not toxic, since the highest dose to produce antinociception<br />

(200 mg/kg) is less than LD50, which indicates its potential<br />

as a therapeutic agent.<br />

EO was significantly active (p < 0.01) in all tested concentrations,<br />

reducing muscle contractions induced by the acetic acid in a<br />

dose-dependent manner (48–78%, l " Fig. 1) when the antinociceptive<br />

effect on the abdominal writhings was evaluated. The intraperitoneal<br />

administration of acetic acid irritates the gastric serous<br />

membrane and produces abdominal writhings due to inflammation,<br />

which is the peripheral component of pain. The inflammatory<br />

response is associated with the release of mediators<br />

such as prostaglandins. The action of anti-inflammatory substances<br />

such as indomethacin results in the inhibition of the enzyme<br />

cyclooxygenase in the arachidonic acid pathway, preventing<br />

the biosynthesis of prostaglandins and prostacyclins and reducing<br />

pain [18, 19].<br />

In the hot-plate test, the antinociceptive potential is characterized<br />

by the behavior of the animal model (biting or licking the<br />

paw) as a response to an external pain stimulus, while the latency<br />

is the time (seconds) before this response emerges [20]. The hotplate<br />

test revealed that EO at 50 mg/kg significantly increased the<br />

time (37–52%, p < 0.05) for mice response to the thermal stimulus<br />

at all analyzed time points (l " Fig. 2 A–D), although it was not<br />

comparable to morphine (up to 151%, l " Fig. 2 B), the positive<br />

control of analgesia. Naloxone was used to determine if EO mimicked<br />

the morphine mechanism of action on opioid receptors.<br />

The effects of morphine and EO at 50 mg/kg were reverted in<br />

the presence of naloxone, which suggests that opioid receptors<br />

are involved in the EO antinociceptive action [19,21, 22].<br />

The EO effect on the first and second phase of pain during the formalin<br />

test was characterized by the reduction of the paw licking<br />

time after the formalin stimulus. The results suggested that its action<br />

on the central and peripheral components of pain might be<br />

dose-dependent because both pain phases were significantly affected<br />

(p < 0.05) only at the highest dose employed, 200 mg/kg,<br />

with 38% (33.97 ± 4.38 s) and 75% (9.13 ± 2.93 s) of paw licking<br />

time reduction, respectively (l " Fig. 3A–B). At 100 mg/kg, EO had<br />

Original Papers<br />

Table 1 Chemical composition of the “Maria Bonita” cultivar essential oil determined<br />

by GC‑MS analysis.<br />

Peak Rt (min) Compound % IRRP exp. IRR lit.<br />

1 4.700 α-pinene 0.19 933 939<br />

2 5.533 sabinene 0.28 972 976<br />

3 5.592 1-octen-3ol 0.07 975 978<br />

4 5.683 β-pinene 0.70 979 980<br />

5 5.850 myrcene 0.14 987 991<br />

6 6.967 limonene 0.20 1029 1031<br />

7 7.050 1,8-cineol 7.47 1035 1033<br />

8 7.375 (Z)-β-ocimene 0.13 1043 1040<br />

9 7.767 γ-terpinene 0.04 1057 1062<br />

10 8.125 – 0.04 1069 –<br />

11 8.583 iso-terpinolene 0.06 1085 1086<br />

12 8.708 – 0.05 1089 –<br />

13 8.992 linalool 69.54 1099 1098<br />

14 10.233 (E)-miroxide 0.13 1137 1142<br />

15 11.292 – 0.12 1169 –<br />

16 11.392 – 0.06 1172 –<br />

17 11.650 terpin-4-ol 0.10 1180 1177<br />

18 12.117 α-terpineol 0.76 1194 1189<br />

19 13.075 – 0.08 1282 –<br />

20 13.542 β-citral 0.11 1236 1240<br />

21 13.933 geraniol 12.55 1249 1255<br />

22 14.542 geraranial 0.18 1265 1270<br />

23 15.158 bornyl acetate 0.29 1283 1285<br />

24 16.458 – 0.10 1321 –<br />

25 16.983 – 0.04 1337 –<br />

26 18.275 neryl acetate 3.58 1375 1365<br />

27 18.725 β-elemene 0.07 1388 1391<br />

28 19.725 β-caryophyllene 0.12 1418 1418<br />

29 20.133 α-trans-bergamotene 1.17 1431 1436<br />

30 20.917 α-humulene 0.04 1455 1454<br />

31 21.100 cis-muurola-4(14)-5-diene 0.06 1460 1460<br />

32 21.717 germacrene-D 0.29 1479 1480<br />

33 21.800 – 0.07 1482 –<br />

34 22.375 – 0.06 1499 –<br />

35 22.567 – 0.06 1506 –<br />

36 22.733 γ-cadinene 0.33 1511 1513<br />

37 25.892 – 0.07 1613 –<br />

38 26.667 epi-α-cadinol 0.65 1639 1640<br />

Fig. 1 Effect of the essential oil of “Maria Bonita” Ocimum basilicum EO<br />

and indomethacin on the abdominal writhings induced by acetic acid<br />

(Dunnettʼs test; ** p < 0.01; n = 8). Abdominal writhing inhibitions for each<br />

treatment are indicated as a percentage.<br />

Medeiros Venâncio A et al. Chemical Composition, Acute … <strong>Planta</strong> Med 2011; 77: 825–829<br />

827


828<br />

Original Papers<br />

Fig. 2 Antinociceptive effect of “Maria Bonita” Ocimum basilicum EO and<br />

morphine on the pain response induced by heat in mice during the hot-plate<br />

test at 0 (A), 15 (B), 30 (C), and 60 (D) min (Dunnettʼs test; * p < 0.05;<br />

a significant antinociceptive effect only on the second phase of<br />

pain (p < 0.05), causing a reduction of 70% (10.73 ± 3.16 s) in the<br />

paw licking time (l " Fig. 3B). The first phase of pain is neurogenic<br />

and occurs through nociceptive neuronal activation by direct action.<br />

The second phase occurs through ventral horn neuronal activation<br />

at the spinal cord level, which is characterized by inflammation<br />

and sensitivity to NSAIDs [19, 22]. Drugs that present central<br />

action, such as narcotics (morphine), inhibit the two phases<br />

of pain, while peripheral drugs only inhibit the second phase [19,<br />

23]. Morphine and EO at 200 mg/kg revealed an antinociceptive<br />

effect on both phases, which suggests that EO has central and peripheral<br />

action.<br />

It is known that (−)-linalool, the main component of EO, causes<br />

multiple effects on the central nervous system such as removing<br />

nonselectively voltage-gated currents in sensory and nonsensory<br />

neurons [24]. (−)-Linalool affects β waves of the human brain [24,<br />

25] and inhibits the neurons in the cerebral cortex of glutamatergic<br />

mice [24]. (−)-Linalool has local anesthetic, anticonvulsant<br />

activities [26, 27], sedative effect on vertebrates, including humans<br />

[24, 25], and can significantly inhibit the carrageenan-induced<br />

paw edema in mice. The anti-inflammatory, antinociceptive,<br />

and antihyperalgic properties suggest that this compound<br />

can be used to reduce or eliminate pain associated with neuronal<br />

sensitization [28]. The antinociceptive activity seems to be associated<br />

with linalool acting on K + -ATP channels, which has an important<br />

role in pain modulation [28], and the antagonistic com-<br />

Medeiros Venâncio A et al. Chemical Composition, Acute … <strong>Planta</strong> Med 2011; 77: 825–829<br />

** p < 0.01; n = 8). Percentage indicates the increase in the latency time for<br />

each time and treatment.<br />

petition for the opioid receptor N-methyl-D-aspartate (NMDA)<br />

[26, 29,30]. The NMDA receptor inhibition produces antinociception<br />

[28]. The results of the present study combined with the<br />

ones of the literature suggest that linalool may be the component<br />

of EO responsible for its antinociceptive properties.<br />

The present study demonstrated that “Maria Bonita” EO at<br />

200 mg/kg can be considered of main interest because it showed<br />

a higher inhibition of the peripheral (75–78%) and central (38%)<br />

pain, mainly in the nociception caused by a chemical stimulus<br />

(acetic acid and formalin), while the concentration of 50 mg/kg<br />

was active in the thermal stimulus. The central activity observed<br />

in the thermal stimulus seems to be associated with opioid receptors<br />

due to the inhibition of the EO effect in the presence of<br />

naloxone, a know antagonist of these nociceptive receptors. The<br />

peripheral activity may be associated with the prevention of the<br />

biosynthesis of prostaglandins and prostacyclins, which are inflammatory<br />

mediators. Therefore, the essential oil of this plant<br />

breeding cultivar showed pharmacological potential.<br />

Acknowledgements<br />

!<br />

The authors want to thank specifically Prof. Dr. Arie F. Blank for donating<br />

the botanical material, Prof. Dr. Gileno de Sá Cardoso for<br />

hosting part of the experimental studies in his laboratory, and all<br />

the people who directly and indirectly cooperated with us.


Fig. 3 Antinociceptive effect of “Maria Bonita”Ocimum basilicum EO and<br />

morphine during the first (A) and second (B) phases of the formalin test<br />

(Dunnettʼs test; * p < 0.05; ** p < 0.01; n = 8). The reduction of the paw<br />

licking or biting time is indicated by percentage.<br />

Affiliations<br />

1 Secretaria de Estado da Saúde, Centro de Investigação Toxicológica,<br />

Aracaju, SE, Brazil<br />

2 Departamento de Fisiologia, Universidade Federal de Sergipe,<br />

São Cristovão, SE, Brazil<br />

3 Departamento de Química, Universidade Federal de Sergipe,<br />

São Cristovão, SE, Brazil<br />

4 Departamento de Engenharia Agronômica,<br />

Universidade Federal de Sergipe, São Cristovão, SE, Brazil<br />

References<br />

1 Adigüzel A, Gulluce M, Sengul M, Ogutcu H, Sahin F, Karaman I. Antimicrobial<br />

effects of Ocimum basilicum (Labiatae) extract. Turkish J Biol<br />

2005; 29: 155–160<br />

2 Dog TL. A reason to season: the therapeutic benefits of spices and culinary<br />

herbs. Explore J Sci Healing 2006; 2: 446–449<br />

3 Hajhashemi A, Sajjadi SE, Heshmati M. Anti-inflammatory and analgesic<br />

properties of Heracleum persicum essential oil and hydroalcoholic<br />

extract in animal models. J Ethnopharmacol 2009; 124: 475–480<br />

4 de Paula JP, Carneiro MRG, Paumgartten FJR. Chemical composition,<br />

toxicity and mosquito repellency of Ocimum selloi oil. J Ethnopharmacol<br />

2003; 88: 253–260<br />

5 Makonnen E, Debella A, Zerihun L, Abebe D, Teka F. Antipyretic properties<br />

of the aqueous and ethanol extracts of the leaves of Ocimum suave<br />

and Ocimum lamiifolium in mice. J Ethnopharmacol 2003; 88: 85–91<br />

Original Papers<br />

6 Telci I, Bayram E, Yilmaz G, Avci B. Variability in essential oil composition<br />

of Turkish basils (Ocimum basilicum L.). Biochem Syst Ecol 2006;<br />

34: 489–497<br />

7 Pessoa LM, Morais SM, Bevilaqua CM, Luciano JHS. Anthelmintic activity<br />

of essential oil of Ocimum gratissimum Linn. and eugenol against Haemonchus<br />

contortus. Vet Parasitol 2002; 109: 59–63<br />

8 Franca CS, Menezes FS, Costa LCB, Niculau ES, Alves PB, Pinto JEB, Marçal<br />

RM. Analgesic and antidiarrheal properties of Ocimum selloi essential<br />

oil in mice. Fitoterapia 2008; 79: 569–573<br />

9 Boskabady MH, Kiani S, Haghiri B. Relaxant effects of Ocimum basilicum<br />

on guinea pig tracheal chains and its possible mechanisms. Daru 2005;<br />

13: 28–33<br />

10 Chiang LC, Ng LT, Cheng PW, Chiang W, Lin CC. Antiviral activities of extracts<br />

and selected pure constituents of Ocimum basilicum. Clin Exp<br />

Pharmacol Physiol 2005; 32: 811–816<br />

11 Muralidharan A, Dhananjayan R. Cardiac stimulant activity of Ocimum<br />

basilicum Linn extracts. Indian J Pharmacol 2004; 36: 163–166<br />

12 Carvalho Filho JL, Blank AF, Alves PB, Ehlert PAB, Melo AS, Cavalcanti SCH,<br />

Arrigoni-Blank MF, Silva-Mann R. Influence of the harvesting time,<br />

temperature and drying period on basil (Ocimum basilicum L.) essential<br />

oil. Rev Bras Farmacogn 2006; 16: 24–30<br />

13 Blank AF, Carvalho Filho JLS, Santos Neto AL, Alves PB, Arrigoni-Blank MF,<br />

Silva-Mann R, Mendonça MC. Caracterização morfológica e agronômica<br />

de acessos do manjericão e alfavaca. Hortic Bras 2004; 22: 113–116<br />

14 Koster R, Anderson M, DeBeer MJ. Acetic acid for analgesic screening.<br />

Fed Proc 1959; 18: 412–413<br />

15 Eddy NB, Leimbach D. Synthetic analgesics. II. Dithienylbutenyl and dithienylbutilamines.<br />

J Pharmacol Exp Ther 1953; 107: 385–393<br />

16 Dubuisson D, Dennis SG. The formalin test: a quantitative study of the<br />

analgesic effects of morphine, merepidine and brain stem stimulation<br />

in rats and cats. Pain 1977; 4: 161–174<br />

17 Letizia CS, Cocchiara J, Lalko J, Api AM. Fragrance material review on linalool.<br />

Food Chem Toxicol 2003; 41: 943–964<br />

18 Le Bars D, Gozariu M, Cadden SW. Animal models of nociception. Pharmacol<br />

Rev 2001; 53: 597–652<br />

19 Marchioro M, Arrigoni-Blank MF, Mourão RHV, Antoniolli AR. Anti-nociceptive<br />

activity of the aqueous extract of Erythrina velutina leaves.<br />

Fitoterapia 2005; 76: 637–642<br />

20 Woolfe G, MacDonald AL. The evaluation of the analgesic action of pethidine<br />

hydrochloride (Demerol). J Pharmacol Exp Ther 1994; 80: 300–<br />

307<br />

21 Zakaria ZA, Sulainamn MR, Gopalan HK, Ghani ZDFA, Mohd RNSR, Mat<br />

Jais AM, Abdullah F. Antinociceptive and anti-inflammatory properties<br />

of Corchorus capsularis leaves chloroform extract in experimental animal<br />

model. Yakugaku Zasshi 2007; 127: 359–365<br />

22 Smith HS. Peripherally-acting opioids. Pain Physician 2008; 11: S121–<br />

S132<br />

23 Shibata M, Ohkubo T, Takahashi H, Inoki R. Modified formalin test: characteristic<br />

biphasic pain response. Pain 1989; 38: 347–352<br />

24 Narusuye K, Kawai F, Matsuzaki K, Miyachi E. Linalool suppresses voltage-gated<br />

currents in sensory neurons and cerebellar Purkinje cells.<br />

J Neural Transm 2005; 112: 193–203<br />

25 Sugawara Y, Hara C, Tamura K, Fuji T, Nakamura K, Masujima T, Aoki T.<br />

Sedative effect on humans of inhalation of essential oil of linalool: sensory<br />

evaluation and physiological measurements using optically active<br />

linalool. Anal Chim Acta 1998; 365: 293–299<br />

26 Elisabetsky E, Brum LF, Souza DO. Anticonvulsant properties of linalool<br />

in glutamate-related seizure models. Phytomedicine 1999; 6: 107–113<br />

27 Ghelardini C, Galeotti N, Salvatore G, Mozzanti G. Local anaesthetic activity<br />

of the essential oil of Lavandula angustifolia. <strong>Planta</strong> Med 1999;<br />

65: 700–703<br />

28 Peana AT, de Montis G, Sechi S, Sircana G, DʼAquila PS, Pippia P. Effects of<br />

(−)-linalool in the acute hyperalgesia induced by carrageenan, L-glutamate<br />

and prostaglandin E2. Eur J Pharmacol 2004; 497: 279–284<br />

29 Silva-Brum LF, Emanuelli T, Souza DO, Elisabetsky E. Effects of linalool<br />

on glutamate release and uptake in mouse cortical synaptosomes.<br />

Neurochem Res 2001; 26: 191–194<br />

30 Peana AT, DʼAquila PS, Chessa ML, Moretti MDL, Serra G, Pippia P.<br />

(−)-Linalool produces antinociception in two experimental models of<br />

pain. Eur J Pharmacol 2003; 460: 37–41<br />

Medeiros Venâncio A et al. Chemical Composition, Acute … <strong>Planta</strong> Med 2011; 77: 825–829<br />

829


830<br />

Original Papers<br />

Clove Oil Reverses Learning and Memory Deficits<br />

in Scopolamine-Treated Mice<br />

Authors Sumita Halder 1 , Ashish Krishan Mehta 2 , Rajarshi Kar 3 , Mohammad Mustafa 3 ,<br />

Pramod Kumari Mediratta 1 , Krishna Kishore Sharma 1<br />

Affiliations<br />

Key words<br />

l " Eugenia caryophyllata<br />

l " Myrtaceae<br />

l " passive avoidance<br />

l " elevated plus‑maze<br />

l " step‑down latency<br />

l " transfer latency<br />

received June 8, 2010<br />

revised Nov. 10, 2010<br />

accepted Nov. 15, 2010<br />

Bibliography<br />

DOI http://dx.doi.org/<br />

10.1055/s-0030-1250605<br />

Published online December 14,<br />

2010<br />

<strong>Planta</strong> Med 2011; 77: 830–834<br />

© Georg Thieme Verlag KG<br />

Stuttgart · New York ·<br />

ISSN 0032‑0943<br />

Correspondence<br />

Prof. Pramod<br />

Kumari Mediratta<br />

Department of Pharmacology<br />

University College<br />

of <strong>Medica</strong>l Sciences<br />

(University of Delhi)<br />

Dilshad Garden<br />

Delhi 110095<br />

India<br />

Phone: + 91 9810 5248 79<br />

Fax: +911122590495<br />

drpramod_k@yahoo.com<br />

1 Department of Pharmacology, University College of <strong>Medica</strong>l Sciences, Delhi, India<br />

2 Department of Physiology, University College of <strong>Medica</strong>l Sciences, Delhi, India<br />

3 Department of Biochemistry, University College of <strong>Medica</strong>l Sciences, Delhi, India<br />

Abstract<br />

!<br />

The present study was performed to examine the<br />

effect of Eugenia caryophyllata (Myrtaceae) on<br />

learning and memory, and also evaluate whether<br />

it can modulate oxidative stress in mice. Passive<br />

avoidance step-down task and elevated plusmaze<br />

were used to assess learning and memory<br />

in scopolamine-treated mice. Oxidative stress parameters<br />

were also assessed in brain samples by<br />

estimating the malondialdehyde (MDA) and reduced<br />

glutathione (GSH) levels at the end of the<br />

study. Scopolamine (0.3 mg/kg, i. p.) produced impairment<br />

of acquisition memory as evidenced by<br />

a decrease in step-down latency and an increase<br />

in transfer latency on day 1, and also impairment<br />

of retention of memory on day 2. Pretreatment<br />

Introduction<br />

!<br />

Halder S et al. Clove Oil Reverses… <strong>Planta</strong> Med 2011; 77: 830–834<br />

Eugenia caryophyllata (Myrtaceae; syn. Syzygium<br />

aromaticum, Caryophyllus aromaticus) commonly<br />

called “clove” is a plant indigenous to Moluca Island<br />

but cultivated in many other parts of the<br />

world including India [1]. In Iran, this plant and<br />

its essential oils have been used as an antiepileptic,<br />

and recent studies have established its anticonvulsant<br />

action [2, 3]. Traditional use of clove<br />

oil includes use in dental care as an antiseptic<br />

and analgesic, the oil being rubbed on the gums<br />

to treat toothaches. Clove oil is widely used in<br />

dentistry to treat dental caries and gingivitis due<br />

to its action against oral bacteria associated with<br />

dental caries and periodontal disease [4,5]. Previous<br />

research has shown antifungal, anticarcinogenic,<br />

antiallergic and antimutagenic activity of<br />

clove oil [6–9]. Its major component is considered<br />

to be eugenol and lesser amounts of beta-caryophyllene<br />

and eugenol acetate [10–12].<br />

Eugenol is reported to activate calcium channels<br />

in dorsal root ganglion cells [13], inhibit calcium<br />

with clove oil (0.05 mL/kg and 0.1 mL/kg) for 3<br />

weeks significantly reversed the increase in acquisition<br />

latency and all the doses (0.025, 0.05,<br />

0.1 mL/kg, i. p.) reversed the increase in retention<br />

latency induced by scopolamine (0.3 mg/kg, i. p.)<br />

in elevated plus-maze. However, 0.05 mL/kg clove<br />

oil attenuated memory deficits in the passive<br />

avoidance step-down task. Brain samples showed<br />

a significant decrease in MDA levels in the group<br />

treated with clove oil (0.05 and 0.025 mL/kg).<br />

GSH levels were also increased in clove oil-treated<br />

mice though the results were not significant.<br />

Thus, it can be concluded that clove oil can reverse<br />

the short-term and long-term memory deficits<br />

induced by scopolamine (0.3 mg/kg, i.p.) and<br />

this effect can, to some extent, be attributed to<br />

decreased oxidative stress.<br />

and potassium channels in guinea pig cardiac<br />

muscles [14] and block the transmission of action<br />

potential in the frog sciatic nerve [15]. Similarly,<br />

beta-caryophyllene, another component of clove<br />

oil has been observed to inhibit calcium and potassium<br />

currents in mammalian cardiac tissues<br />

and abolish conjunctival reflex [14–16]. The role<br />

of clove oil as an antiepileptic suggests its effect<br />

on the central nervous system [3]. It still remains<br />

to be explored whether clove oil has any effect on<br />

learning and memory since many drugs which<br />

are used in the treatment of epilepsy also modulate<br />

cognitive functions [17]. Increased oxidative<br />

stress in the brain has also been suggested to play<br />

a role in memory impairment, and the role of<br />

antioxidants in preventing oxidative stress-induced<br />

memory deficits is well established [18,<br />

19]. Eugenol has been known to demonstrate<br />

neuroprotective action in some studies and to<br />

possess antioxidant properties as well [20–23].<br />

Since eugenol constitutes the major component<br />

of clove oil, it can be possible that clove oil too ex-


hibits such antioxidant potential and modulates memory and<br />

learning processes.<br />

Various animal models have been used to evaluate memory and<br />

learning in animals. The passive avoidance apparatus and the elevated<br />

plus-maze are two such protocols wherein the animal is<br />

trained, and acquisition and retention of task memory are assessed.<br />

Elevated plus-maze (EPM) is a model of anxiety but it<br />

can also be used for the assessment of learning and memory<br />

[24]. In memory assessment, the animal is placed at the end of<br />

the open arm facing outwards (away from the centre), whereas<br />

for the assessment of anxiety, the animal is placed at the centre<br />

of the maze. In both these models, acquisition latency is indicative<br />

of short-term memory, and retention latency denotes longterm<br />

memory in animals [25]. Scopolamine is a known agent<br />

causing memory impairment, and piracetam is a proven nootropic<br />

agent known to improve this scopolamine-induced memory<br />

deficit [26–29].<br />

The present study was undertaken to examine the effect of clove<br />

bud oil on learning and memory and its effects on the parameters<br />

of oxidative stress in mice.<br />

Materials and Methods<br />

!<br />

Animals<br />

Young (6 weeks old) male Swiss albino mice weighing 20–25 g<br />

were used in the study. The animals were procured from the Central<br />

Animal House, University College of <strong>Medica</strong>l Sciences, Delhi.<br />

Animals were housed in groups of 4 per cage with free access to<br />

pellet diet and water in a temperature controlled facility. All the<br />

experiments were performed at daytime between 09:30 and<br />

15:30 h. Care of animals was as per the guidelines of the Committee<br />

for the Purpose of Control and Supervision of Experiments on<br />

Animals (CPCSEA), Ministry of Environment and Forest, Govt. of<br />

India, New Delhi. The study was duly approved by the Institutional<br />

Animal Ethics Committee, University College of <strong>Medica</strong>l Sciences,<br />

Delhi (Approval No. IEC‑AR/10/2008 dated September 26, 2008).<br />

Plant material<br />

Clove bud oil was obtained from M/s. Kancor Ingredients Limited.<br />

As per the literature provided by the manufacturer, the bud oil<br />

was obtained by the steam distillation from the dried flower<br />

buds of Eugenia caryophyllata to obtain a colourless pale yellow<br />

liquid of specific gravity 1.0426 at 25°C and refractive index<br />

1.533 at 20°C (sample code no. 085407, Reference no. 1 of 730/<br />

07-08). The characterisation of a sample of clove bud oil using<br />

GC‑MS revealed a high concentration of eugenol (87.34%), eugenyl<br />

acetate (5.18%), and beta-caryophyllene (2.01%).<br />

For the purpose of the study, the clove bud oil was suspended in<br />

double distilled water along with a few drops of Tween 80 to prepare<br />

solutions of the required doses (0.025, 0.05, 0.1 mL/kg).<br />

Drugs and dosing schedule<br />

Scopolamine (Sigma Aldrich) was dissolved in double distilled<br />

water and administered at a dose of 0.3 mg/kg. Piracetam (Nootropil<br />

– M/s UCB) of 98% purity was administered at 200 mg/kg as<br />

the standard drug.<br />

Animals were divided into six groups (n = 8). Group I was chronically<br />

administered 0.9% saline (i. p. once per day × 3 weeks), and<br />

at the end of 3 weeks, the animals were challenged with saline<br />

only. Group II was also administered 0.9% saline as above and at<br />

the end of 3 weeks was challenged with scopolamine (0.3 mg/kg,<br />

Original Papers<br />

i. p.). Groups III, IV, and V were chronically treated with clove oil<br />

0.025, 0.05, and 0.1 mL/kg/day, i.p., respectively × 3 weeks and at<br />

the end of 3 weeks were challenged with scopolamine (0.3 mg/<br />

kg, i.p.). Group VI was administered piracetam (200 mg/kg,<br />

i. p. × 3 weeks) and at the end of 3 weeks was challenged with<br />

scopolamine (0.3 mg/kg, i. p.). At the end of 3 weeks, the experiments<br />

were performed and the animals were sacrificed to dissect<br />

out the brain for oxidative stress studies.<br />

Assessment of memory and learning<br />

Step-down passive avoidance task: This test was performed by the<br />

method described by Joshi et al. [30] with modifications. The passive<br />

avoidance apparatus consisted of a box (27 × 27 × 27 cm)<br />

having three walls of wood and one wall of Plexiglas, featuring a<br />

grid floor (3 mm stainless steel rods set 8 mm apart) with a<br />

wooden platform (10 × 7 × 1.7 cm) kept at the centre of the grid<br />

floor. Each mouse was placed gently onto the wooden platform<br />

placed in the middle during the training period. When the mouse<br />

stepped down from the platform and placed all its paws on the<br />

grid floor, the unscrambled intermittent electric shock (1 Hz,<br />

0.5 s, 60 V DC) was delivered over a period of 15 s, and the stepdown<br />

latency (SDL) was recorded. SDL was defined as the time<br />

taken by the mouse to step down from the wooden platform to<br />

the grid floor with all its paws on the grid floor. Animals were<br />

then given a second training which was performed 90 min after<br />

the first one. Ninety minutes after the third training on the same<br />

day (day 1), the acquisition test was performed when no electric<br />

shock was given, and SDL was noted. The retention test was performed<br />

24 h after training (on day 2), when each mouse was<br />

again placed on the platform and the SDL was measured. An<br />

upper cut-off time of 600 s was set as per the method described<br />

by Reddy and Kulkarni [31].<br />

Transfer latency (TL) in elevated plus-maze: The test was performed<br />

by the method described by Ukai et al. [32]. On the first<br />

trial (training on day 1), a mouse was placed at the end of one of<br />

the open arms facing away from the central platform, and the<br />

time it took for the mouse to move from the open arm to either<br />

of the enclosed arms (TL) was recorded. If the mouse did not enter<br />

the enclosed arm within 90 s, it was pushed gently to guide its<br />

entrance in the enclosed arm, and in such case the transfer latency<br />

was assigned to 90 s. Then, the mouse was gently taken<br />

out of the plus-maze 10 s after it entered the enclosed arm and<br />

was returned to its home cage. Twenty-four hours later (on day<br />

2), the second trial (retention test) was performed. To accomplish<br />

this, the mouse was again put into the plus-maze, and TL was recorded<br />

up to a maximum of 90 s.<br />

Estimation of oxidative stress: At the end of the study period, i. e.,<br />

after obtaining results of cognitive tests, the animals were sacrificed<br />

by deep ether anaesthesia. The brains were quickly dissected<br />

out, washed with ice-cold sodium phosphate buffer,<br />

weighed and stored over ice. The brains were further processed<br />

within 30 min of dissection, and the estimation of oxidative<br />

stress parameters were conducted on the same working day.<br />

Brain tissue was homogenised with 10 times (w/v) sodium phosphate<br />

buffer (7.4 pH, ice cold, mixture of KH 2PO4 and Na2HPO4).<br />

The homogenate was centrifuged at 3000 rpm for 15 min. The estimated<br />

parameters of oxidative stress were malondialdehyde<br />

(MDA) and reduced glutathione (GSH).<br />

Malondialdehyde (MDA) estimation: Estimation of brain lipid peroxidation<br />

was done by measuring MDA levels, as described by<br />

Ohkawa et al [33]. To 0.5 mL of the supernatant of the homogenate,<br />

acetic acid (20%, pH 3.5) 1.5 mL, thiobarbituric acid (0.8%),<br />

Halder S et al. Clove Oil Reverses… <strong>Planta</strong> Med 2011; 77: 830–834<br />

831


832<br />

Original Papers<br />

and sodium lauryl sulphate (8.1%) 0.2 mL were added and the<br />

mixture heated at 100 °C for 1 h. After cooling this mixture, 5 mL<br />

of butanol : pyridine (15 : 1 v/v) and 1 mL of distilled water were<br />

added. The mixture was vortexed and then centrifuged at<br />

4000 rpm for 10 min. Thereafter, the organic layer was separated<br />

and the absorbance measured at 532 nm using a spectrophotometer.<br />

Comparing the absorbance with that of a standard of known<br />

concentration, the concentration of MDA was determined by a<br />

linear standard curve and expressed as nmol/g wet brain tissue.<br />

Reduced glutathione (GSH) estimation: GSH was estimated by the<br />

method described by Ellman [34]. To 0.5 mL of the supernatant of<br />

the homogenate obtained above, 1 mL tricarboxylic acid (5%) was<br />

added and centrifuged to remove the proteins. To 0.1 mL of this<br />

homogenate, 4 mL of phosphate buffer (pH 8.4), 0.5 mL of DTNB,<br />

and 0.4 mL double distilled water were added. This mixture was<br />

vortexed and absorbance read at 412 nm within 15 min. Various<br />

concentrations (5–50 µg) of standard glutathione were subjected<br />

to the steps mentioned above and the absorbance plotted against<br />

the known concentrations of GSH to give a standard curve. The<br />

concentration of GSH in the samples were determined by a linear<br />

standard graph and expressed as µg/g wet brain tissue.<br />

Statistical analysis<br />

Results of the above experiments were expressed as mean ± SEM,<br />

and the difference between means was analysed by analysis of<br />

variance (ANOVA) followed by Dunnettʼs multiple comparisons<br />

test.<br />

Results<br />

!<br />

Scopolamine (0.3 mg/kg, i. p.), when administered to the salinetreated<br />

group, was found to significantly increase the acquisition<br />

latency (TL1; p < 0.05) as well as retention latency (TL2; p < 0.01)<br />

compared to the group challenged with saline. Clove oil at doses<br />

of 0.05 mL/kg and 0.1 mL/kg significantly reversed the scopolamine-induced<br />

acquisition deficit (TL1; p < 0.01 and p < 0.05, respectively),<br />

and all the doses reversed retention deficit (TL2) induced<br />

by scopolamine (0.3 mg/kg, i. p.) in elevated plus-maze<br />

(p < 0.001). Piracetam (200 mg/kg, i. p.) pretreatment also reversed<br />

the scopolamine-induced deficit in acquisition (TL1;<br />

p < 0.05) and retention latencies (TL2; p < 0.001) significantly<br />

(l " Fig. 1).<br />

Scopolamine (0.3 mg/kg, i. p.), when administered to the salinetreated<br />

group, was found to significantly decrease (p < 0.05) the<br />

SDL on day 1 (acquisition) as well as on day 2 (retention). At a<br />

dose of 0.05 mL/kg, clove oil significantly attenuated the scopola-<br />

Halder S et al. Clove Oil Reverses… <strong>Planta</strong> Med 2011; 77: 830–834<br />

mine-induced decrease in SDL on day 1 (acquisition; p < 0.01) as<br />

well as on day 2 (retention; p < 0.05). Treatment with piracetam<br />

(200 mg/kg, i.p.) also reversed the scopolamine-induced acquisition<br />

and retention deficits significantly (p < 0.01) (l " Fig. 2).<br />

Chronic administration of clove oil (0.025 and 0.05 mg/kg, i. p.)<br />

significantly decreased MDA levels as compared to the salinetreated<br />

group (p < 0.01 and p < 0.01, respectively). Piracetam also<br />

showed a significant decrease in MDA levels compared to the saline-treated<br />

group (p < 0.01). GSH levels did not show a significant<br />

increase in any of the treated groups as compared to the saline-treated<br />

group (l " Fig. 3).<br />

Discussion<br />

!<br />

Clove (Eugenia caryophyllata) is an evergreen plant grown in<br />

tropical areas of the world including India. The active medicinal<br />

agent of this botanical is an essential oil which is obtained as a<br />

colourless or light yellowish fluid, extracted from dried flower<br />

buds by steam distillation [1]. Clove oil is mainly constituted of<br />

eugenol, beta-caryophyllene, eugenol acetate, as well as alphahumulen<br />

[35] and has been demonstrated to be effective against<br />

pentylenetetrazole-induced and maximal electroshock seizures,<br />

confirming the claims of its uses in epileptic disorders in Iranian<br />

folk medicine [3]. These findings indicate that clove oil, which has<br />

eugenol as the major constituent, may exert some action on the<br />

nervous system. It could be possible that it also exercises some<br />

effect on the central nervous system influencing learning and<br />

memory processes in the brain as well.<br />

Traditional medicine claims that clove oil is effective in relieving<br />

pain, improves memory, reduces depression and acts as a mind<br />

and body stimulant, thus affecting the central nervous system,<br />

but no conclusive study has been conducted so far to prove such<br />

effects of clove oil. The present study was aimed to explore the<br />

effect of clove oil on learning and memory and evaluate its role<br />

in modulating oxidative stress in mice.<br />

In the present study, we have demonstrated that in the stepdown<br />

latency test, clove bud oil produced significant improvement<br />

in passive avoidance acquisition and retention of memory<br />

in mice. Scopolamine (0.3 mg/kg, i.p.) produced deficits in learning<br />

as well as memory retention since the animals showed a decrease<br />

in step-down latency after prior training. Clove oil was<br />

chronically administered at three doses (0.025, 0.05, 0.1 mL/kg,<br />

i. p.) for three weeks. The treatment with a dose of 0.05 mL/kg<br />

significantly attenuated the scopolamine-induced deficits in acquisition<br />

and retention. However, its administration at doses<br />

higher and lower than 0.05 mL/kg failed to reverse the scopola-<br />

Fig. 1 Effect of chronic Eugenia caryophyllata (EC)<br />

and piracetam (PIR) administration for 3 weeks on<br />

transfer latency (TL) on elevated plus-maze on day 1<br />

and 2 in scopolamine (SAL + SCO)-treated mice.<br />

* P < 0.05 as compared to the saline (SAL + SAL)<br />

group (TL1), ** p < 0.01 as compared to the SAL +<br />

SCO group (TL1), € p < 0.05 as compared to the<br />

SAL + SAL group (TL1), €€ p < 0.05 as compared to<br />

the SAL + SCO group (TL1), a p < 0.01 as compared<br />

to the SAL + SAL group (TL2), b p < 0.001 as compared<br />

to the SAL + SCO group (TL2), c p < 0.05 as<br />

compared to the SAL + SCO group (TL2).


mine-induced memory impairment in mice in the passive avoidance<br />

task.<br />

Additional evidence for the attenuation of scopolamine-induced<br />

memory deficit of clove oil was obtained from studies on the elevated<br />

plus-maze test in which mice showed a natural aversion to<br />

open and high spaces and therefore spent more time in the enclosed<br />

arms. Itoh et al. [36] suggested that TL might be shortened<br />

if the animal had previously experienced entering the arms. This<br />

shortened TL could be related to memory. In the study presented<br />

in this report, shortened TL was obtained on day 2 (retention latency)<br />

as compared to day 1 (acquisition latency) in the salinetreated<br />

group, and the effect was statistically significant. TL on<br />

days 1 and 2 was significantly increased in scopolamine-treated<br />

(0.3 mg/kg) mice compared to the saline-treated group indicating<br />

thereby a memory impairment. Chronic treatment with clove oil<br />

for three weeks prior to scopolamine injection (0.3 mg/kg) significantly<br />

reversed the effects of scopolamine-induced alterations in<br />

the TLs. Chronic administration of clove oil at higher doses (0.05<br />

and 0.1 mL/kg) reversed the scopolamine-induced increase in TL<br />

on day 1 as well as on day 2. This observation suggests that clove<br />

oil at these doses improves acquisition as well as retention of the<br />

learned task. However, a lower dose (0.025 mL/kg) of the agent<br />

showed improvement in retention but not in acquisition latencies.<br />

Piracetam, a known nootropic drug, was shown to improve<br />

acquisition as well as retention in parallel experiments.<br />

Original Papers<br />

Fig. 2 Effect of chronic Eugenia caryophyllata (EC)<br />

and piracetam (PIR) administration for 3 weeks on<br />

step-down latency (SDL) of the passive avoidance<br />

task on day 1 and 2 in scopolamine (SAL + SCO)treated<br />

mice.* P < 0.05 when compared to the saline<br />

(SAL + SAL) group (SDL1), ** p < 0.01 when compared<br />

to the SAL + SCO group (SDL1), *** p < 0.01<br />

when compared to the SAL + SCO group (SDL2),<br />

$ p < 0.05 when compared to the SAL + SCO group<br />

(SDL2), a p < 0.05 when compared to the SAL + SAL<br />

group (SDL2).<br />

Fig. 3 Effect of chronic Eugenia caryophyllata (EC)<br />

and piracetam (PIR) administration for 3 weeks on<br />

malondialdehyde (MDA) and reduced glutathione<br />

(GSH) levels in brain tissue of mice.* P < 0.05 when<br />

compared to the SAL + SAL group, ** p < 0.01 when<br />

compared to the SAL + SAL group.<br />

Recent studies have shown that cognitive disorders characterised<br />

by insidious onset of memory impairment are very often related<br />

to increased oxidative stress [18, 19] which is reflected by an increase<br />

in lipid peroxidation and a decrease in reduced glutathione<br />

[33, 34]. Evaluation of the brain samples after the chronic<br />

treatment revealed the clove oil-treated samples to have lower<br />

MDA levels compared to the saline-treated group. Clove oil at<br />

doses of 0.025 and 0.05 mL/kg demonstrated lower MDA levels<br />

which were statistically significant when compared to the saline-treated<br />

group. Further, the medium dose of 0.05 mL/kg of<br />

clove oil showed the lowest MDA levels similar to the piracetamtreated<br />

group. Brain levels of GSH were found to be relatively increased<br />

in piracetam- and clove oil-treated groups, though this<br />

difference was not statistically significant.<br />

The reduction in MDA levels at a dose of 0.05 mL/kg clove oil correlates<br />

well with our findings of changes in passive avoidance and<br />

elevated plus-maze. However, a higher dose of 0.1 mL/kg of clove<br />

oil has exhibited improvement in elevated plus-maze only, without<br />

a significant reduction in MDA or an increase in GSH levels. A<br />

lower dose of 0.025 mL/kg showed significant improvement in<br />

memory retention in elevated plus-maze only along with a significant<br />

reduction of MDA levels but no significant improvement<br />

in SDL. Thus at a higher dose (0.1 mL/kg), a reversal in effect was<br />

observed. The constituents in clove oil apart from eugenol could<br />

be responsible for this effect. Though eugenol is a known antioxidant<br />

[20–23], other constituents like beta-caryophyllene, euge-<br />

Halder S et al. Clove Oil Reverses… <strong>Planta</strong> Med 2011; 77: 830–834<br />

833


834<br />

Original Papers<br />

nol acetate, and alpha-humulen could probably exert a modulating<br />

influence at a higher dose. This finding further points to the<br />

fact that plant products have the best therapeutic effect at an optimum<br />

dose and at doses higher or lower to it, the effect can be<br />

suboptimal. The GSH levels in all the groups were not statistically<br />

significant as compared to the saline-treated group, thus suggesting<br />

the involvement of other mechanisms that are associated<br />

with reducing oxidative stress and subsequent improvement in<br />

cognitive function in clove oil-treated mice.<br />

Lesser improvement of scopolamine-induced memory deficit<br />

produced by chronic administration of a higher dose of clove oil<br />

(0.1 mL/kg) could be due to a lesser improvement in oxidative<br />

stress as observed by an insignificant decrease in the brain MDA<br />

levels at this dose. Brain MDA levels in clove oil-treated mice<br />

were found to be increased relatively at a higher dose (0.1 mL/<br />

kg) compared to those at lower doses (0.025 and 0.05 mL/kg).<br />

Treatment with 0.2 mL/kg clove oil was found to increase the<br />

MDA levels even further significantly (data not shown) in subsequent<br />

experiments going on in our laboratory. Hence, the lesser<br />

improvement of scopolamine-induced memory deficit at a relatively<br />

higher dose of clove oil (0.1 mL/kg) as observed in the passive<br />

avoidance task could be probably due to better correlation of<br />

this test to oxidative stress as compared to elevated plus-maze.<br />

Thus, the present study strongly suggests that clove oil could reverse<br />

short-term and long-term memory deficits. Reduction in<br />

lipid peroxides could be one of the mechanisms of reducing oxidative<br />

stress in clove oil-treated mice leading to subsequent improvement<br />

in learning and memory. However, further studies are<br />

needed to figure out the role of other mechanisms as well.<br />

Acknowledgements<br />

!<br />

We would like to thank Kancor Ingredients Limited, Kerala, India,<br />

for generously providing us with clove bud oil for the study.<br />

References<br />

1 Tyler VE, Brady LR, Robbers JE. Pharmacognosy. Philadelphia: Lea and<br />

Febiger; 1998: 103–135<br />

2 Avicenna A. Ghanoon dar teb. Tehran: Soroosh; 1988: 244–251<br />

3 Pourgholami MH, Kamalinejad M, Javadi M, Majzoob S, Sayyah M. Evaluation<br />

of the anticonvulsant activity of the essential oil of Eugenia<br />

caryophyllata in male mice. J Ethnopharmacol 1999; 64: 167–171<br />

4 Cai L, Wu CD. Compounds from Syzygium aromaticum possessing<br />

growth inhibitory activity against oral pathogens. J Nat Prod 1996;<br />

59: 987–990<br />

5 Hiroak N, Ryui U, Nozomik KS, Kenji K. Role of endothelium and adventitia<br />

on eugenol induced relaxation of rabbit ear artery precontracted<br />

by histamine. J Smooth Muscle Res 1998; 34: 123–137<br />

6 Chami F, Chami N, Bennis S, Bouchikhi T, Remmal A. Oregano and clove<br />

essential oils induce surface alteration of Saccharomyces cerevisiae.<br />

Phytother Res 2005; 19: 405–408<br />

7 Kim HM, Lee EH, Hong SH, Song HJ, Shin MK, Kim SH, Shin TY. Effect of<br />

Syzygium aromaticum extract on immediate hypersensitivity in rats.<br />

J Ethnopharmacol 1998; 60: 125–131<br />

8 Miyazawa M, Hisama M. Suppression of chemical mutagen induced<br />

SOS response by alkylphenols from clove (Syzygium aromaticum) in<br />

Salmonella typhymurium TA1535/pSK1002 umu test. J Agric Food<br />

Chem 2001; 49: 4019–4025<br />

9 Zheng GQ, Kenney PM, Lam LKT. Sesquiterpenes from clove (Eugenia<br />

caryophyllata). J Nat Prod 1992; 55: 999–1003<br />

10 Chaieb K, Hajlaoui H, Zmantar T, Kahla-Nakbi AB, Rouabhia M, Mahdouani<br />

K, Bakhrouf A. The chemical composition and biological activity<br />

of clove essential oil, Eugenia caryophyllata (Syzigium aromaticum L.<br />

Myrtaceae): a short review. Phytother Res 2007; 21: 501–506<br />

Halder S et al. Clove Oil Reverses… <strong>Planta</strong> Med 2011; 77: 830–834<br />

11 Pawar VC, Thakur VS. In vitro efficacy of 75 essential oils against Aspergillus<br />

niger. Mycoses 2006; 49: 316–323<br />

12 Prashar A, Locke IC, Evans CS. Cytotoxicity of clove (Syzygium aromaticum)<br />

oil and its major components to human skin cells. Cell Prolif<br />

1999; 39: 241–248<br />

13 Okhubo T, Kitamura K. Eugenol activates Ca 2+ -permeable currents in rat<br />

dorsal root ganglion cells. J Dent Res 1997; 76: 1737–1744<br />

14 Sensch O, Vierling W, Brandt W, Reiter W. Effects of inhibition of calcium<br />

and potassium currents in guinea-pig cardiac contraction: comparison<br />

of beta-caryophyllene oxide, eugenol, and nifedipine. Br J Pharmacol<br />

2000; 131: 1089–1096<br />

15 Kozam G. The effect of eugenol on nerve transmission. Oral Surg 1977;<br />

44: 799–805<br />

16 Ghelardini C, Galeotti N, Mannelli LDC, Mazzanti G, Bartolini A. Local<br />

anaesthetic activity of beta-caryophyllene. Farmaco 2001; 56: 387–<br />

389<br />

17 Bharal N, Mehta AK, Agarwal NK, Mediratta PK, Sharma KK. Melatonin<br />

protects against maximal electroshock seizures induced cognitive impairment<br />

in mice. Pharm Rev 2009; 7: 132–135<br />

18 Gupta S, Garg GR, Bharal N, Mediratta PK, Banerjee BD, Sharma KK. Reversal<br />

of propoxur-induced impairment of step-down passive avoidance,<br />

transfer latency and oxidative stress by piracetam and ascorbic<br />

acid in rats. Environ Toxicol Pharmacol 2009; 28: 403–408<br />

19 Mehta KD, Garg GR, Mehta AK, Arora T, Sharma AK, Khanna N, Tripathi<br />

AK, Sharma KK. Reversal of propoxur-induced impairment of memory<br />

and oxidative stress by 4′-chlorodiazepam in rats. Naunyn Schmiedebergs<br />

Arch Pharmacol 2010; 381: 1–10<br />

20 Jirovetz L, Buchbauer G, Stoilova I, Stoyanova A, Krastanov A, Schmidt E.<br />

Chemical composition and antioxidant properties of clove leaf essential<br />

oil. J Agric Food Chem 2006; 54: 6303–6307<br />

21 Kim B, Kim J, Kim H, Heo M. Biological screening of 100 plants for cosmetic<br />

use. II. Antioxidative activity and free radical scavenging activity.<br />

Int J Cosmet Sci 1997; 19: 299–307<br />

22 Ogata M, Hoshi M, Urano S, Endo T. Antioxidant activity of eugenol and<br />

related monomeric and dimeric compounds. Chem Pharm Bull 2000;<br />

48: 1467–1469<br />

23 Wie MB, Won MH, Lee KH, Shin JH, Lee JC, Suh HW, Song DK, Kim YH.<br />

Eugenol protects neuronal cells from excitotoxic and oxidative injury<br />

in primary cortical cultures. Neurosci Lett 1997; 225: 93–96<br />

24 Sharma AC, Kulkarni SK. Evaluation of learning and memory mechanisms<br />

employing plus maze in rats and mice. Prog Neuropsychopharmacol<br />

Biol Psychiatry 1992; 16: 117–125<br />

25 Patil RA, Jagdale SC, Kasture SB. Antihyperglycemic, antistress and<br />

nootropic activity of roots of Rubia cordifolia Linn. Indian J Exp Biol<br />

2006; 44: 987–992<br />

26 Li H, Matsumoto K, Tohda M, Yamamoto M, Watanabe H. NMDA antagonists<br />

potentiate scopolamine-induced amnesic effect. Behav Brain Res<br />

1997; 83: 225–228<br />

27 Joshi H, Parle M. Nootropic activity of calyces of Hibiscus sabdariffa<br />

Linn. Iranian J Pharmacol Ther 2006; 5: 15–20<br />

28 Joshi H, Parle M. Evaluation of nootropic potential of Ocimum santum<br />

Linn in mice. Indian J Exp Biol 2006; 44: 133–136<br />

29 De Mello N, Carobrez AP. Elevated T maze as an animal model of memory:<br />

effects of scopolamine. Behav Pharmacol 2002; 13: 139–148<br />

30 Joshi H, Kaur N, Chauhan J. Evaluation of nootropic effect of Argyreia<br />

speciosa in mice. J Health Sci 2007; 53: 382–388<br />

31 Reddy DS, Kulkarni SK. The effects of neurosteroids on acquisition and<br />

retention of a modified passive-avoidance learning task in mice. Brain<br />

Res 1998; 791: 108–116<br />

32 Ukai M, Miura M, Kameyama T. Effects of galanin on passive avoidance<br />

response, elevated plus-maze learning, and spontaneous alternation<br />

performance in mice. Peptides 1995; 16: 1283–1286<br />

33 Okhawa H, Ohishi N, Yagi K. Assay of lipid peroxides in animal tissue by<br />

thiobarbituric acid reaction. Anal Biochem 1979; 95: 351–358<br />

34 Ellman GL. Tissue sulphydryl groups. Arch Biochem Biophys 1959; 82:<br />

70–77<br />

35 Ozturk A, Ozbek H. The anti-inflammatory activity of Eugenia caryophyllata<br />

essential oil: an animal model of anti-inflammatory activity.<br />

Eur J Gen Med 2005; 2: 159–163<br />

36 Itoh J, Nabeshima T, Kameyama T. Utility of elevated plus-maze for dissociation<br />

of amnesic and behavioural effects of drugs in mice. Eur J<br />

Pharmacol 1991; 194: 71–76


Goldenseal (Hydrastis canadensis L.) Extracts<br />

Synergistically Enhance the Antibacterial Activity<br />

of Berberine via Efflux Pump Inhibition<br />

Authors Keivan A. Ettefagh 1 , Johnna T. Burns 1 , Hiyas A. Junio 1 , Glenn W. Kaatz 2 , Nadja B. Cech 1<br />

Affiliations<br />

Key words<br />

l " Hydrastis canadensis<br />

l " Ranunculaceae<br />

l " synergy<br />

l " alkaloid<br />

l " berberine<br />

l " Staphylococcus aureus<br />

received Sept. 2, 2010<br />

revised Nov. 5, 2010<br />

accepted Nov. 17, 2010<br />

Bibliography<br />

DOI http://dx.doi.org/<br />

10.1055/s-0030-1250606<br />

Published online December 14,<br />

2010<br />

<strong>Planta</strong> Med 2011; 77: 835–840<br />

© Georg Thieme Verlag KG<br />

Stuttgart · New York ·<br />

ISSN 0032‑0943<br />

Correspondence<br />

Nadja B. Cech<br />

Department<br />

of Chemistry/Biochemistry<br />

The University<br />

of North Carolina Greensboro<br />

435 Sullivan Bldg<br />

Greensboro, NC 27402<br />

USA<br />

Phone: + 13 36334 30 17<br />

Fax: + 1336334 5402<br />

nadja_cech@uncg.edu<br />

1 Department of Chemistry/Biochemistry, The University of North Carolina Greensboro, Greensboro, NC, USA<br />

2 Division of Infectious Disease, John Dingell Department of Veteran Affairs <strong>Medica</strong>l Center<br />

and Department of Internal Medicine, Wayne State University School of Medicine, Detroit, MI, USA<br />

Abstract<br />

!<br />

Goldenseal (Hydrastis canadensis L.) is used to<br />

combat inflammation and infection. Its antibacterial<br />

activity in vitro has been attributed to its alkaloids,<br />

the most abundant of which is berberine.<br />

The goal of these studies was to compare the composition,<br />

antibacterial activity, and efflux pump<br />

inhibitory activity of ethanolic extracts prepared<br />

from roots and aerial portions of H. canadensis.<br />

Ethanolic extracts were prepared separately from<br />

roots and aerial portions of six H. canadensis<br />

plants. Extracts were analyzed for alkaloid concentration<br />

using LC‑MS and tested for antimicrobial<br />

activity against Staphylococcus aureus (NCTC<br />

8325-4) and for inhibition of ethidium bromide<br />

efflux. Synergistic antibacterial activity was observed<br />

between the aerial extract (FIC 0.375) and<br />

to a lesser extent the root extract (FIC 0.750) and<br />

berberine. The aerial extract inhibited ethidium<br />

Introduction<br />

!<br />

The rise in infections from multidrug resistant<br />

bacteria is recognized worldwide as a major<br />

health crisis. According to the European Antimicrobial<br />

Resistance Surveillance System, as many<br />

as 52 million of the world population may carry<br />

multidrug resistant bacteria. One such resistant<br />

strain, methicillin-resistant Staphylococcus aureus<br />

(MRSA), is estimated to be responsible for over<br />

18000 annual deaths [1] in the US alone. Better<br />

methods to treat infections from resistant bacteria<br />

are needed. There is a long history of the use<br />

of botanical medicines to treat inflammation and<br />

infection. It is often argued that the inherent complexity<br />

of such preparations, which may lead to<br />

synergistic interactions, may make them more effective<br />

than their pharmaceutical counterparts [2,<br />

3]. Furthermore, if such botanical medicines act<br />

through multiple different pathways, it may make<br />

Original Papers<br />

bromide efflux from wild-type S. aureus but had<br />

no effect on the expulsion of this compound from<br />

an isogenic derivative deleted for norA. Our studies<br />

indicate that the roots of H. canadensis contain<br />

higher levels of alkaloids than the aerial portions,<br />

but the aerial portions synergize with berberine<br />

more significantly than the roots. Furthermore,<br />

extracts from the aerial portions of H. canadensis<br />

contain efflux pump inhibitors, while efflux<br />

pump inhibitory activity was not observed for<br />

the root extract. The three most abundant H. canadensis<br />

alkaloids, berberine, hydrastine, and canadine,<br />

are not responsible for the efflux pump inhibitory<br />

activity of the extracts from H. canadensis<br />

aerial portions.<br />

Supporting information available online at<br />

http://www.thieme-connect.de/ejournals/toc/<br />

plantamedica<br />

the development of resistance more difficult. For<br />

these reasons, further research into the use of botanical<br />

medicines to combat bacterial infections is<br />

warranted.<br />

The botanical medicine goldenseal (Hydrastis<br />

canadensis L.) is the subject of this research. Goldenseal<br />

preparations are popular in the international<br />

market [4, 5] and are among the top 20 best<br />

selling botanical dietary supplements in the US<br />

[6]. Crude extracts and isolated compounds from<br />

goldenseal have demonstrated antibacterial activity<br />

in vitro and in clinical trials [7–11]. The<br />

antibacterial activity of goldenseal has typically<br />

been attributed to alkaloids, especially berberine<br />

[11, 12], which has shown activity against various<br />

gram-positive bacteria, including MRSA [13].<br />

However, there has been some suggestion that<br />

other compounds present in complex goldenseal<br />

preparations might enhance the antimicrobial activity<br />

of berberine [7, 14]. We have observed pro-<br />

Ettefagh KA et al. Goldenseal (Hydrastis canadensis … <strong>Planta</strong> Med 2011; 77: 835–840<br />

835


836<br />

Original Papers<br />

nounced antimicrobial activity of extracts from the aerial portions<br />

of goldenseal, which could not be attributed to berberine<br />

alone. We hypothesize that these extracts contain efflux pump<br />

inhibitors that synergistically enhance the antimicrobial activity<br />

of berberine. Bacterial efflux pumps are membrane bound proteins<br />

that pump toxins out of bacterial cells [15]. Overexpression<br />

of efflux pumps contributes to the development of resistance in<br />

bacteria, including S. aureus [16]. Inhibition of efflux pumps may<br />

enhance the effectiveness of antimicrobial agents that are substrates<br />

for these pumps and decrease the minimum inhibitory<br />

concentration (MIC) for the antimicrobials [17].<br />

The goals of these studies were (1) to compare alkaloid content in<br />

extracts prepared from below ground (roots and rhizomes) and<br />

above ground (leaves and stems) portions of H. canadensis, (2) to<br />

evaluate the antibacterial activity of H. canadensis extracts in<br />

combination with the antibacterial agent berberine, and (3) to<br />

determine whether H. canadensis extracts act as inhibitors of<br />

NorA, the major chromosomal S. aureus efflux pump. Ultimately,<br />

our objective was to show whether synergists other than the major<br />

known alkaloids are present in H. canadensis, and to determine<br />

which part of the plant would have higher levels of such<br />

synergists. These questions are important in the manufacture<br />

and quality control of dietary supplements from H. canadensis.<br />

More broadly, these studies are relevant because we demonstrate<br />

an analytical approach by which synergistic activity can be investigated<br />

prospectively in crude extracts of botanical medicines.<br />

Materials and Methods<br />

!<br />

Cell lines, chemicals, and biochemicals<br />

Staphyloccus aureus NCTC 8325-4 [18] and its isogenic norA deletion<br />

mutant K1758 [19] were used. Mueller Hinton broth, carbonyl<br />

cyanide m-chloro-phenylhydrazone (CCCP, purity > 98%<br />

by TLC), berberine (purity > 98% by HPLC), (1R,9S)-(-)-β-hydrastine<br />

(purity > 98% by HPLC), and DMSO were purchased from Sigma<br />

Aldrich and canadine (tetrahydroberberine, purity > 98% by<br />

HPLC, stereochemistry unconfirmed) was obtained from Sequoia<br />

Research. Acetic acid was purchased from Fisher Chemical. Ethanol<br />

(95%), HPLC grade acetonitrile, and HPLC grade methanol<br />

were obtained from Pharmaco-AAPER. Nanopure water was prepared<br />

with a nanodiamond water purification system from Barnstead.<br />

Plant material<br />

Hydrastis canadensis L. (Ranunculaceae) was cultivated in its native<br />

habitat (a hardwood forest in Hendersonville, NC, N 35°<br />

24.277′, W 082 °20.993′, 702.4 m elevation) and harvested in September<br />

of 2008. A voucher specimen was deposited at the Herbarium<br />

of the University of North Carolina at Chapel Hill<br />

(NCU583414) and identified by Dr. Alan S. Weakly. Individual H.<br />

canadensis plants were harvested and numbered (l " Table 1).<br />

Extraction and LC‑MS<br />

The extracts (50% ethanol: 50% nanopure water) were prepared<br />

using 1 g of powdered leaves and stems or roots and rhizomes<br />

per 5 mL of solvent (1 : 5 w: v) according to the standard procedures<br />

in the US dietary supplements industry [20]. Henceforth,<br />

the root and rhizome extract will be designated “root” and the<br />

leaf and stem extract will be designated “aerial”. Plant material<br />

was blended with solvent, macerated for 24 hr and filtered under<br />

vacuum. Extracts were stored in amber bottles at room temper-<br />

Ettefagh KA et al. Goldenseal (Hydrastis canadensis … <strong>Planta</strong> Med 2011; 77: 835–840<br />

Table 1 Quantity of alkaloids in extracts from the root and rhizome or leaf<br />

and stem (aerial) portions of 6 individual H. canadensis plants and 2 “pooled<br />

samples”. Concentrations were calculated from calibration curves for standards<br />

of each alkaloid analyzed with LC‑MS in the positive ion mode. The<br />

“pooled root” and “pooled leaf” extracts were prepared from larger batches<br />

of plant material (including multiple individual plants) and used for antimicrobial<br />

testing.<br />

Extract Berberine (mM) Hydrastine (mM) Canadine (mM)<br />

Root 1 29.9 ± 0.4 2.30 ± 0.04 0.069 ± 0.011<br />

Root 2 25.3 ± 0.5 2.08 ± 0.04 0.061 ± 0.010<br />

Root 3 39.6 ± 0.5 3.34 ± 0.06 0.078 ± 0.013<br />

Root 4 70.0 ± 0.9 5.23 ± 0.10 0.056 ± 0.009<br />

Root 5 25.0 ± 0.3 3.50 ± 0.07 0.074 ± 0.011<br />

Root 6 28.9 ± 0.4 2.42 ± 0.05 0.074 ± 0.012<br />

Average root 36.5 ± 0.5 3.15 ± 0.06 0.069 ± 0.011<br />

Aerial 1 8.5 ± 0.1 1.23 ± 0.02 0.042 ± 0.007<br />

Aerial 2 8.3 ± 0.1 1.58 ± 0.03 0.046 ± 0.007<br />

Aerial 3 8.8 ± 0.1 1.13 ± 0.02 0.029 ± 0.004<br />

Aerial 4 9.2 ± 0.1 0.77 ± 0.01 0.050 ± 0.008<br />

Aerial 5 6.2 ± 0.8 1.20 ± 0.02 0.059 ± 0.009<br />

Aerial 6 5.7 ± 0.1 0.83 ± 0.02 0.044 ± 0.007<br />

Average aerial 7.8 ± 0.1 1.12 ± 0.02 0.045 ± 0.007<br />

Pooled<br />

Sample root<br />

16.6 ± 0.3 3.79 ± 0.07 0.069 ± 0.011<br />

Pooled<br />

Sample aerial<br />

8.0 ± 0.1 2.61 ± 0.05 0.054 ± 0.008<br />

ature. Biological activity and HPLC profile were retained in these<br />

storage conditions for > 1 year. An entire below ground portion<br />

(roots and rhizome) and its corresponding aerial portions (leaves<br />

and stems) of six individual plants were extracted separately. The<br />

extracts were numbered so that in each case, the number for the<br />

root and aerial extract of a single plant matched. Two larger<br />

“pooled extracts” were also prepared, one using 100 g of root/rhizome<br />

material and one using 100 g of aerial portions. All bioassay<br />

data presented here were collected using these “pooled extracts”.<br />

Analysis of alkaloid content was accomplished with liquid chromatography–mass<br />

spectrometry (LC‑MS) using an ion trap mass<br />

spectrometer with electrospray source (LCQ Advantage; Thermo)<br />

coupled to reversed phase HPLC (HP1100; Agilent). A C-18 column<br />

(50 mm × 2.1 mm, 3 µm particle size, 110 Å pore size, Prevail<br />

packing; Grace and 0.5 µm precolumn filter; MacMod Analytical)<br />

was employed for the analysis of all extracts, using a 0.2 mL/min<br />

flow rate and a 10 µL injection volume. The samples were analyzed<br />

using the following gradient, where A = 1% acetic acid in<br />

nanopure water and B = HPLC grade acetonitrile: 100–68% A from<br />

0 to 5 min; 68% − 0% A from 5 to 20 min; 0% A from 20 to 25 min,<br />

100% B from 25–40 min. Mass spectrometric detection was<br />

conducted in the positive ion mode with a scan range of 50–<br />

2000 m/z. Capillary temperature was 275 °C, sheath gas pressure<br />

was 20 (arbitrary units), and spray, capillary, and tube lens voltages<br />

were 4.5 kV, 3 V, and 60 V, respectively.<br />

Standards of the alkaloids berberine, hydrastine, and canadine<br />

were prepared at stock concentrations of 1 mg/mL in methanol.<br />

Calibration curves over a concentration range of 0.05 to 100 µM<br />

were plotted as peak area of the relevant selected ion trace versus<br />

concentration. Extracts were diluted as necessary in 50 : 50 ethanol<br />

: water to provide alkaloid concentrations within the linear<br />

range of the calibration curve. Alkaloids were identified in the extracts<br />

by matching retention time and fragmentation patterns<br />

with the relevant standards.


Checkerboard assay with Staphyloccus aureus<br />

NCTC 8325-4<br />

Extracts were dried under nitrogen and redissolved in Mueller<br />

Hinton broth containing 10% DMSO and filtered through a<br />

0.45 µm PVDF filter. Broth microdilution MIC assays were performed<br />

according to Clinical and Laboratory Standards Institute<br />

(CLSI) guidelines [21]. Extracts were standardized to berberine<br />

and tested in combination with berberine with a checkerboard<br />

assay [22] over a concentration range of 5 to 300 µg/mL (where<br />

these represent concentration of berberine either in the standard<br />

or the extract). A purified standard of berberine, which has welldocumented<br />

antimicrobial activity [23–25], served as the positive<br />

control. The assay was performed in triplicate in 96-well<br />

plates with 1 × 10 5 CFU/mL S. aureus, 2% final DMSO content,<br />

and a final well volume of 250 µL. The negative (vehicle) control<br />

consisted of 2% DMSO in broth. MIC values were determined at<br />

the point at which there was no significant difference between<br />

OD600 of the vehicle control and the treatment. OD600 for wells<br />

containing only extracts or berberine (without bacteria) was subtracted<br />

from the relevant assay wells to minimize interference.<br />

FIC indices were calculated according to equation 1, where A<br />

and B are the compounds (or extracts) tested in combination, MI-<br />

CA is defined as the minimum inhibitory concentration of A<br />

alone, MICB is defined as the minimum inhibitory concentration<br />

of B alone, and [B] is defined as the MIC of B in the presence of a A,<br />

and [A] is defined as the MIC of A in the presence of B.<br />

FIC Index ¼ FICA þ FICB ¼ A ½ Š<br />

þ<br />

MICA B ½ Š<br />

MICB<br />

(equation 1)<br />

Efflux pump assay<br />

Wild type S. aureus and its isogenic norA deletion mutant (NCTC<br />

8325-4 and K1758, respectively) were grown in Mueller Hinton<br />

broth to OD 600 = 0.740. Ethidium bromide and CCCP were added<br />

to final concentrations of 25 µM and 100 µM, respectively, and<br />

the solution was incubated at 20°C with agitation (300 RPM) for<br />

20 min. This solution was diluted to OD 600 = 0.400 with fresh<br />

Mueller Hinton broth containing 10 µg/mL ethidium bromide<br />

and 100 µM CCCP. The solution was spun down in 1 mL aliquots<br />

at 20°C for 5 min at 13000 × g in a Spectrafuge 24D centrifuge<br />

(Labnet). The supernatant was discarded and the red pellets<br />

placed on ice. At time of measurement, these pellets were thawed<br />

for 5 min and resuspended with 1 mL fresh Mueller Hinton broth<br />

containing 2% DMSO with or without inhibitor. Fluorescence<br />

measurements were made using a Spex FluoroMax-2 spectrofluorometer<br />

(Instruments S. A., Inc.) every second for 300 s, with<br />

λ ex = 530, λ emiss = 600, and slit widths of 5 mm.<br />

Original Papers<br />

Fig. 1 Structures of major alkaloid constituents of<br />

Hydrastis canadensis, berberine (1), (1R,9S)-(-)-β-hydrastine<br />

(2), and canadine (tetrahydroberberine) (3).<br />

Statistics<br />

Statistical significance of means versus control was evaluated using<br />

single factor ANOVA (calculated with Microsoft Excel version<br />

2007) with p < 0.05 considered significant.<br />

Supporting information<br />

Chromatograms and CID fragmentation patterns for berberine,<br />

hydrastine, and canadine as well as data on inhibition of ethidium<br />

bromide efflux by the individual alkaloids are available as<br />

Supporting Information.<br />

Results<br />

!<br />

The alkaloids berberine, hydrastine, and canadine (l " Fig. 1) were<br />

detected in all extracts, as confirmed based on matching retention<br />

times, molecular weights, and CID fragmentation patterns<br />

with standard compounds (Fig. 1S, Supporting Information). The<br />

configuration of hydrastine indicated in l " Fig. 1 is that reported<br />

for the standard, but the configuration of the canadine standard<br />

was not confirmed. The isolation of a ± mixture of canadine from<br />

H. canadensis has been previously reported [26], but it was not<br />

possible with the LC‑MS method employed to distinguish stereoisomers<br />

of hydrastine or canadine.<br />

Alkaloids were found in higher concentrations in root than in leaf<br />

extracts (l " Table 1). This was true for each of the individual root<br />

and leaf samples, and average values among the 6 root and 6 aerial<br />

portion samples. For berberine, the root extracts contained an<br />

average of 5 times more berberine (36.45 ± 0.47 mM in roots versus<br />

7.77 ± 0.10 mM in aerial portions, p = 0.002), 3 times more hydrastine<br />

(3.145 ± 0.058 mM in roots versus 1.123 ± 0.021 mM in<br />

aerial portions, p = 0.002), and 1.5 times more canadine (0.069 ±<br />

0.011 mM in roots versus 0.045 ± 0.007 in aerial portions,<br />

p = 0.001) than did the aerial extracts. Consistent with previous<br />

reports [27], berberine was the most abundant of the alkaloids<br />

(an average of 6.12% in roots, 1.31% in aerial portions), followed<br />

by hydrastine (0.60% in roots, 0.22% in aerial portions) and, finally,<br />

canadine (0.11% in roots, 0.008% in aerial portions).<br />

As indicated by the FIC value of 0.375 (l " Table 2) and by the convex<br />

shape of the isobologram (l " Fig. 2), the extract prepared<br />

from a pooled sample of H. canadensis leaves synergistically enhanced<br />

the antimicrobial activity of berberine. A suggestion of<br />

synergistic enhancement by the root extract was observed as<br />

well (l " Fig. 2), but the results did not comply with the stringent<br />

guidelines of an FIC < 0.5 indicating synergy [28]. For validation<br />

purposes, berberine was tested in combination with itself<br />

(l " Fig. 2), and the effect was additive (FIC of 1.00, linear isobologram).<br />

The berberine standard also served as the positive control<br />

for the antimicrobial assays, and gave an MIC of 150 µg/mL, consistent<br />

with literature values [29–32].<br />

Ettefagh KA et al. Goldenseal (Hydrastis canadensis … <strong>Planta</strong> Med 2011; 77: 835–840<br />

837


838<br />

Original Papers<br />

Table 2 MIC and FIC indices of Hydrastis canadensis extracts or isolated alkaloids<br />

in combination with berberine. MIC indicates the minimum inhibitory<br />

concentration of the compound alone, while FIC (calculated with equation 1),<br />

indicates the degree of interaction between the given compound or extract<br />

and berberine.<br />

MIC (µg/mL) FIC Index<br />

Berberine 150 1.00<br />

Hydrastine > 300 0<br />

Canadine > 300 0<br />

Root extract 150 0.750<br />

Aerial extract 75 0.375<br />

Fig. 2 Isobolograms indicating inhibition of bacteria growth by H. canadensis<br />

extracts and added berberine. Berberine alone was also included in<br />

the assay for validation and to serve as a positive control. Extracts were<br />

dissolved in 2% DMSO in Mueller Hinton broth. Assays were performed in<br />

triplicate in a 96-well plate with 5 × 10 5 CFU/mL S. aureus.<br />

Fig. 3 Percent fluorescence over time for S. aureus (NCTC 8325-4) loaded<br />

with ethidium bromide and treated with various extracts and controls.<br />

Treatments included the known efflux pump inhibitor CCCP (positive control)<br />

and extracts from the roots and aerial portions of H. canadensis. All<br />

extracts and CCCP were dissolved in Mueller Hinton broth containing 2%<br />

DMSO. Data points represent the average of three separate experiments<br />

(using 3 different pellets of S. aureus). Error bars are ± standard error.<br />

Ettefagh KA et al. Goldenseal (Hydrastis canadensis … <strong>Planta</strong> Med 2011; 77: 835–840<br />

Fig. 4 Percent fluorescence over time for the norA-deleted S. aureus<br />

(K1758). Aside from the use of a different strain of bacteria, conditions<br />

were identical to those employed for l " Fig. 3.<br />

The extract prepared from pooled aerial portions of H. canadensis<br />

was also shown to inhibit efflux of ethidium bromide from wild<br />

type S. aureus (l " Fig. 3), but not its norA-deleted derivative<br />

(l " Fig. 4). For wild-type S. aureus, the percent of ethidum bromide<br />

fluorescence after 3 minutes compared to the initial reading<br />

for bacteria in broth alone was 66.7% ± 5.5% for the root extract<br />

(50 µg/mL), 79.2% ± 1.2% for the aerial extract (50 µg/mL), and<br />

84.73% ± 1.29% for the positive control (CCCP at 20 µg/mL). The<br />

results for the CCCP and the aerial extract were significantly different<br />

from that of the negative control (ethidium bromide<br />

loaded bacteria in Mueller Hinton broth with 2% DMSO), with p<br />

values of 0.002 and 0.02, respectively. There was no significant<br />

difference in % fluorescence between the root extract and the<br />

negative control after 3 minutes. For the norA-deleted S. aureus,<br />

no significant inhibition of ethidium bromide efflux was observed<br />

for any of the treatments (H. canadensis aerial extract, H.<br />

canadensis root extract, or CCCP) after 3 min (l " Fig. 4).<br />

Discussion<br />

!<br />

Goldenseal roots (rather than aerial portions) are more commonly<br />

employed medicinally because of their higher alkaloid<br />

content [27]. Our results do show higher alkaloid content in the<br />

root extracts, but we show that when the extracts are standardized<br />

to berberine content, extracts from the aerial portions of the<br />

plants synergize the antimicrobial activity of berberine (l " Fig. 2).<br />

This finding indicates that some constituent(s) other than berberine<br />

in the extract from H. canadensis aerial portions synergistically<br />

enhance the antimicrobial activity of berberine. This synergistic<br />

enhancement is not due to hydrastine or canadine in the<br />

leaf extracts. Purified standards of these compounds exhibited no<br />

significant antimicrobial activity (MIC values of > 300 µg/mL,<br />

l " Table 2), and did not enhance the antimicrobial activity of berberine<br />

(FIC = 0, l " Table 2).<br />

On the basis of literature reporting the presence of efflux pump<br />

inhibitors in berberine containing plants [33], we hypothesized<br />

that the synergists present in the extracts of H. canadensis aerial<br />

portions might act as inhibitors of the NorA efflux pump, the ma-


jor chromosomal efflux pump expressed by S. aureus. Indeed, our<br />

data (l " Figs. 3 and 4) show that extracts from H. canadensis aerial<br />

portions do contain NorA efflux pump inhibitors. Ethidium bromide<br />

is a known substrate of NorA and fluoresces strongly (due<br />

to intercalation with DNA) when inside bacterial cells [33]. With<br />

the ethidium bromide efflux assay, cells are loaded with ethidium<br />

bromide and fluorescence is monitored over time. A decrease in<br />

fluorescence indicates efflux of ethidium bromide from the cells,<br />

and in the presence of an efflux pump inhibitor, fluorescence<br />

should decrease more slowly. This effect is observed with CCCP,<br />

a known efflux pump inhibitor, and with the extract prepared<br />

from aerial portions of H. canadensis (l " Fig. 3). If inhibition of<br />

NorA is responsible for the altered behavior of wild type<br />

S. aureus in the presence of CCCP and H. canadensis extracts, no<br />

effect should be observed with a norA-deleted strain. This, indeed,<br />

was the case (l " Fig. 4). The efflux pump inhibitory activity<br />

observed was not due to the presence of berberine, hydrastine, or<br />

canadine, as evidenced by the fact that the root extract, which<br />

had higher alkaloid concentration, did not show activity in the<br />

assay (l " Fig. 3). To verify this, the alkaloids were each tested individually<br />

and showed no significant inhibition of ethidium bromide<br />

efflux (Fig. 2S, Supporting Information).<br />

Proponents of the use of botanical dietary supplements often<br />

argue that their complexity leads to enhanced efficacy due to<br />

synergistic interactions among multiple constituents. Here we<br />

demonstrate that extracts from the aerial portions of Hydrastis<br />

canadensis contain both the known antimicrobial agent berberine<br />

and other compounds (hitherto unidentified) that synergistically<br />

increase the antimicrobial activity of berberine. We also<br />

show that this enhancement of antimicrobial activity is likely to<br />

occur through inhibition of the NorA efflux pump. This finding is<br />

particularly interesting given that Hydrastis canadensis is listed<br />

as endangered by CITES in much of its native habitat. Our results<br />

show that the aerial portions of this plant, which can be harvested<br />

sustainably, are a good source of antibacterial compounds.<br />

Furthermore, we demonstrate that berberine is effective in vitro<br />

at lower dosages when used in the complex phytochemical matrix<br />

of the plant aerial portions than in the root portions. Follow-up<br />

studies to evaluate the toxicity of extracts prepared from<br />

roots versus aerial portions of H. canadensis and to test their antibacterial<br />

activity in vivo are warranted.<br />

Acknowledgements<br />

!<br />

This publication was made possible by Grant Number 1 R15<br />

AT005005-01 from the National Center for Complementary and<br />

Alternative Medicine (NCCAM), a component of the National Institutes<br />

of Health (NIH). We thank William Burch for providing<br />

goldenseal plant material for this study, and Dr. Robert Cannon,<br />

Dr. Joseph Falkinham, and Dr. Nicholas Oberlies for helpful advice.<br />

References<br />

1 Klevens RM, Morrison MA, Nadle J, Petit S, Gershman K, Ray S, Harrison<br />

LH, Lynfield R, Dumyati G, Townes JM, Craig AS, Zell ER, Fosheim GE,<br />

McDougal LK, Carey RB, Fridkin SK. Invasive methicillin-resistant Staphylococcus<br />

aureus infections in the United States. JAMA 2007; 298:<br />

1763-1771<br />

2 Spelman K, Duke JA, Bogenschutz-Godwin MJ. The synergy principle at<br />

work with plants, pathogens, insects, herbivores and humans. In:<br />

Cseke L, Kirakosyan A, Kaufman PB, editors. Natural products from<br />

plants, 2nd edition. Boca Raton: CRC Taylor and Francis; 2006: 475–<br />

501<br />

Original Papers<br />

3 Wagner H, Ulrich-Merzenich G. Synergy research: approaching a new<br />

generation of phytopharmaceuticals. Phytomedicine 2009; 16: 97–110<br />

4 Bannerman JE. Goldenseal in world trade: pressures and potentials.<br />

Herbalgram 1997; 2: 51–53<br />

5 Sinclair A, Catling PM. Cultivating the increasingly popular medicinal<br />

plant, goldenseal: Review and update. Am J Altern Agric 2001; 16:<br />

131–140<br />

6 Blumenthal M. Herb market levels after five years of boom. Herbalgram<br />

1999; 47: 64–65<br />

7 Hwang BY, Roberts SK, Chadwick L, Wu CD, Kinghorn DA. Antimicrobial<br />

constitutents from goldenseal (the rhizomes of Hydrastis canadensis)<br />

against selected oral pathogens. <strong>Planta</strong> Med 2003; 69: 623–627<br />

8 Khosla PK, Neeraj VI, Gupta SK, Satpathy G. Berberine, a potential drug<br />

for trachoma. Rev Int Trach Pathol Ocul Trop Subtrop Sante Publique<br />

1992; 69: 147–165<br />

9 Knight SE. Goldenseal (Hydrastis canadensis) versus penicillin: a comparison<br />

of effects on Staphylococcus aureus, Streptococcus pyogenes,<br />

and Pseudomonas aeruginosa. Bios 1999; 70: 3–10<br />

10 Mahady GB, Pendland SL, Stoia A, Chadwick L. In vitro suceptibility of<br />

Helicobacter pylori to isoquinoline alkaloids from Sanguinaria canadensis<br />

and Hydrastis canadensis. Phytother Res 2003; 17: 217–221<br />

11 Scazzocchino F, Cometa MF, Tomassini L, Palmery M. Antibacterial activity<br />

of Hydrastis canadensis extract and its major isolated alkaloids.<br />

<strong>Planta</strong> Med 2001; 67: 561–564<br />

12 Chadwick LR, Wu CD, Kinghorn AD. Isolation of alkaloids from goldenseal<br />

(Hydrastis canadensis rhizomes) using pH-zone refining countercurrent<br />

chromatography. J Liq Chromatogr Relat Technol 2001; 24:<br />

2445–2453<br />

13 Villinski JR, Dumas ER, Chai HB, Pezzuto JM, Angerhofer CK, Gafner S.<br />

Antibacterial activity and alkaloid content of Berberis thunbergii, Berberis<br />

vulgaris and Hydrastis canadensis. Pharm Biol 2003; 41: 551–557<br />

14 Gentry EJ, Jampani HB, Keshavarz-Shokri A, Morton MD, Velde DV, Telikepalli<br />

H, Mitscher LA, Shawar R, Humble D, Baker W. Antitubercular<br />

natural products: Berberine from the roots of commercial Hydrastis<br />

canadensis powder. Isolation of inactive 8-oxotetrahydrothalifendine,<br />

canadine, b-hydrastine, and two new quinic acid esters, hycandinic acid<br />

esters-1 and 2. J Nat Prod 1998; 61: 1187–1193<br />

15 Nikaido H. Multiple antibiotic resistance and efflux. Curr Opin Microbiol<br />

1998; 1: 516–523<br />

16 Webber MA, Piddock LJV. The importance of efflux pumps in bacterial<br />

antibiotic resistance. J Antimicrob Chemother 2003; 51: 9–11<br />

17 Lewis K, Ausubel FM. Prospects for plant-derived antibacterials. Nat<br />

Biotechnol 2006; 24: 1504–1507<br />

18 Novick R. Properties of cryptic high-frequency transducing phage of<br />

Staphylococcus aureus. Virology 1967; 33: 155–166<br />

19 Price CT, Kaatz GW, Gustafson JE. The multidrug efflux pump NorA is<br />

not required for salicylate induced reduction in drug accumulation by<br />

Staphylococcus aureus. Int J Antimicrob Agents 2002; 20: 206–213<br />

20 Cech RA. Making plant medicine. Williams: Horizon Herbs; 2000: 276<br />

21 No author. Methods for dilution antimicrobial susceptibility tests for<br />

bacteria that grow aerobically; approved standard, Vol. 26, 7th edition.<br />

Wayne: Clinical and Laboratory Standards Institute; 2006: M7–A7<br />

22 Eliopolous GM, Moellering RC. Antimicrobial combinations. In: Lorian V,<br />

editor. Antibiotics in laboratory medicine. Baltimore, MD: Williams<br />

and Wilkins; 1996: 330–396<br />

23 Amin AH, Subbaiah TV, Abbasi KM. Berberine sulfate: antimicrobial activity,<br />

bioassay, and mode of action. Can J Microbiol 1969; 15: 1067–<br />

1076<br />

24 Yu HH, Kim KJ, Cha JD, Kim HK, Lee YE, Choi NY, You YO. Antimicrobial<br />

activity of berberine alone and in combination with ampicillin or oxacillin<br />

against methicillin-resistant Staphylococcus aureus. J Med Food<br />

2005; 8: 454–461<br />

25 Scazzocchio F, Cometa MF, Tomassini L, Palmery M. Antibacterial activity<br />

of Hydrastis canadensis extract and its major isolated alkaloids. <strong>Planta</strong><br />

Med 2001; 67: 561–564<br />

26 Monkovic I, Spenser ID. Biosynthesis of berberine and berverastine. Can<br />

J Chem 1965; 43: 2017–2026<br />

27 Upton R. Goldenseal root Hydrastis canadensis: Standards of analysis,<br />

quality control, and therapeutics. Santa Cruz: American Herbal Pharmacopoeia;<br />

2001<br />

28 Odds FC. Synergy, antagonism, and what the chequerboard puts between<br />

them. J Antimicrob Chemother 2003; 52: 1<br />

Ettefagh KA et al. Goldenseal (Hydrastis canadensis … <strong>Planta</strong> Med 2011; 77: 835–840<br />

839


840<br />

Original Papers<br />

29 Stermitz FR, Tawara-Matsuda J, Lorenz P, Mueller P, Zenewicz LA, Lewis K.<br />

5′Methoxyhydnocarpin-D and phenophorbide A: Berberis species<br />

components that potentiate berberine growth inhibition of resistant<br />

Staphylococcus aureus. J Nat Prod 2000; 63: 1146–1149<br />

30 Wang D, Yu L, Xiang H, Fan J, He L, Guo N, Feng H, Deng X. Global transcriptional<br />

profiles of Staphylococcus aureus treated with berberine<br />

chloride. FEMS Microbiol Lett 2008; 279: 217–225<br />

31 Yu H-H, Kim K-J, Cha J-D, Kim H-K, Lee Y-E, Choi N-Y, You Y-O. Antimicrobial<br />

activity of berberine alone and in combination with ampicillin or<br />

oxacillin against methicillin-resistant Staphylococcus aureus. J Med<br />

Food 2005; 8: 454–461<br />

Ettefagh KA et al. Goldenseal (Hydrastis canadensis … <strong>Planta</strong> Med 2011; 77: 835–840<br />

32 Tegos G, Stermitz FR, Lomovskaya O, Lewis K. Multidrug pump inhibitors<br />

uncover remarkable activity of plant antimicrobials. Antimicrob<br />

Agents Chemother 2002; 46: 3133–3141<br />

33 Stermitz FR, Lorenz P, Tawara JN, Zenewicz LA, Lewis K. Synergy in a medicinal<br />

plant: antimicrobial action of berberine potentiated by 5′-methoxyhydnocarpin,<br />

a multidrug pump inhibitor. Proc Nat Acad Sci<br />

2000; 97: 1433–1437


Neuroprotective Diarylheptanoids<br />

from the Leaves and Twigs<br />

of Juglans sinensis against<br />

Glutamate-Induced Toxicity<br />

in HT22 Cells<br />

Heejung Yang, Sang Hyun Sung, Jinwoong Kim,<br />

Young Choong Kim<br />

College of Pharmacy and Research Institute of Pharmaceutical<br />

Science, Seoul National University, Seoul, Korea<br />

Abstract<br />

!<br />

A new diarylheptanoid, juglanin C (1), was isolated from the 80%<br />

methanolic extract of the leaves and twigs of Juglans sinensis with<br />

three known diarylheptanoids, juglanin A (2), juglanin B (3), and<br />

(5R)-5-hydroxy-7-(4-hydroxy-3-methoxyphenyl)-1(4-hydroxyphenyl)-3-heptanone<br />

(4), using bioactivity-guided fractionation<br />

and chromatographic techniques. Among the isolated diarylheptanoids,<br />

compounds 1 and 2 significantly showed neuroprotective<br />

activities against glutamate-induced toxicity in HT22 cells.<br />

These two diarylheptanoids significantly reduced the overproduction<br />

of cellular peroxide in glutamate-injured HT22 cells.<br />

Moreover, these two diarylheptanoids significantly maintained<br />

antioxidative defense systems, including glutathione, glutathione<br />

reductase, and glutathione peroxidase, under glutamate-induced<br />

oxidative stress in HT22 cells.<br />

Key words<br />

Juglans sinensis · Juglandaceae · diarylheptanoid · HT22 hippocampal<br />

cells · antioxidant activity · neuroprotective<br />

Supporting information available online at<br />

http://www.thieme-connect.de/ejournals/toc/plantamedica<br />

Reactive oxygen species are free radicals containing oxygen<br />

atoms which are highly reactive due to the presence of unpaired<br />

valence shell electrons that causes significant damage to cell<br />

structures and platelet functions, protein denaturation, and lipid<br />

peroxidation [1, 2]. It has been reported that natural antioxidants<br />

such as terpenoids, flavonoids, and diarylheptanoids distributed<br />

in many medicinal plants could suppress the oxidative stress implicated<br />

in many neurodegenerative diseases [3–5]. Thus, it has<br />

been attempted to seek antioxidants having neuroprotective activity<br />

from natural resources [6].<br />

Glutamate induces neurotoxicity impairing the glutamate/cystine<br />

antiporter, which results in the decrease at the cellular level<br />

of free cysteine and the depletion of glutathione in HT22 cells,<br />

mouse hippocampal neuronal cells, and subsequently causes cell<br />

death attributable to oxidative stress [7]. Therefore, the glutamate-injured<br />

HT22 cells have been widely used as an in vitro assay<br />

system to elucidate the action mechanism of neuroprotective<br />

compounds [8–11].<br />

As a part of our continuing research seeking neuroprotective<br />

compounds from natural resources, we recently found that the<br />

80% methanolic extract of the leaves and twigs of Juglans sinensis<br />

Dode. (Juglandaceae), Korean walnut tree, significantly protected<br />

HT22 cells from the glutamate insult. J. sinensis is a deciduous native<br />

tree to Korea and China and has been used for the treatment<br />

of airway inflammatory diseases, leucorrhea, scabies, and elephantiasis<br />

in Korean traditional medicine [12]. It was reported<br />

that the phenolic compounds from the walnut tree leaves exerted<br />

antioxidative activities [13]. The genus Juglans contains ellagic acid,<br />

gallic acid, and flavonoids that inhibit the oxidation of human<br />

plasma and low-density lipoproteins (LDL) in vitro, along with<br />

polyphenols, diarylheptanoids, and terpenoids [14–16]. To date,<br />

only a few reports on the biological activities of this tree are<br />

available. Moreover, to the best of our knowledge, there was no<br />

report on its neuroprotective activity. In the present study, we attempted<br />

to isolate neuroprotective compounds from the leaves<br />

and twigs of J. sinensis employing bioactivity–guided fractionation<br />

techniques. We describe the isolation and identification of a<br />

new diarylheptanoid (1), along with three known diarylheptanoids<br />

(2–4), from the EtOAc fraction and their neuroprotective<br />

activities in glutamate-injured HT22 cells. The three known diarylheptanoids<br />

were identified as juglanin A (2) [15], juglanin B<br />

(3) [15], and (5R)-5-hydroxy-7-(4-hydroxy-3-methoxyphenyl)-1<br />

(4-hydroxyphenyl)-3-heptanone (4) [17]. The structures of these<br />

isolated compounds were unequivocally determined by 1D and<br />

2D NMR experiments and MS analysis, as well as in comparison<br />

with the literature (l " Fig. 1).<br />

Fig. 1 Structures of compounds 1–4.<br />

Letters<br />

Yang H et al. Neuroprotective Diarylheptanoids from… <strong>Planta</strong> Med 2011; 77: 841–845<br />

841


842<br />

Letters<br />

The molecular formula of compound 1 was determined as<br />

C20H22O4 on the basis of 13 C NMR analysis and HREIMS (m/z<br />

326.1518, calcd. for 326.1514 [M + ]). The 1 H NMR (l " Table 1) and<br />

HSQC spectra displayed the presences of two aromatic rings, a<br />

methylenedioxy group, a hydroxyl group, and six methylene<br />

groups. The 13 C NMR and DEPT spectra (l " Table 1) indicated seven<br />

quaternary carbons, six methine carbons, and seven methylene<br />

carbons in consistence with 1 H NMR and HSQC spectra analyses.<br />

The aliphatic chains from C-7 to C-13 were determined by<br />

the correlations in the 1 H- 1 H COSY analysis (l " Fig. 2). The HMBC<br />

correlations of H-7 (δH 2.85 and 2.41) with C-5 (δC 107.6) and C-19<br />

(δC 126.3), H-13 (δH 2.72) with C-15 (δC 129.5) and C-18 (δC 134.2)<br />

indicated that the aliphatic chain was attached at two aromatic<br />

rings (l " Fig. 2). Taken together, the structure of compound 1<br />

was similar to that of juglanin B (3) previously reported from Juglans<br />

spp. [15], except for the correlations of two protons (δH 5.99<br />

and 5.97) with C-3 (δC 142.4) and C-4 (δC 147.6) in HMBC analysis.<br />

These spectral data indicated the substitution of a hydroxyl group<br />

and a methoxy group in the A-ring of compound 3 by a methylenedioxy<br />

group. The absolute configuration of a hydroxyl group<br />

at C-11 was determined to be 11R by comparison of CD spectra<br />

data of compound 3 with those in the literature [18, 19] and the<br />

application of modified Mosherʼs method (l " Fig. 2) [20]. Consequently,<br />

the structure of 1 was determined as (11R)-11,17-dihydroxy-3,4-methylenedioxy-[7,0]-metacyclophane,<br />

and the compound<br />

was named juglanin C.<br />

The isolated diarylheptanoids 1–4 were evaluated for their neuroprotective<br />

activities in vitro. Among these diarylheptanoids,<br />

compounds 1 and 2 exhibited significant neuroprotective activities<br />

at the concentrations ranging from 1 to 10 µM compared to<br />

trolox, the positive control (97.8 ± 1.8% at 10 µM) (l " Table 2; the<br />

MTT assay showed a similar trend to the LDH assay; data not<br />

shown). It was noteworthy that the neuroprotective activity of<br />

compound 1 was superior to that of compound 3 in spite of the<br />

structural similarity between compounds 1 and 3. The difference<br />

in the neuroprotective activity of the two compounds may be explained<br />

by the presence of a methylenedioxy group in compound<br />

1. We attempted to elucidate the underlying mechanism of the<br />

neuroprotective activities of compounds 1 and 2 (93.7 ± 0.7%<br />

and 91.1 ± 1.3% at 10 µM for LDH assay, respectively). Since glutamate-induced<br />

toxicity is mediated by oxidative stress [21], the effects<br />

of the two compounds on the antioxidative defense system<br />

of glutamate-injured HT22 cells were determined. The effects of<br />

compounds 1 and 2 on the cellular peroxide contents were measured<br />

using the specific fluorescent dye, 2, 7-DCF‑DA (l " Table 3).<br />

If HT22 cells were exposed to glutamate, the cellular peroxide<br />

content was rapidly increased within 3 h after glutamate insult<br />

[22]. The increased cellular peroxides induced by the glutamate<br />

insult were effectively decreased by the treatment with compounds<br />

1 or 2. Glutamate-induced oxidative stress has been also<br />

known to deplete intracellular GSH and reduce the activities of<br />

antioxidant enzymes [23, 24]. We further investigated the effects<br />

of compounds 1 and 2 on GSH content and the activities of antioxidant<br />

enzymes in HT22 cells. Under our experimental conditions,<br />

the exposure of HT22 cells to glutamate resulted in a significant<br />

reduction in the activities of superoxide dismutase (SOD),<br />

glutathione reductase (GR), and glutathione peroxidase (GSH‑Px)<br />

(l " Table 4). The reduced activities of GR and GSH‑Px were significantly<br />

restored by the treatment with compounds 1 or 2 in HT22<br />

cells. However, compounds 1 and 2 did not significantly increase<br />

the SOD activity reduced by glutamate. These two compounds also<br />

effectively prevented the glutamate-induced GSH depletion.<br />

Yang H et al. Neuroprotective Diarylheptanoids from… <strong>Planta</strong> Med 2011; 77: 841–845<br />

Table 1 1 Hand 13 C NMR spectroscopic data for compounds 1 and 3 in<br />

DMSO-d 6.<br />

1 3<br />

No. δH (J in Hz) b a, b δC δH (J in Hz) b a, b δC 1 123.7, s 125.5, s<br />

2 120.5, s 126.2, s<br />

3 142.4, s 140.7, s<br />

4 147.6, s 148.3, s<br />

5 6.65 (1H, s) 107.6, d 6.69 (1H, s) 111.7, d<br />

6 131.6, s 130.3, s<br />

7 2.85 (1H, m), 30.4, t 2.88 (1H, m), 30.0, t<br />

2.41 (1H, m)<br />

2.46 (1H, m)<br />

8 1.87 (1H, m), 26.4, t 1.95 (1H, m), 26.3, t<br />

1.76 (1H, m)<br />

1.81 (1H, m)<br />

9 1.49–1.31 22.4, t 1.65 (1H, m), 22.6, t<br />

(2H, m)<br />

1.44 (1H, m)<br />

10 1.66 (1H, m), 39.4, t 1.81 (1H, m), 39.5, t<br />

1.40 (1H, m)<br />

1.53 (1H, m)<br />

11 3.70 (1H, m) 66.7, d 4 (1H, t,<br />

J = 9.8)<br />

66.7, d<br />

12 2.08 (1H, m), 34.4, t 2.22 (1H, m), 34.4, t<br />

1.49 (1H, m)<br />

1.65 (1H, m)<br />

13 2.72 (2H, m) 26.5, t 2.82 (2H, m) 26.4, t<br />

14 129.4, s 129.2, s<br />

15 6.97 (1H, m) 129.5, d 7.01 (1H,<br />

d, J = 8.2)<br />

129.9, d<br />

16 6.76 (1H, d, 115.7, d 6.78 (1H, 115.9, d<br />

J = 8.0)<br />

d, J = 8.2)<br />

17 152.2, s 151.0, s<br />

18 6.98 (1H, s) 134.2, d 7.16 (1H, s) 133.8, d<br />

19 6.50 (1H, s) 126.3, d 6.73 (1H, s) 125.1, d<br />

OCH3 3.83 (1H, s) 56.0, q<br />

OCH2O 5.99 (1H, s),<br />

5.97 (1H, s)<br />

100.4, t<br />

a 13 Multiplicity of C NMR data was determined by DEPT experiments (s = quaternary,<br />

d = methine, t = methylene, q = methyl). b1H and 13C NMR data were measured at 400<br />

and 100 MHz, respectively<br />

Fig. 2 Key 2D NMR correlations and Δδ (δS-δR) values obtained from MTPA<br />

esters for compound 1.<br />

However, compounds 3 and 4, which did not show significant<br />

neuroprotective activities in LDH assay (57.4 ± 2.0% and 41.6 ±<br />

7.1% at 10 µM, respectively), did not reduce the increased cellular<br />

peroxide content caused by the glutamate insult. The restoration<br />

of reduced intracellular GSH content and antioxidant enzymes<br />

including SOD, GR, and GSH‑Px activities also did not accompany<br />

the treatment with compounds 3 and 4 (data not shown).<br />

Taken together, compounds 1 and 2 isolated from J. sinensis significantly<br />

attenuated glutamate-induced neurotoxicity in HT22


Table 2 Protective effects of compounds 1–4 isolated from J. sinensis on the<br />

glutamate-induced toxicity in HT22 cells.<br />

Groups ED50 (µM)<br />

1 2.29 ± 0.12<br />

2 3.92 ± 0.07<br />

3 7.39 ± 0.10<br />

4 15.61 ± 1.21<br />

Trolox 3.52 ± 0.11<br />

Trolox was used as a positive control. The values are expressed as means ± SD of triplicate<br />

experiments<br />

Table 4 Protective effects of compounds 1 and 2 on the activities of antioxidant<br />

enzymes and glutathione level in the glutamate-injured HT22 cells.<br />

Groups Conc. SOD<br />

(µM) (U/mg<br />

protein)<br />

Control 27.065 ±<br />

0.738<br />

Glutamate- 18.320 ±<br />

injured<br />

1.571<br />

1 10 19.140 ±<br />

1.343<br />

5 18.585 ±<br />

0.388<br />

1 16.686 ±<br />

1.280<br />

2 10 20.641 ±<br />

0.723<br />

5 22.840 ±<br />

1.410**<br />

1 22.940 ±<br />

0.421*<br />

GR<br />

(mU/mg<br />

protein)<br />

6.240 ±<br />

0.315<br />

2.323 ±<br />

0.201<br />

3.960 ±<br />

0.056*<br />

3.816 ±<br />

0.381*<br />

3.298 ±<br />

0.174*<br />

4.243 ±<br />

0.345**<br />

4.461 ±<br />

0.255**<br />

3.239 ±<br />

0.266*<br />

GSH‑Px<br />

(mU/mg<br />

protein)<br />

0.971 ±<br />

0.011<br />

0.466 ±<br />

0.006<br />

0.580 ±<br />

0.006***<br />

0.584 ±<br />

0.005***<br />

0.513 ±<br />

0.009**<br />

0.557 ±<br />

0.012**<br />

0.578 ±<br />

0.006***<br />

0.571 ±<br />

0.010**<br />

GSSG/<br />

total<br />

GSH ratio<br />

2.768 ±<br />

0.082<br />

1.401 ±<br />

0.013<br />

1.766 ±<br />

0.016***<br />

1.612 ±<br />

0.041**<br />

1.656 ±<br />

0.022***<br />

1.722 ±<br />

0.158*<br />

1.628 ±<br />

0.027**<br />

1.595 ±<br />

0.049**<br />

HT22 cells were pretreated with compounds 1–4, respectively, for 1 h before the exposure<br />

to 5 mM glutamate and then maintained for 8–12 h. Each value represents the<br />

mean ± SD of three experiments. Results differ significantly from the glutamate-treated;<br />

* p < 0.05, ** p < 0.01, *** p < 0.001<br />

cells through a mechanism that included the scavenging of reactive<br />

oxygen species. From these results, it could be postulated<br />

that diarylheptanoids of J. sinensis have potential as a natural<br />

antioxidant resource for the treatment of neurodegenerative disorders.<br />

Further studies are ongoing to unravel the specific cellular<br />

and molecular mechanisms of diarylheptanoids from J. sinensis.<br />

Materials and Methods<br />

!<br />

The leaves and twigs of J. sinensis were collected at the Medicinal<br />

Plant Garden, College of Pharmacy, Seoul National University,<br />

Goyang, Korea, in September 2008 and air-dried. J. sinensis was<br />

identified by professor Jong Hee Park, College of Pharmacy, Pusan<br />

National University. A voucher specimen (SNUPH-0329) has been<br />

stored in the Herbarium at the Medicinal Plant Garden, College of<br />

Pharmacy, Seoul National University.<br />

Dried leaves and twigs (20 kg) of J. sinensis were extracted with<br />

80% MeOH (15 L × 3) in an ultrasonic apparatus (3 h × 3). After removal<br />

of the solvent in vacuo, the 80% MeOH extract (2.2 kg) was<br />

suspended in H2O and successively partitioned into n-hexane<br />

fraction, CHCl3 fraction, EtOAc fraction, and n-BuOH fraction, respectively.<br />

The EtOAc fraction (200 g) which showed the most<br />

Table 3 Protective effects of compounds 1 and 2 on cellular peroxidation in<br />

glutamate-induced toxicity in HT22 cells.<br />

Groups Conc. DCF‑DA Fluorescence<br />

(µM) (Arbitrary unit)<br />

Control 176.1 ± 2.7<br />

Glutamate-injured 329.5 ± 27.0<br />

1 10 239.3 ± 11.9***<br />

5 242.9 ± 3.1***<br />

1 267.9 ± 4.5*<br />

2 10 193.1 ± 13.3***<br />

5 229.3 ± 19.9***<br />

1 310.0 ± 6.0<br />

Trolox 10 184.3 ± 9.4***<br />

5 202.0 ± 14.9***<br />

1 238.1 ± 15.6**<br />

Letters<br />

HT22 cells were pretreated with compounds 1–4, respectively, for 1 h before the exposure<br />

to 5 mM glutamate and then maintained for 8–12 h. The content of intracellular<br />

peroxide was determined using the fluorescent dye 2, 7-DCF‑DA. The values are<br />

expressed as means ± SD of triplicate experiments. Results differ significantly from the<br />

glutamate-treated; * p < 0.05, ** p < 0.01, *** p < 0.001<br />

significant neuroprotective activity (63.3 ± 4.8% at 50 µg/mL and<br />

41.1 ± 5.4% at 10 µg/mL) in glutamate-injured HT22 cells was subjected<br />

to repeated open column chromatographies with a gradient<br />

elution of solvents. The EtOAc fraction was subjected to silica<br />

gel column chromatography (2 kg, 20 × 60 cm) and eluted<br />

with a mixture of CHCl 3:MeOH increasing polarity (50 : 1, 30: 1,<br />

10:1, 5:1, 3 : 1, 0:1, v/v, 20 L) to give ten fractions (HoE1-HoE10).<br />

Compound 2 (25 mg) was obtained from HoE2 by ODS silica gel<br />

column chromatography (MeOH : H2O = 7:3, 9:1, v/v, 5 × 20 cm,<br />

4 L) followed by Sephadex LH-20 (MeOH, 2 × 60 cm, 500 mL).<br />

HoE3 was subjected to ODS MPLC (5 × 20 cm) and eluted with<br />

MeOH : H 2O (6 : 4, v/v, 4 L) to yield 3 subfractions (HoE3A‑HoE3C).<br />

HoE3B was refined by Sephadex LH-20 (2 × 60 cm) with MeOH<br />

(500 mL) to give compound 1 (40 mg). Compound 3 (20 mg) was<br />

isolated from HoE3C using Sephadex LH-20 (MeOH, 2 × 60 cm,<br />

300 mL), followed by ODS MPLC (5 × 20 cm) with MeOH : H2O<br />

(7 : 3, v/v, 1 L) as an eluent. The HoE10 fraction was fractionated<br />

into 6 subfractions (HoE10A‑HoE10F) by ODS silica gel column<br />

(MeOH : H2O 8 :2, v/v, 10 × 20 cm, 4 L), and compound 4 (25 mg)<br />

was refined from HoE10F by Sephadex LH-20 (MeOH, 2 × 60 cm)<br />

and eluted with MeOH (300 mL).<br />

Isolates<br />

Juglanin C (1): colorless needle; m.p.; [α] D 25 : − 12.2 (c 1.13, MeOH);<br />

UV (MeOH): λmax (log ε) 297.9 (3.74) nm; IR: νmax 3255 (OH),<br />

1638 (CH=CH), 1509, 1406, 1258, 1222, 1051, 934 cm −1 ;CD(c<br />

0.0005, MeOH): Δε (nm) = + 25.2 (213), ‑18.2 (236), ‑8.4 (255),<br />

‑1.5 (277), ‑6.7 (298); FABMS: m/z = 327 [M + H] + ,326[M] + ;<br />

HREIMS: m/z = 326.1514 [M] + (calcd. m/z = 326.1518); 1 Hand 13 C<br />

NMR spectra data (DMSO-d6): see l " Table 1.<br />

(S)-MTPA ester (1a): 1 H NMR (CDCl3, 500 MHz): 5.69 (1H, m, H-<br />

11α), 2.99 (1H, d, J = 17.0 Hz, H-7a), 2.80 (1H, d, J = 17.6 Hz, H-<br />

13a), 2.47 (1H, m, H-7b), 2.44 (1H, m, H-13b), 2.32 (1H, m, H-<br />

8a), 2.23 (1H, t, J = 13.3 Hz, H-12a), 2.02 (1H, m, H-8b), 1.84 (1H,<br />

m, H-10a), 1.72 (1H, t, J = 11.3 Hz, H-10b), 1.61 (1H, m, H-12b),<br />

1.60 (1H, m, H-9a), 1,50 (1H, m, H-9b).<br />

(R)-MTPA ester (1b): 1 H NMR (CDCl3, 500 MHz): 5.58 (1H, m, H-<br />

11α), 2.94 (2H, m, H-7a and H-13a), 2.73 (1H, t, J = 13.9 Hz, H-<br />

13b), 2.47 (1H, t, J = 12.6 Hz, H-7b), 2.25 (2H, m, H-8a and H-<br />

Yang H et al. Neuroprotective Diarylheptanoids from… <strong>Planta</strong> Med 2011; 77: 841–845<br />

843


844<br />

Letters<br />

12a), 2.07 (1H, m, H-8b), 1.82 (1H, m, H-10a), 1.77 (1H, m, H-<br />

12b), 1.67 (1H, t, J = 11.3 Hz, H-10b), 1.58 (1H, m, H-9a), 1,48<br />

(1H, m, H-9b).<br />

An immortalized mouse hippocampal cell line, HT22 cells, was a<br />

gift from Dr. Ki-Sun Kwon at the Korea Research Institute of Bioscience<br />

& Biotechnology. HT22 cells were seeded onto 12 or 24well<br />

plates at a density of 2 × 10 5 or 1 × 10 5 cells/mL, respectively,<br />

in DMEM (Dulbeccoʼs modified Eagleʼs medium) containing 10%<br />

heat-inactivated FBS with penicillin (100 IU/mL) and streptomycin<br />

(100 µg/mL) and incubated at 37 °C in a humidified atmosphere<br />

of 95% air and 5% CO2. After 12 to 24 h incubation, cells<br />

were pretreated with each of the four diarylheptanoids at different<br />

concentrations for 1 h and then exposed to 5 mM L-glutamate.<br />

The purity of each one of the diarylheptanoids was higher<br />

than 95.0%. After 8 h, the supernatants of the cultures were assessed<br />

for glutamate-induced toxicity using LDH (lactate dehydrogenase)<br />

assay [25], and the residues of the cultures were evaluated<br />

for the neuroprotective effects using MTT (3-[4,5-dimethylthiozol-2-yl]-2,5-diphenyltetrazolium<br />

bromide) assay [26].<br />

Trolox (Sigma, > 97% purity) was used as a positive control for<br />

the bioassays. For measuring the cellular peroxide contents, the<br />

specific fluorescent dye, 2,7-DCF‑DA, was used [27]. The activities<br />

of SOD, GR, and GSH‑px were measured according to the<br />

previously reported methods [28–30]. Total GSH content in the<br />

supernatant was determined spectrophotometrically using the<br />

enzymatic cycling method [31].<br />

Supporting information<br />

Original spectral data of 1, detailed descriptions of the preparation<br />

of (S)-MTPA and (R)-MTPA ester, and bioassay protocols are<br />

available as Supporting Information.<br />

Acknowledgements<br />

!<br />

This study was supported by a grant (2010K000832) from the<br />

Brain Research Center of the 21st Century Frontier Research Program<br />

funded by the Ministry of Science and Technology, Korea.<br />

We thank Dr. Ki-Sun Kwon (the Korea Research Institute of Bioscience<br />

& Biotechnology, Daejon, South Korea) for his generous<br />

gift of HT22 cells.<br />

References<br />

1 de Diego-Otero Y, Romero-Zerbo Y, el Bekay R, Decara J, Sanchez L, Rodriguez-de<br />

Fonseca F, del Arco-Herrera I. Alpha-tocopherol protects<br />

against oxidative stress in the fragile X knockout mouse: an experimental<br />

therapeutic approach for the Fmr1 deficiency. Neuropsychopharmacology<br />

2009; 34: 1011–1026<br />

2 Kinsella JE, Frankel E, German B, Kanner J. Possible mechanisms for the<br />

protective role of antioxidants in wine and plant foods. Food Technol<br />

1993; 47: 85–89<br />

3 Dinda B, Chowdhury DR, Mohanta BC. Naturally occurring iridoids,<br />

secoiridoids and their bioactivity. An updated review, part 3. Chem<br />

Pharm Bull 2009; 57: 765–796<br />

4 Spencer JP, Vauzour D, Rendeiro C. Flavonoids and cognition: the molecular<br />

mechanisms underlying their behavioural effects. Arch Biochem<br />

Biophys 2009; 492: 1–9<br />

5 Ramassamy C. Emerging role of polyphenolic compounds in the treatment<br />

of neurodegenerative diseases: a review of their intracellular targets.<br />

Eur J Pharmacol 2006; 545: 51–64<br />

6 Zhao B. Natural antioxidants protect neurons in Alzheimerʼs disease<br />

and Parkinsonʼs disease. Neurochem Res 2009; 34: 630–638<br />

Yang H et al. Neuroprotective Diarylheptanoids from… <strong>Planta</strong> Med 2011; 77: 841–845<br />

7 Fukui M, Song JH, Choi J, Choi HJ, Zhu BT. Mechanism of glutamate-induced<br />

neurotoxicity in HT22 mouse hippocampal cells. Eur<br />

J Pharmacol 2009; 617: 1–11<br />

8 Kang TH, Baek HY, Kim YC. Protective effect of jakyak-gamcho-tang extract<br />

and its constituents against t-BHP-induced oxidative damage in<br />

HT22 cells. Am J Chin Med 2005; 33: 181–189<br />

9 Breyer A, Elstner M, Gillessen T, Weiser D, Elstner E. Glutamate-induced<br />

cell death in neuronal HT22 cells is attenuated by extracts from<br />

St. Johnʼs wort(Hypericum perforatum L.). Phytomedicine 2007; 14:<br />

250–255<br />

10 Satoh T, Izumi M. Neuroprotective effects of phenylenediamine derivatives<br />

independent of an antioxidant pathway in neuronal HT22 cells.<br />

Neurosci Lett 2007; 418: 102–105<br />

11 Jamarkattel-Pandit N, Pandit NR, Kim MY, Park SH, Kim KS, Choi H, Kim<br />

H, Bu Y. Neuroprotective effect of defatted sesame seeds extract against<br />

in vitro and in vivo ischemic neuronal damage. <strong>Planta</strong> Med 2010; 76:<br />

20–26<br />

12 Bae KH. Medicinal Plants of Korea. Seoul: Kyo-Hak Publishing Co.;<br />

2000: 49<br />

13 Pereira JA, Oliveira I, Sousa A, Valentao P, Andrade PB, Ferreira IC, Ferreres<br />

F, Bento A, Seabra R, Estevinho L. Walnut (Juglans regia L.) leaves:<br />

phenolic compounds, antibacterial activity and antioxidant potential<br />

of different cultivars. Food Chem Toxicol 2007; 45: 2287–2295<br />

14 Anderson KJ, Teuber SS, Gobeille A, Cremin P, Waterhouse AL, Steinberg<br />

FM. Walnut polyphenolics inhibit in vitro human plasma and LDL oxidation.<br />

J Nutr 2001; 131: 2837–2842<br />

15 Liu JX, Di DL, Wei XN, Han Y. Cytotoxic diarylheptanoids from the pericarps<br />

of walnuts (Juglans regia). <strong>Planta</strong> Med 2008; 74: 754–759<br />

16 Fukuda T, Ito H, Yoshida T. Antioxidative polyphenols from walnuts<br />

(Juglans regia L.). Phytochemistry 2003; 63: 795–801<br />

17 Shin D, Kinoshita K, Koyama K, Takahashi K. Antiemetic principles of<br />

Alpinia officinarum. J Nat Prod 2002; 65: 1315–1318<br />

18 Joshi BS, Pelletier SW, Newton MG, Lee D, McGaughey GB, Puar MS. Extensive<br />

1D, 2D NMR spectra of some [7.0]metacyclophanes and X‑ray<br />

analysis of (±)-myricanol. J Nat Prod 1996; 59: 759–764<br />

19 Takeda Y, Fujita T, Shingu T, Ogimi C. Studies on the bacterial gall of<br />

Myrica rubra – isolation of a new [7,0]-metacyclophan from the gall<br />

and Dl-beta-phenyllactic acid from the culture of gall-forming bacteria.<br />

Chem Pharm Bull 1987; 35: 2569–2573<br />

20 Hoye TR, Jeffrey CS, Shao F. Mosher ester analysis for the determination<br />

of absolute configuration of stereogenic (chiral) carbinol carbons. Nat<br />

Protoc 2007; 2: 2451–2458<br />

21 Fu Y, Koo MW. EGCG protects HT‑22 cells against glutamate-induced<br />

oxidative stress. Neurotox Res 2006; 10: 23–30<br />

22 Lafon-Cazal M, Pietri S, Culcasi M, Bockaert J. NMDA-dependent superoxide<br />

production and neurotoxicity. Nature 1993; 364: 535–537<br />

23 Almeida A, Heales SJ, Bolanos JP, Medina JM. Glutamate neurotoxicity is<br />

associated with nitric oxide-mediated mitochondrial dysfunction and<br />

glutathione depletion. Brain Res 1998; 790: 209–216<br />

24 Coyle JT, Puttfarcken P. Oxidative stress, glutamate, and neurodegenerative<br />

disorders. Science 1993; 262: 689–695<br />

25 Ma CJ, Kim SR, Kim J, Kim YC. Meso-dihydroguaiaretic acid and licarin A<br />

of Machilus thunbergii protect against glutamate-induced toxicity in<br />

primary cultures of a rat cortical cells. Br J Pharmacol 2005; 146: 752–<br />

759<br />

26 Lee KY, Kim SH, Jeong EJ, Park JH, Kim YC, Sung SH. New secoisolariciresinol<br />

derivatives from Lindera obtusiloba stems and their neuroprotective<br />

activities. <strong>Planta</strong> Med 2010; 76: 294–297<br />

27 Goodman Y, Mattson MP. Secreted forms of beta-amyloid precursor<br />

protein protect hippocampal neurons against amyloid beta-peptideinduced<br />

oxidative injury. Exp Neurol 1994; 128: 1–12<br />

28 McCord JM, Fridovich I. Superoxide dismutase. An enzymic function for<br />

erythrocuprein (hemocuprein). J Biol Chem 1969; 244: 6049–6055<br />

29 Carlberg I, Mannervik B. Purification and characterization of the flavoenzyme<br />

glutathione reductase from rat liver. J Biol Chem 1975; 250:<br />

5475–5480<br />

30 Flohe L, Gunzler WA. Assays of glutathione peroxidase. Methods Enzymol<br />

1984; 105: 114–121<br />

31 Tietze F. Enzymic method for quantitative determination of nanogram<br />

amounts of total and oxidized glutathione: applications to mammalian<br />

blood and other tissues. Anal Biochem 1969; 27: 502–522


eceived October 6, 2010<br />

revised November 16, 2010<br />

accepted November 18, 2010<br />

Bibliography<br />

DOI http://dx.doi.org/10.1055/s-0030-1250609<br />

Published online December 14, 2010<br />

<strong>Planta</strong> Med 2011; 77: 841–845<br />

© Georg Thieme Verlag KG Stuttgart · New York ·<br />

ISSN 0032‑0943<br />

Correspondence<br />

Prof. PhD Young Choong Kim<br />

College of Pharmacy<br />

and Research Institute<br />

of Pharmaceutical Sciences<br />

Seoul National University<br />

San 56-1, Sillim-Dong, Gwanak-Gu<br />

Seoul 151-742<br />

Republic of Korea<br />

Phone: + 82 2880 7842<br />

Fax: +8228882933<br />

youngkim@snu.ac.kr<br />

Steroidal Saponins from<br />

the Rhizomes of Dioscorea bulbifera<br />

and Their Cytotoxic Activity<br />

Hai Liu 1,2 , Gui-Xin Chou 1,2 , Jun-Ming Wang1 , Li-Li Ji1, 2 ,<br />

1, 2<br />

Zheng-Tao Wang<br />

1 The MOE Key Laboratory for Standardization of Chinese Medicines,<br />

Institute of Chinese Materia <strong>Medica</strong>, Shanghai University<br />

of Traditional Chinese Medicine, Shanghai, P. R. China<br />

2 Shanghai R & D Centre for Standardization of Chinese Medicines,<br />

Shanghai, P. R. China<br />

Letters<br />

Abstract<br />

!<br />

Two new steroidal saponins, diosbulbisides D (1)andE(2), along<br />

with five known saponins (3–7) were isolated from the rhizomes<br />

of Dioscorea bulbifera L. Their structures were elucidated on the<br />

basis of spectral data. Compounds 6 and 7, two3-O-trisaccharides<br />

of diosgenin spirostanes, showed moderate cytotoxic activity<br />

against human hepatocellular carcinoma cells, with IC50 values<br />

of 3.89 µM and 7.47 µM on SMMC7721, and 10.87 µM and<br />

19.10 µM on Bel-7402 cell lines, respectively.<br />

Key words<br />

steroidal saponin · Dioscorea bulbifera L. · Dioscoreaceae · cytotoxicity<br />

Supporting information available online at<br />

http://www.thieme.connect.de/ejournals/toc/plantamedica<br />

The rhizome of Dioscorea bulbifera L. (Dioscoreaceae) has been<br />

commonly used as a medicinal herb in China and Bangladesh for<br />

treatment of goiter, leprosy, and cancers [1,2]. Clerodane diterpenoids<br />

[3–9] and steroidal saponins [10–12] from the Dioscorea<br />

species have been reported to show potent biological activities<br />

such as antitumor, antifungal, and anti-inflammatory activities<br />

[5, 10,13]. In our continuing investigation on the chemistry of<br />

this plant [9,10], two new steroidal saponins, 1 and 2, were isolated,<br />

along with five known saponins, diosgenin-3-O-[α-<br />

L-rhamnopyranosyl-(1 → 4)-β-D-glucopyranoside] (3) [14], pennogenin-3-O-[α-L-rhamnopyranosyl-(1<br />

→ 4)-β-D-glucopyranoside]<br />

(4) [15], diosgenin-3-O-[α-L-rhamnopyranosyl-(1 → 2)-β-<br />

D-glucopyranoside] (5) [14], diosgenin-3-O-[α-L-rhamnopyranosyl-(1<br />

→ 2)-[α-L-rhamnopyranosyl-(1 → 3)]-β-D-glucopyranoside]<br />

(6) [16], and diosgenin-3-O-[α-L-rhamnopyranosyl-(1 → 2)-<br />

[α-L-rhamnopyranosyl-(1 → 4)]-β-D-glucopyranoside] (7) [14]<br />

(l " Fig. 1). Their cytotoxic activities were evaluated using in vitro<br />

assay.<br />

Compound 1 was obtained as a white powder. Its molecular formula<br />

was deduced to be C51H80O22 from the positive-ion<br />

HRFTMS (m/z 1067.5035 [M + Na] + , calcd. 1067.5038). The 1 H<br />

and 13 CNMRspectra(l " Table 1)of1 showed the presence of four<br />

sugar residues, indicated by four anomeric proton and carbon<br />

signals at δH 4.99 (J = 7.6 Hz)/δC 100.1, 5.94 (s)/102.7, 5.82 (s)/<br />

104.0, and 4.91 (J = 7.6 Hz)/104.9. Acid hydrolysis of 1 yielded<br />

glucose and rhamnose by TLC and GC analyses. In addition, three<br />

methyl singlet signals at δ H 2.06, 1.13, and 1.03, one methyl doublet<br />

signal at δH 1.08 (J = 6.7 Hz), an oxymethylene [δH 4.05 and<br />

Liu H et al. Steroidal Saponins from… <strong>Planta</strong> Med 2011; 77: 845–848<br />

845


eceived October 6, 2010<br />

revised November 16, 2010<br />

accepted November 18, 2010<br />

Bibliography<br />

DOI http://dx.doi.org/10.1055/s-0030-1250609<br />

Published online December 14, 2010<br />

<strong>Planta</strong> Med 2011; 77: 841–845<br />

© Georg Thieme Verlag KG Stuttgart · New York ·<br />

ISSN 0032‑0943<br />

Correspondence<br />

Prof. PhD Young Choong Kim<br />

College of Pharmacy<br />

and Research Institute<br />

of Pharmaceutical Sciences<br />

Seoul National University<br />

San 56-1, Sillim-Dong, Gwanak-Gu<br />

Seoul 151-742<br />

Republic of Korea<br />

Phone: + 82 2880 7842<br />

Fax: +8228882933<br />

youngkim@snu.ac.kr<br />

Steroidal Saponins from<br />

the Rhizomes of Dioscorea bulbifera<br />

and Their Cytotoxic Activity<br />

Hai Liu 1,2 , Gui-Xin Chou 1,2 , Jun-Ming Wang1 , Li-Li Ji1, 2 ,<br />

1, 2<br />

Zheng-Tao Wang<br />

1 The MOE Key Laboratory for Standardization of Chinese Medicines,<br />

Institute of Chinese Materia <strong>Medica</strong>, Shanghai University<br />

of Traditional Chinese Medicine, Shanghai, P. R. China<br />

2 Shanghai R & D Centre for Standardization of Chinese Medicines,<br />

Shanghai, P. R. China<br />

Letters<br />

Abstract<br />

!<br />

Two new steroidal saponins, diosbulbisides D (1)andE(2), along<br />

with five known saponins (3–7) were isolated from the rhizomes<br />

of Dioscorea bulbifera L. Their structures were elucidated on the<br />

basis of spectral data. Compounds 6 and 7, two3-O-trisaccharides<br />

of diosgenin spirostanes, showed moderate cytotoxic activity<br />

against human hepatocellular carcinoma cells, with IC50 values<br />

of 3.89 µM and 7.47 µM on SMMC7721, and 10.87 µM and<br />

19.10 µM on Bel-7402 cell lines, respectively.<br />

Key words<br />

steroidal saponin · Dioscorea bulbifera L. · Dioscoreaceae · cytotoxicity<br />

Supporting information available online at<br />

http://www.thieme.connect.de/ejournals/toc/plantamedica<br />

The rhizome of Dioscorea bulbifera L. (Dioscoreaceae) has been<br />

commonly used as a medicinal herb in China and Bangladesh for<br />

treatment of goiter, leprosy, and cancers [1,2]. Clerodane diterpenoids<br />

[3–9] and steroidal saponins [10–12] from the Dioscorea<br />

species have been reported to show potent biological activities<br />

such as antitumor, antifungal, and anti-inflammatory activities<br />

[5, 10,13]. In our continuing investigation on the chemistry of<br />

this plant [9,10], two new steroidal saponins, 1 and 2, were isolated,<br />

along with five known saponins, diosgenin-3-O-[α-<br />

L-rhamnopyranosyl-(1 → 4)-β-D-glucopyranoside] (3) [14], pennogenin-3-O-[α-L-rhamnopyranosyl-(1<br />

→ 4)-β-D-glucopyranoside]<br />

(4) [15], diosgenin-3-O-[α-L-rhamnopyranosyl-(1 → 2)-β-<br />

D-glucopyranoside] (5) [14], diosgenin-3-O-[α-L-rhamnopyranosyl-(1<br />

→ 2)-[α-L-rhamnopyranosyl-(1 → 3)]-β-D-glucopyranoside]<br />

(6) [16], and diosgenin-3-O-[α-L-rhamnopyranosyl-(1 → 2)-<br />

[α-L-rhamnopyranosyl-(1 → 4)]-β-D-glucopyranoside] (7) [14]<br />

(l " Fig. 1). Their cytotoxic activities were evaluated using in vitro<br />

assay.<br />

Compound 1 was obtained as a white powder. Its molecular formula<br />

was deduced to be C51H80O22 from the positive-ion<br />

HRFTMS (m/z 1067.5035 [M + Na] + , calcd. 1067.5038). The 1 H<br />

and 13 CNMRspectra(l " Table 1)of1 showed the presence of four<br />

sugar residues, indicated by four anomeric proton and carbon<br />

signals at δH 4.99 (J = 7.6 Hz)/δC 100.1, 5.94 (s)/102.7, 5.82 (s)/<br />

104.0, and 4.91 (J = 7.6 Hz)/104.9. Acid hydrolysis of 1 yielded<br />

glucose and rhamnose by TLC and GC analyses. In addition, three<br />

methyl singlet signals at δ H 2.06, 1.13, and 1.03, one methyl doublet<br />

signal at δH 1.08 (J = 6.7 Hz), an oxymethylene [δH 4.05 and<br />

Liu H et al. Steroidal Saponins from… <strong>Planta</strong> Med 2011; 77: 845–848<br />

845


846<br />

Letters<br />

3.69/δC 75.2 (CH2)], two carbonyl groups (δC 210.6 and 205.8), a<br />

tetrasubstituted olefinic group (δC 145.8 and 142.7), and a trisubstituted<br />

olefinic group [δH 5.38/δC 141.0, 121.5] suggested<br />

that 1 had a C27 cholestane steroidal skeleton [10]. In HMBC experiments,<br />

the correlations from H2-15 to C-16 (δC 205.8), and<br />

from Me-21, H2-23, and H2-24 to C-22 (δC 210.6) indicated the<br />

locations of the two carbonyl groups (l " Fig. 2). The double bond<br />

between C-17 (δC 142.7) and C-20 (δC 145.8) was determined by<br />

the HMBC correlations from Me-18 to C-17 and from Me-21 to δC<br />

142.7 and 145.8 (l " Fig. 2). The correlation between H-3 (δH 3.99)<br />

and H-9 (δH 1.05) in the NOESY spectrum indicated H-3 in α-orientation.<br />

The 25 R configuration was deduced based on the coupling<br />

difference between H-26a and H-26b, which showed a Δa b<br />

(δHa–δHb) of 0.36 [17]. The double bond at C-17(20) of 1 in cisconfiguration<br />

(Z-configuration) was revealed by the NOESY correlation<br />

between Me-18 and Me-21. Thus, the aglycone of 1 was<br />

identified as (25R)-3,26-dihydroxycholesta-5,17-diene-16,22-dione<br />

[10, 18]. The glycosidic and the monosaccharide residues<br />

linkages were also deduced from HMBC analyses, in which C-3<br />

(δC 77.9) correlated with H-1′, C-2′ (δC 78.5) with H-1′′, C-3′ (δC<br />

87.5) with H-1′′′, and C-26 (δC 75.2) correlated with H-1′′′′. The<br />

β-configuration of D-glucopyranosyl was determined by 1H NMR coupling constants ( 3J1, 2 > 7 Hz) for anomeric protons,<br />

while L-rhamnopyranosyl had its α-configuration deduced by<br />

comparing the 13C NMR data for the C-3 and C-5 of rhamnose<br />

[19]. Thus, the structure of 1 was established as (25R)-26-[(β-Dglucopyranosyl)oxy]-3β-[(O-α-L-rhamnopyranosyl-(1<br />

→ 3)-[α-Lrhamnopyranosyl-(1<br />

→ 2)]-β-D-glucopyranosyl)oxy]-cholesta-<br />

5,17-diene-16,22-dione and named diosbulbiside D.<br />

Compound 2 was obtained as a white powder. Its molecular formula<br />

was deduced to be C45H72O18 from the HRFTMS data (m/z<br />

923.4640 [M + Na] + , calcd. 923.4616). The 1Hand13C NMR spectra<br />

of 2 (l " Table 1) exhibited a C27 furospirostenol skeleton with<br />

one glucosyl and two rhamnopyranosyls (l " Table 1). The aglycone<br />

part of 2 was similar to that of nuatigenin [14], with the exception<br />

of an extra hydroxyl in 2. The 17-OH substitution was de-<br />

Liu H et al. Steroidal Saponins from … <strong>Planta</strong> Med 2011; 77: 845–848<br />

Fig. 1 Structures of compounds 1–7.<br />

Fig. 2 Key HMBC correlations (arrow) of compound 1.<br />

duced from the HMBC correlation of the quaternary carbon at δC<br />

88.3 to the proton signals at δH 4.64 (H-16), 1.03 (H-18), and 1.29<br />

(H-21). The HMBC correlations of H-1′ (δH 4.97) with C-3 (δC<br />

76.6), H-1′′ (δH 5.94) with C-2′ (δC 76.9), and H-1′′′ (δH 5.83) with<br />

C-3′ (δC 85.9) indicated the glycosidic and sugar sequencing linkages.<br />

Accordingly, the structure of 2 was elucidated as (22S,25S)-<br />

17α,26-dihydroxy-22,25-epoxyfurost-5-ene-3β-yl-O-α-L-rhamnopyranosyl-(1<br />

→ 3)-[α-L-rhamnopyranosyl-(1 → 2)]-β-D-glucopyranoside<br />

and was assigned to the trivial name diosbulbisin E.<br />

The cytotoxic activities of compounds 1–7 were evaluated against<br />

proliferation of the human hepatocellular carcinoma Bel-7402<br />

and SMMC7721 cell lines, following an MTT assay [20]. Among<br />

the tested compounds, 6 and 7 exhibited moderate cytotoxic activities,<br />

with IC50 values of 10.87 µM (6) and 19.10 µM (7) against<br />

the Bel-7402 cell line while for the SMMC7721 cell line, the IC50


Table 1 1 H (500 MHz) and 13 C NMR (125 MHz) data of diosbulbisides D (1)<br />

and E (2) with J values (in Hz) in parentheses.<br />

No. Diosbulbiside D Diosbulbiside E<br />

δ H δ C δ H δ C<br />

1a<br />

1b<br />

1.78 m<br />

0.99 m<br />

37.3<br />

1.80 m<br />

1.01 m<br />

36.0<br />

2a<br />

2b<br />

2.20 m<br />

1.93 m<br />

30.1<br />

2.17 m<br />

1.92 m<br />

28.5<br />

3 3.99 m 77.9 3.86 m 76.6<br />

4a<br />

4b<br />

2.82 m<br />

2.82 m<br />

38.7<br />

2.79 m<br />

2.79 m<br />

37.2<br />

5 141.0 139.2<br />

6 5.38 d (5.3) 121.5 5.35 d (5.2) 120.4<br />

7a<br />

7b<br />

1.88 m<br />

1.57 m<br />

31.8<br />

1.97 m<br />

1.68 m<br />

31.0<br />

8 1.57 m 30.9 1.53 m 30.9<br />

9 1.05 m 50.0 0.99 m 48.7<br />

10 37.2 35.6<br />

11a<br />

11b<br />

1.59 m<br />

1.59 m<br />

21.0<br />

1.65 m<br />

1.63 m<br />

19.5<br />

12a<br />

12b<br />

2.23 m<br />

1.59 m<br />

36.2<br />

2.40 m<br />

1.67 m<br />

30.3<br />

13 43.5 43.8<br />

14 1.48 m 50.6 2.13 m 51.4<br />

15a<br />

15b<br />

2.28 m<br />

2.10 m<br />

38.1<br />

2.22 m<br />

1.70 m<br />

30.7<br />

16 205.8 4.64 m 88.5<br />

17 142.7 88.3<br />

18 1.03 s 16.9 1.03 s 15.5<br />

19 1.13 s 19.4 1.15 s 18.0<br />

20 145.8 2.61 m 40.0<br />

21 2.06 s 15.9 1.29 d (7.2) 8.6<br />

22 210.6 119.0<br />

23a<br />

23b<br />

2.88 m<br />

2.88 m<br />

38.9<br />

2.21 m<br />

2.21 m<br />

32.8<br />

24a<br />

24b<br />

2.21 m<br />

1.94 m<br />

28.1<br />

2.45 m<br />

2.20 m<br />

30.9<br />

25 2.09 m 33.5 84.6<br />

26a<br />

26b<br />

4.05 m<br />

3.69 dd (9.6, 6.1)<br />

75.2<br />

3.96 m<br />

3.96 m<br />

68.6<br />

27<br />

3-Glc<br />

1.08 d (6.7) 17.6 1.41 s 22.6<br />

1′ 4.99 d (7.6) 100.1 4.97 d (7.8) 98.4<br />

2′ 4.14 m 78.5 4.14 m 76.9<br />

3′ 4.23 m 87.5 4.24 m 85.9<br />

4′ 4.13 m 70.1 4.12 m 68.4<br />

5′ 3.89 m 78.2 3.96 m 76.3<br />

6′a<br />

6′b<br />

2′-Rha<br />

4.55 m<br />

4.43 m<br />

62.4<br />

4.50 m<br />

4.40 m<br />

60.8<br />

1′′ 5.94 s 102.7 5.94 s 101.2<br />

2′′ 4.83 m 72.5 4.83 m 71.0<br />

3′′ 4.61 m 72.9 4.57 m 71.3<br />

4′ 4.40 m 73.9 4.37 m 72.4<br />

5′′ 4.94 m 70.0 4.93 m 68.4<br />

6′′<br />

3′-Rha<br />

1.84 d (6.2) 18.8 1.83 d (6.2) 17.2<br />

1′′′ 5.82 s 104.0 5.83 s 102.4<br />

2′′′ 4.58 m 72.6 4.55 m 71.0<br />

3′′′ 4.92 m 72.7 4.94 m 71.2<br />

4′′′ 4.40 m 73.7 4.36 m 72.1<br />

5′′′ 4.84 m 70.7 4.85 m 69.1<br />

6′′′ 1.73 d (6.3) 18.5 1.53 d (6.1) 17.0<br />

Table 1 1 H (500 MHz) and 13 C NMR (125 MHz) data of diosbulbisides D (1)<br />

and E (2) with J values (in Hz) in parentheses. (continued)<br />

No. Diosbulbiside D Diosbulbiside E<br />

δH δC δH δC 26-Glc<br />

1′′′′ 4.91 d (7.6) 104.9<br />

2′′′′ 4.10 m 75.3<br />

3′′′′ 4.02 m 78.6<br />

4′′′′ 4.31 m 71.8<br />

5′′′′ 4.32 m 78.7<br />

6′′′′a<br />

6′′′′b<br />

4.63 m<br />

4.47 m<br />

63.0<br />

values were 3.89 (6) and 7.47 µM (7), respectively. Structurally,<br />

both 6 and 7 are diosgenin saponins bearing a trisaccharide moiety<br />

at C-3, differently from other tested saponins.<br />

Materials and Methods<br />

!<br />

Letters<br />

Optical rotations were measured on a KRÜSS P8000-T polarimeter.<br />

Melting points were taken on a BÜCHI Melting Point B-540<br />

(uncorrected). The 1D and 2D NMR spectra were recorded on a<br />

Bruker AV500 spectrometer in pyridine-d5. The HRFTMS were<br />

determined on an Ionspec 4.7 T HisRes MALDI-FTMS. Column<br />

chromatography (CC) was performed using silica gel (200–300<br />

and 300–400 mesh; Qingdao Marine Chemical Co.), D101 macroporous<br />

resin (Tianjin Pesticide Co.), ODS‑A (50 µm; YMC), and<br />

Sephadex LH-20 (GE Healthcare) as the stationary phases. Thinlayer<br />

chromatography was performed on HSGF254 (0.2 mm;<br />

Qingdao Marine Chemical Co.) and RP-18 F254 (0.25 mm: Merck)<br />

plates. GC analysis was recorded on a Thermo Finnigan Trace DSQ<br />

gas chromatograph equipped with a mass spectrometry detector.<br />

The rhizomes of D. bulbifera L. were collected in September 2006<br />

from Qingyang, Anhui Province, China, and authenticated by Professor<br />

Shou-Jin Liu (Anhui College of Traditional Chinese Medicine,<br />

Anhui Province, China). A voucher specimen (LH0609-1)<br />

was deposited in the herbarium of the Institute of Chinese Materia<br />

<strong>Medica</strong>, Shanghai University of Traditional Chinese Medicine.<br />

The aqueous ethanol extract of the dried and pulverized rhizomes<br />

of D. bulbifera (30 kg) was partitioned with petroleum<br />

ether and ethyl acetate successively, and the residual aqueous solution<br />

was applied to D101 macroporous resin column chromatography<br />

(CC) to yield a saponin-rich fraction, which was further<br />

separated by silica gel CC, Sephadex LH-20, and reversed phase<br />

silica gel (RP-18) columns to afford seven compounds, 1–7. (Detailed<br />

information is shown in Supporting Information.)<br />

Isolates<br />

Diosbulbiside D (25R)-26-[(β-D-glucopyranosyl)oxy]-3β-[(O-α-Lrhamnopyranosyl-(1<br />

→ 3)-[α-L-rhamnopyranosyl-(1 → 2)]-β-Dglucopyranosyl)oxy]-cholesta-5,17-diene-16,22-dione<br />

(1): white<br />

powder, m.p. 182–183 °C, [α] D 25 − 74.4° (c 0.048, MeOH), 1 H‑NMR<br />

and 13 C NMR data: see l " Table 1, HRFTMS m/z: 1067.5035 [M +<br />

Na] + (calcd. for C51H80O22Na, 1067.5038).<br />

Diosbulbiside E (22S,25S)-17α,26-dihydroxy-22,25-epoxyfurost-<br />

5-en-3β-yl-O-α-L-rhamnopyranosyl-(1 → 3)-[α-L-rhamnopyranosyl-(1<br />

→ 2)]-β-D-glucopyranoside (2): white powder, m. p.<br />

Liu H et al. Steroidal Saponins from… <strong>Planta</strong> Med 2011; 77: 845–848<br />

847


848<br />

Letters<br />

290–291 °C, [α] D 25 − 94.0° (c 0.051, MeOH), 1 H‑NMR and 13 C NMR<br />

data: see l " Table 1, HRFTMSm/z: 923.4640 [M + Na] + (calcd. for<br />

C45H72O18Na, 923.4616).<br />

Acid hydrolysis and sugar analysis<br />

The hydrolysis and GC‑MS analysis of the chiral derivatives of the<br />

sugars of compounds 1 and 2 (1.5 mg, each) were achieved as<br />

previous described [10].<br />

MTT colorimetric assay<br />

The cell growth inhibition was measured by a microculture tetrazolium<br />

(MTT) assay, using two human hepatocellular carcinoma<br />

cells (Bel-7402 and SMMC7721; Cell Bank, Type Culture Collection<br />

of the Chinese Academy of Sciences) [10, 20], and 10hydroxycamptothecine<br />

(HCP; Shanghai R & D Center for Standardization<br />

of Chinese Medicines) as a positive control.<br />

Supporting information<br />

Details on extraction and isolation and copies of the spectra for<br />

compounds 1 and 2 are available as Supporting Information.<br />

Acknowledgements<br />

!<br />

Financial support from the National Natural Science Foundation<br />

of China (NSFC) to Dr. Hai Liu (No. 30701082) and National Basic<br />

Research Program of China (2006CB504704) are acknowledged.<br />

We are also grateful to Prof. Shou-Jin Liu for collecting material<br />

to this study. The authors thank Miss Jie Li and Prof. Kan Ding for<br />

the cytotoxicity assay.<br />

References<br />

1 Gao HY, Kuroyanagi M, Wu L, Kawahara N, Yasuno T, Nakamura Y. Antitumor-promoting<br />

constituents from Dioscorea bulbifera L. in JB6<br />

mouse epidermal cells. Biol Pharm Bull 2002; 25: 1241–1243<br />

2 Murray RDH, Jorge ZD, Khan NH, Shahjahan M, Quaisuddin M. Diosbulbin<br />

D and 8-epidiosbulbin-E acetate, norclerodane diterpenoid<br />

from Dioscorea bulbifera tubers. Phytochemistry 1984; 23: 623–625<br />

3 Komori T. Glycosides from Dioscorea bulbifera. Toxicon 1997; 35:<br />

1531–1535<br />

4 Teponno RB, Tapondjou AL, Abou-Mansour E, Stoeckli-Evans H, Tane P,<br />

Barboni L. Bafoudiosbulbins F and G, further clerodane diterpenoids<br />

from Dioscorea bulbifera L. var sativa and revised structure of bafoudiosbulbin<br />

B. Phytochemistry 2008; 69: 2374–2379<br />

5 Teponno RB, Tapondjou AL, Gatsing D, Djoukeng JD, Abou-Mansour E,<br />

Tabacchi R, Tane P, Stoekli-Evans H, Lontsi D. Bafoudiosbulbins A, and<br />

B, two anti-salmonellal clerodane diterpenoids from Dioscorea bulbifera<br />

L. var sativa. Phytochemistry 2006; 67: 1957–1963<br />

6 Zheng SZ, Guo Z, Shen T, Zhen XD, Shen XW. Three new apianen lactones<br />

from Dioscorea bulbifera L. Indian J Chem Sect B 2003; 42: 946–949<br />

7 Teponno RB, Tapondjou AL, Ju-Jung H, Nam JH, Tane P, Park HJ. Three<br />

new clerodane diterpenoids from the bulbils of Dioscorea bulbifera L.<br />

var sativa. Helv Chim Acta 2007; 90: 1599–1605<br />

Liu H et al. Steroidal Saponins from … <strong>Planta</strong> Med 2011; 77: 845–848<br />

8 Wang G, Liu JS, Lin BB, Wang GK, Liu JK. Two new furanoid norditerpenes<br />

from Dioscorea bulbifera. Chem Pharm Bull 2009; 57: 625–627<br />

9 Liu H, Chou GX, Guo YL, Ji LL, Wang JM, Wang ZT. Norclerodane diterpenoids<br />

from the rhizomes of Dioscorea bulbifera. Phytochemistry 2010;<br />

71: 1174–1180<br />

10 Liu H, Chou GX, Wu T, Guo YL, Wang SC, Wang CH, Wang ZT. Steroidal<br />

sapogenins and glycosides from the rhizomes of Dioscorea bulbifera.<br />

J Nat Prod 2009; 72: 1964–1968<br />

11 Teponno RB, Tapondjou AL, Djoukeng JD, Abou-Mansour E, Tabacchi R,<br />

Tane P, Lontsi D, Park HJ. Isolation and NMR assignment of a pennogenin<br />

glycoside from Dioscorea bulbifera L. var sativa. Nat Prod Sci<br />

2006; 12: 62–66<br />

12 Li SS, Deng JZ, Zhao SX. Steroids from tuber of Dioscorea bulbifera L.<br />

J Plant Resour Environ 1999; 8: 61–62<br />

13 Sautour M, Mitaine-Offer AC, Lacaille-Dubois MA. The Dioscorea genus:<br />

a review of bioactive steroid saponins. J Nat Med 2007; 61: 91–101<br />

14 Agrawal PK, Jain DC, Gupta RK, Thakur RS. Carbon-13 NMR spectroscopy<br />

of steroidal sapogenins and steroidal saponins. Phytochemistry<br />

1985; 24: 2479–2496<br />

15 Mahato SB, Sahu NP, Ganguly AN. Steroidal saponins from Dioscorea floribunda:<br />

Structures of floribundasaponins A and B. Phytochemistry<br />

1981; 20: 1943–1946<br />

16 Han XW, Yu H, Liu XM, Bao X, Yu B, Li C, Hui YZ. Complete 1 Hand 13 C<br />

NMR assignments of diosgenyl saponins. Magn Reson Chem 1999; 37:<br />

140–144<br />

17 Agrawal PK. Assigning stereodiversity of the 27-Me group of furostanetype<br />

steroidal saponins via NMR chemical shifts. Steroids 2005; 70:<br />

715–724<br />

18 Mimaki Y, Watanabe K, Sakagami H, Sashida Y. Steroidal glycosides<br />

form the leaves of Cestrum nocturnum. J Nat Prod 2002; 65: 1863–1868<br />

19 Kasai R, Okihara M, Asakawa J, Mizutani K, Tanaka O. 13 C nmr study of<br />

α-anomeric and β-anomeric pairs of d-mannopyranosides and l-rhamnopyranosides.<br />

Tetrahedron 1979; 35: 1427–1432<br />

20 Carmichael J, Degraff WG, Gazdar AF, Minna JD, Mitchell JB. Evaluation of<br />

a tetrazolium based semiautomated colorimetric assay assessment of<br />

chemosensitivity testing. Cancer Res 1987; 47: 936–942<br />

received July 4, 2010<br />

revised November 21, 2010<br />

accepted November 24, 2010<br />

Bibliography<br />

DOI http://dx.doi.org/10.1055/s-0030-1250633<br />

Published online December 16, 2010<br />

<strong>Planta</strong> Med 2011; 77: 845–848<br />

© Georg Thieme Verlag KG Stuttgart · New York ·<br />

ISSN 0032‑0943<br />

Correspondence<br />

Prof. Dr. Zheng-Tao Wang<br />

The MOE Key Laboratory<br />

for Standardization of Chinese Medicines<br />

Institute of Traditional Chinese Medicine<br />

Shanghai University<br />

of Traditional Chinese Medicine<br />

1200 Cailun Road<br />

201210 Shanghai<br />

P. R. China<br />

Phone: + 86 2151 3225 07<br />

Fax: +862151322519<br />

wangzht@hotmail.com


Complete Structural Assignment<br />

of Serratol, a Cembrane-Type<br />

Diterpene from Boswellia serrata,<br />

and Evaluation of Its<br />

Antiprotozoal Activity<br />

Thomas J. Schmidt 1 , Marcel Kaiser2,3 , Reto Brun 2,3<br />

1 Institut für Pharmazeutische Biologie und Phytochemie (IPBP),<br />

Westfälische Wilhelms-Universität Münster, Münster, Germany<br />

2 Swiss Tropical and Public Health Institute, Basel, Switzerland<br />

3 University of Basel, Basel, Switzerland<br />

Abstract<br />

!<br />

From the dichloromethane extract obtained from the gum resin<br />

of Boswellia serrata Roxb. (Burseraceae), a well-known medicinal<br />

plant resin (“Indian Olibanum”), the cembrane-type diterpene<br />

serratol was isolated in high yield. Its structure, previously reported<br />

without clear specification of double-bond geometry and<br />

without specification of stereochemistry, was reanalysed by<br />

means of spectroscopic measurements and unambiguously assigned<br />

as S(−)-cembra-3E,7E,11E‑triene-1-ol. Full assignment of<br />

all NMR data is reported for the first time. The compound was<br />

found to be identical with a cembrenol previously isolated from<br />

B. carteri. Serratol was tested for in vitro activity against four protozoan<br />

human pathogens, namely, Trypanosoma brucei rhodesiense<br />

(East African Human Trypanosomiasis, sleeping sickness),<br />

T. cruzi (Chagasʼ disease), Leishmania donovani (Kala-Azar), and<br />

Plasmodium falciparum (Tropical Malaria). It was found active<br />

against T. brucei and P. falciparum. These activities were 10- to<br />

15-fold higher than its cytotoxicity against rat skeletal myoblasts.<br />

While some reports exist on potential anti-inflammatory activity<br />

of Boswellia diterpenes, this is the first report on antiprotozoal<br />

activity of such a compound.<br />

Key words<br />

Boswellia serrata Roxb. · Burseraceae · cembrane diterpene ·<br />

serratol · antiprotozoal activity<br />

Supporting information available online at<br />

http://www.thieme-connect.de/ejournals/toc/plantamedica<br />

Pardhy and Bhattacharyya, in 1978, first reported on the isolation<br />

and structure elucidation of serratol, a cembrane diterpene, from<br />

the gum resin of Boswellia serrata Roxb. (“Indian Olibanum”; Burseraceae)<br />

and assigned its structure as cembra-4,8,12-triene-1-ol<br />

[1]. Although not explicitly stating a double-bond geometry, they<br />

depicted the structure of serratol with two Z-configured double<br />

bonds. The absolute stereochemistry at C-1 was left unassigned<br />

[1]. Klein and Obermann [2], in the same year, reported on a<br />

new cembrenol of the same constitution which they isolated<br />

from the essential oil of Olibanum, the resin of Boswellia carteri<br />

Burdw. (Syn. B. sacra Flueck.). These authors assigned the double-bond<br />

geometry of their compound as all-E on grounds of the<br />

NMR data, and the configuration at C-1 as S.<br />

We have now reisolated serratol from B. serrata and found its<br />

NMR data (see l " Table 1) identical with those of sarcophytol M,<br />

Letters<br />

Table 1 13 Cand 1 H‑NMR data (125/500 MHz, CDCl 3) of serratol. All assignments<br />

were confirmed by HSQC and HMBC spectra.<br />

Position δC (ppm) mult. δH (ppm) a mult. J (Hz)<br />

1 76.94 s<br />

2 34.86 t 2.19 m<br />

2.15 m<br />

3 121.03 d 5.234 br dd (t) 6.5, 7.5<br />

4 136.64 s<br />

5 39.59 t 2.19 m (2H)<br />

6 24.93 t 2.277 dddd (ddt)<br />

2.09 m<br />

7 126.04 d 4.879 br dd (t) 5.5, 6.5<br />

8 133.37 s<br />

9 40.02 t 2.10 m<br />

1.92 m<br />

10 23.87 t 2.06 m (2H)<br />

11 123.28 d 4.986 br dd (t) 7.0, 7.0<br />

12 135.62 s<br />

13 33.59 t 1.91 m<br />

1.73 m<br />

14 35.05 t 1.618 m (2H)<br />

15 34.66 d 1.682 m<br />

16 16.92b q 0.924 d (3H) 7.0<br />

17 16.75b q 0.931 d (3H) 7.0<br />

18 15.17 q 1.536 s (3H)<br />

19 15.28 q 1.555 s (3H)<br />

20 16.33 q 1.537 s (3H)<br />

a 1H‑NMR shift values reported with two digits were taken from HSQC cross signals.<br />

b Assignment interchangeable<br />

a cembratrienol of the soft coral Sarcophyton glaucum Quoy &<br />

Gaimard [3]. Sarcophytol M possesses the same constitution and<br />

double-bond configuration as reported for the B. carteri cembrenol<br />

[2] but opposite configuration at C-1, i.e., it is the enantiomer<br />

of the former. Based on HSQC and HMBC spectra, all proton and<br />

carbon resonances of serratol were resolved and unambiguously<br />

assigned for the first time. The specific optical rotation of our isolate<br />

([α] D 20 − 97, CHCl3) was essentially identical with the published<br />

value for serratol (− 93 [1]) and showed the same sign as<br />

reported for the cembrenol from B. carteri (− 152 [2]), while values<br />

of + 57 (CHCl3) and + 160 (EtOH) were reported for sarcophytol<br />

M [3]. Serratol from B. serrata is thus identical with the cembrenol<br />

from B. carteri (l " Fig. 1).<br />

It is noteworthy that serratol is present in the oleoresin in quite<br />

high concentrations. Calculated from our preparative yield, the<br />

content of serratol in the resin would amount to approximately<br />

1.2%.<br />

A number of reports exist on pharmacological activity of cembrane<br />

diterpenes from the Boswellia species, such as an interesting<br />

anti-inflammatory potential of incensol [4, 5]. The anti-infective<br />

potential of such compounds has not been studied so far.<br />

In the course of our search for natural compounds with antiprotozoal<br />

activity (e.g. [6, 7]), serratol was therefore tested for activity<br />

against Trypanosoma brucei rhodesiense, T. cruzi, Leishmania<br />

donovani,andPlasmodium falciparum. It was found active against<br />

T. brucei rhodesiense and P. falciparum (IC 50 1.1 µg/mL = 3.8 µM<br />

and 0.72 µg/mL = 2.5 µM, respectively). Its activity was much<br />

weaker than the positive control drugs, melarsoprol and chloroquine<br />

(0.004 and 0.07 µg/mL against T. brucei and P. falciparum,<br />

respectively) but, interestingly enough, it displayed cytotoxicity<br />

against the L6 rat skeletal myoblast cell line only at a much higher<br />

concentration (IC50 11.4 µg/mL = 39 µM) and thus showed a cer-<br />

Schmidt TJ et al. Complete Structural Assignment… <strong>Planta</strong> Med 2011; 77: 849–850<br />

849


850<br />

Letters<br />

Fig. 1 Structure of<br />

S-(−)-cembra-3E,7E,11Etriene-1-ol<br />

(serratol).<br />

tain degree of selectivity. It was essentially inactive against T.<br />

cruzi and L. donovani (IC50 = 44 and 12 µM). In spite of its relatively<br />

moderate activity, serratol might be an interesting starting<br />

point for further studies on antiprotozoal effects of cembranoid<br />

diterpenes.<br />

Material and Methods<br />

!<br />

Oleoresin of B. serrata was obtained from Pharmasan GmbH,<br />

batch No. PS0208030. A voucher sample (number PB226) was retained<br />

at the documentation file of the Institut für Pharmazeutische<br />

Biologie und Phytochemie, Münster. The material was powdered<br />

and 998 g were extracted exhaustively (Soxhlet) with dichloromethane<br />

to yield 665 g of crude extract. 100 g of the extract<br />

were separated in three portions of 33 g each by VLC on<br />

300 g silica 60 with n-hexane/EtOAc 95: 5 (1500 mL) and EtOAc<br />

(500 mL). The hexane-rich lipophilic fractions were combined<br />

(14.8 g) and further separated by CC on silica 60 (column dimensions;<br />

126 × 8 cm; 1500 g silica) using n-hexane/EtOAc 98:2<br />

(5000 mL), 95: 5 (12 000 mL), 90: 10 (5000 mL), 50: 50 (1000<br />

mL), and 0: 100 (2000 mL). Serratol was eluted in pure form (TLC<br />

control) between 12 300 and 13600 mL. The total yield was<br />

1831 mg of TLC and GC pure serratol in the form of a colourless<br />

resin.<br />

GC/EIMS analyses were carried out with an Agilent 6890N/5973<br />

system; optical rotation was measured with a Perkin Elmer Polarimeter<br />

341; CD and UV spectra were obtained with a JASCO J-815<br />

spectropolarimeter, and NMR spectra were recorded on a Varian<br />

INOVA spectrometer.<br />

Antiprotozoal tests were performed according to well-established<br />

protocols as reported previously [6].<br />

S-(−)-cembra-3E,7E,11E-triene-1-ol (serratol): GC/EIMS: m/z (rel.<br />

int. %): 290 [M] + (100); 275 [M‑CH3] + (2); 261 [M‑CHO] + (1); 247<br />

[M‑CH3CO] + (67); UV (n-hexane): λ,(logε): 190 nm (end absorb-<br />

20<br />

ance, 4.4); CD (n-hexane): λ, (Δε): 202 (−12.7); 190 (−7.6); [α] D<br />

−97° (n-hexane c 1.0); −79° (CHCl3 c 1.0). 1Hand 13C‑NMR see<br />

l " Table 1.<br />

Schmidt TJ et al. Complete Structural Assignment… <strong>Planta</strong> Med 2011; 77: 849–850<br />

Supporting information<br />

Copies of all spectra are available as Supporting Information.<br />

Acknowledgements<br />

!<br />

The authors cordially thank Mairin Lenz for excellent technical<br />

assistance.<br />

References<br />

1 Pardhy RS, Bhattacharyya S. Structure of serratol, a new diterpene<br />

cembranoid alcohol from Boswellia serrata Roxb. Indian J Chem 1977;<br />

16 B: 171–173<br />

2 Klein E, Obermann H. (S)-1-Isopropyl-4,8,12-trimethyl-cyclotetradeca-<br />

3E,7E,11E-trien-1-ol, ein neues Cembrenol aus dem ätherischen Öl von<br />

Olibanum. Tetrahedron Lett 1978; 4: 349–352<br />

3 Kobayashi M, Osabe K. Marine terpenes and terpenoids. VII. Minor<br />

cembranoid derivatives, structurally related to the potent anti-tumorpromoter<br />

sarcophytol A, from the soft coral Sarcophyton glaucum.<br />

Chem Pharm Bull 1989; 37: 631–636<br />

4 Moussaieff A, Shohami E, Kashman Y, Fride E, Schmitz ML, Renner F, Fiebich<br />

BL, Munoz E, Ben-Neriah Y, Mechoulam R. Incensole acetate, a novel<br />

anti-inflammatory compound isolated from Boswellia resin, inhibits<br />

nuclear factor-kB activation. Mol Pharmacol 2007; 72: 1657–1664<br />

5 Moussaieff A, Shein NA, Tsenter J, Grigoriadis S, Simeonidou C, Alexandrovich<br />

AG, Trembovler V, Ben-Neriah Y, Schmitz ML, Fiebich BL, Munoz<br />

E, Mechoulam R, Shohami E. Incensole acetate: a novel neuroprotective<br />

agent isolated from Boswellia carterii. J Cereb Blood Flow Metab 2008;<br />

28: 1341–1352<br />

6 Nour AMM, Khalid SA, Brun R, Kaiser M, Schmidt TJ. The antiprotozoal<br />

activity of sixteen Asteraceae species native to Sudan and bioactivityguided<br />

isolation of xanthanolides from Xanthium brasilicum. <strong>Planta</strong><br />

Med 2009; 75: 1363–1368<br />

7 Nour AMM, Khalid SA, Brun R, Kaiser M, Schmidt TJ. The antiprotozoal<br />

activity of methylated flavonoids from Ageratum conyzoides L.<br />

J Ethnopharmacol 2010; 129: 127–130<br />

received September 27, 2010<br />

revised November 17, 2010<br />

accepted November 19, 2010<br />

Bibliography<br />

DOI http://dx.doi.org/10.1055/s-0030-1250612<br />

Published online December 14, 2010<br />

<strong>Planta</strong> Med 2011; 77: 849–850<br />

© Georg Thieme Verlag KG Stuttgart · New York ·<br />

ISSN 0032‑0943<br />

Correspondence<br />

Prof. Dr. Thomas J. Schmidt<br />

Institut für Pharmazeutische Biologie<br />

und Phytochemie (IPBP)<br />

Westfälische Wilhelms-Universität Münster<br />

Hittorfstraße 56<br />

D-48149 Münster<br />

Germany<br />

Phone: + 49 2518 3333 78<br />

Fax: +492518338341<br />

thomschm@uni-muenster.de


NMR Fingerprinting for Analysis<br />

of Hoodia Species and Hoodia Dietary Products<br />

Authors Jianping Zhao 1 , Bharathi Avula 1 , Vaishali C. Joshi 1 , Natascha Techen 1 , Yan-Hong Wang 1 ,<br />

Troy J. Smillie 1 , Ikhlas A. Khan 1,2<br />

Affiliations<br />

Key words<br />

l " NMR<br />

l " fingerprinting<br />

l " multivariate analysis<br />

l " Hoodia species<br />

l " Asclepiadaceae<br />

l " botanical<br />

l " dietary supplement<br />

received July 30, 2010<br />

revised October 28, 2010<br />

accepted Nov. 2, 2010<br />

Bibliography<br />

DOI http://dx.doi.org/<br />

10.1055/s-0030-1250583<br />

Published online December 2,<br />

2010<br />

<strong>Planta</strong> Med 2011; 77: 851–857<br />

© Georg Thieme Verlag KG<br />

Stuttgart · New York ·<br />

ISSN 0032‑0943<br />

Correspondence<br />

Prof. Dr. Ikhlas A. Khan<br />

National Center<br />

for Natural Products Research<br />

Research Institute<br />

of Pharmaceutical Sciences<br />

University of Mississippi<br />

P. O. Box 1848<br />

University, MS 38677<br />

USA<br />

Phone: + 166 2915 78 21<br />

Fax: + 166 2915 7989<br />

ikhan@olemiss.edu<br />

Correspondence<br />

Dr. Jianping Zhao<br />

National Center<br />

for Natural Products Research<br />

Thad Cochran Research Center<br />

University of Mississippi<br />

P. O. Box 1848<br />

University, MS 38677<br />

USA<br />

Phone: + 166 2915 71 51<br />

Fax: + 166 2915 7989<br />

jianping@olemiss.edu<br />

1 National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, University of Mississippi,<br />

University, MS, USA<br />

2 Department of Pharmacognosy, School of Pharmacy, University of Mississippi, University, MS, USA<br />

Abstract<br />

!<br />

Hoodia gordonii, a succulent plant growing in<br />

African arid regions, is used as a botanical dietary<br />

supplement for weight loss. The increasing concerns<br />

on the quality and safety of Hoodia products<br />

call for the needs of more science-based information,<br />

as well as objective and efficient tools for inspection.<br />

In the present study, NMR fingerprinting<br />

and multivariate analysis techniques were applied<br />

for the identification, discrimination, and<br />

quality analysis of Hoodia plant materials and<br />

commercial products. Four Hoodia species,<br />

namely H. gordonii (five authenticated samples),<br />

H. currorii (one authenticated sample), H. parvi-<br />

Introduction<br />

!<br />

Hoodia gordonii (Masson) Sweet ex Decne (Asclepiadaceae),<br />

a succulent plant, grows primarily in<br />

the semi-deserts of South Africa, Botswana, Namibia,<br />

and Angola. There are about 13 species in<br />

the genus Hoodia [1]. Being fleshy succulents and<br />

growing in arid regions, H. gordonii is traditionally<br />

used as food by the indigenous people in the<br />

Kalahari Desert [2]. H. gordonii has gained popularity<br />

in recent years because of its anorectic activity.<br />

Varieties of Hoodia products prepared in<br />

powder, capsule, liquid, or tea form are sold in<br />

health food stores and on the internet as herbal<br />

weight-loss supplements. The demand for<br />

weight-loss products containing H. gordonii is increasing,<br />

especially after the Ephedra product was<br />

banned because of its side effects.<br />

H. gordonii grows slowly [3]. It takes several years<br />

to mature and grows in a limited area with extreme<br />

soil and weather conditions, making the<br />

plant vulnerable to overexploitation and short of<br />

supply. The gap between supply and demand of<br />

H. gordonii plant materials might be reflected to<br />

some extent by the existence of adulteration<br />

Original Papers<br />

flora (three authenticated samples), and H. rushii<br />

(one authenticated sample), were investigated;<br />

the chemicals and characteristic spectral signals<br />

that made most contributions for their differentiations<br />

were revealed. With the aid of NMR fingerprint<br />

analysis, ten Hoodia products sold on<br />

the dietary supplement market were assessed for<br />

their chemical composition and quality. The study<br />

demonstrated that the NMR fingerprinting approach<br />

could be a promising and efficient tool for<br />

the authentication of botanicals.<br />

Supporting information available online at<br />

http://www.thieme‑connect.de/ejournals/toc/<br />

plantamedica<br />

problems for Hoodia commercial products. Many<br />

Hoodia products on the market were found to be<br />

counterfeits, causing more concerns about their<br />

safety and quality [4, 5]. There is a crucial need to<br />

develop effective methods for authentication and<br />

quality analysis of H. gordonii plant material and<br />

commercial products.<br />

So far, H. gordonii is the only sought-after species<br />

for trade and the only source material for manufacturing<br />

Hoodia dietary supplement products.<br />

Previously, our group developed UPLC‑UV‑MS<br />

and HPTLC methods for identifying and quantifying<br />

P57, an oxypregnane glycoside purported as<br />

the active principle in H. gordonii, and several<br />

other pregnane glycosides in different species of<br />

Hoodia genus and related genera [4, 6]. Interestingly,<br />

it was found that besides H. gordonii, other<br />

species of Hoodia genus, namely H. currorii (synonym<br />

H. macranth), H. parviflora, and H. ruschii, also<br />

contained P57 and other pregnane glycosides.<br />

The finding provided the information that it<br />

might be possible to use these other Hoodia species<br />

as substitutes for H. gordonii. However, there<br />

is no sufficient knowledge about the safety and<br />

efficacy of using these related species as supple-<br />

Zhao J et al. NMR Fingerprinting for… <strong>Planta</strong> Med 2011; 77: 851–857<br />

851


852<br />

Original Papers<br />

ments, and even the information about their chemical compositions<br />

is rare. In our previous studies, the chemical fingerprint<br />

analyses of the Hoodia species by using HPLC and HPTLC were reported<br />

[6, 7]. The chromatographic methods can provide information<br />

about the content levels of different pregnane glycosides<br />

in different Hoodia species. However, it is difficult to discriminate<br />

H. gordonii from the other three Hoodia species even though at<br />

least 11 pregnane glycosides reference compounds were used as<br />

markers.<br />

Nuclear magnetic resonance (NMR) spectroscopy is one of the<br />

most powerful chemical analytical tools and widely used for<br />

characterization and structure determination of natural products.<br />

NMR spectra can provide qualitative and quantitative information<br />

regarding the components of a mixture. In principle, any<br />

components containing hydrogen can be detected by 1 H NMR.<br />

Hence, it provides unbiased detections for compounds over all organic<br />

chemical classes and allows direct quantitative comparisons<br />

[8]. NMR has therefore become one of the most useful techniques<br />

contributing to the emerging field of metabolomics [9,<br />

10]. NMR spectroscopy combined with multivariate analysis<br />

techniques have been successfully used to solve problems such<br />

as plant species and cultivar discrimination, metabolite profiling,<br />

and quality assessment of food or herbal medicines [11, 12].<br />

The aim of the present work was to study the variability of metabolite<br />

fingerprints of the Hoodia species and to develop an efficient<br />

method for identification, differentiation, and quality analysis<br />

of H. gordonii plant materials and its products by using the<br />

NMR fingerprinting approach.<br />

Materials and Methods<br />

!<br />

Chemicals<br />

Methanol, acetonitrile, water, and formic acid were of HPLC grade,<br />

purchased from Fisher Scientific. Deuterated methanol (CD3OD,<br />

99.8% D), chloroform (CDCl3, 99.9% D), and D2O (99.9% D) were obtained<br />

from Cambridge Isotope Laboratories, and sodium 3-trimethylsilyl<br />

[2,2,3,3- 2 H(4)] propanoate (TSP) was purchased from<br />

Sigma-Aldrich, Inc.<br />

The standard compounds (P57, 93.6% purity and hoodigoside L,<br />

87.0% purity) were isolated at the National Center for Natural<br />

Products Research (NCNPR). The identity and purity were confirmed<br />

by chromatographic (TLC, HPLC) methods, by the analysis<br />

of the spectroscopic data (IR, 1D- and 2D‑NMR, HR–ESI–MS), and<br />

by comparison with published spectroscopic data [13, 14].<br />

Plant materials<br />

Authenticated samples of H. gordonii were obtained from the<br />

Universiteit Van Die Vrystaat, South Africa (code numbers: 2925,<br />

2926, and 2927), the Missouri Botanical Garden (code number:<br />

2821), and the American Herbal Pharmacopoeia (AHP) (code<br />

number: 3164). The samples of H. parviflora were obtained from<br />

the AHP (code numbers: 3162 and 3163) and a domestic supplier<br />

(code number: 3230). H. ruschii (code number: 2822) and H. currorii<br />

(code number: 2823) samples were obtained from the Missouri<br />

Botanical Garden. The plant material was authenticated by<br />

Dr. Vaishali Joshi. Hoodia commercial products (sample codes:<br />

HCP-1 to HCP-10) claiming to consist solely of 100% pure H. gordonii<br />

were obtained from the market in the USA. Voucher specimens<br />

of all samples were deposited at the National Center for<br />

Natural Products Research (NCNPR), University of Mississippi,<br />

Mississippi, USA.<br />

Zhao J et al. NMR Fingerprinting for… <strong>Planta</strong> Med 2011; 77: 851–857<br />

Sample preparation<br />

The dried plant materials were ground with an electric blender<br />

(IKA Works Inc.) into powder and sieved through a 2-mm sieve.<br />

The commercial Hoodia products investigated in this study were<br />

mixed and triturated with a mortar and pestle prior to further<br />

preparation. The homogenized sample (200 mg) was weighed<br />

and ultrasonicated in 3 mL of methanol for 20 min followed by<br />

centrifugation for 15 min at 4500 rpm. The supernatant was transferred<br />

to a 15-mL vial. The extraction procedure was repeated<br />

thrice, and the respective supernatants combined. The extract<br />

was dried by removing the solvent under vacuum with a Thermo<br />

Savant SpeedVac system (Thermo Scientific, MA). Parallel triplicate<br />

experiments were conducted for each sample. The dried extract<br />

was dissolved in 0.4 mL of CD3OD containing 0.05% (w/v)<br />

TSP, then transferred into a NMR tube for NMR measurement.<br />

NMR measurements<br />

1 H NMR spectra were measured on a Bruker AVANCE DRX400<br />

NMR spectrometer operating at a proton NMR frequency of<br />

400.13 MHz under a temperature of 298 K with Bruker Topspin<br />

software (v. 1.3). A pulse program with 60 dB power level of selective<br />

irradiation for water presaturation during relaxation delay<br />

was used to suppress the residual water signal. For each sample,<br />

16 transients were collected into 64 k data points. The spectra<br />

were recorded with the following parameters: spectral width =<br />

4845 Hz, relaxation delay = 5 s, pulse width = 11.00 µs (90 °). FIDs<br />

were Fourier transformed with LB = 0.3 Hz. The spectra were<br />

manually phased and baseline corrected using Mnova (v. 5.2,<br />

Mestrelab Research S. L.), and calibrated to the signal of TSP at<br />

chemical shift δ = 0.00 ppm. 2D NMR spectra (COSY, TOCSY, HSQC,<br />

and HMBC) were recorded with the standard acquisition programs<br />

for spectral interpretation and component identification.<br />

Multivariate data analysis<br />

1 H NMR spectra were bucketed with equal bin width of 0.02 ppm<br />

over a region of δ 0.5 to 8.5 ppm after phase and baseline corrections.<br />

The spectral regions from δ 4.75 to 5.05 ppm and 3.27 to<br />

3.37 ppm were excluded to eliminate the effects of water suppression<br />

and the residual methanol signal. Binned data were normalized<br />

to the total sum and converted to ASCII format. The data<br />

sets were then imported and subjected to orthogonal partial least<br />

squares project to latent structures-discriminant analysis<br />

(OPLS‑DA), using software SIMCA‑P + (v. 12.0; Umetrics). The data<br />

sets were Pareto-scaled. The OPLS‑DA model was cross-validated<br />

through the exclusion of every 7th spectra in each cross validation<br />

round. Model quality was assessed by means of the goodness<br />

of fit parameter (R 2 ) and cross validation predictive ability [Q 2 (Y)].<br />

To identify and select the significant and reliable variables that<br />

make most contributions to the classification from the OPLS‑DA<br />

model, the VIP (variable importance in the projection) plot, S-plot<br />

[Cov(t,X) vector vs. Corr(t,X) vector], and loading plot with a jackknifed<br />

confidence interval (CIJKjk) were computed using SIMCA‑P<br />

software. Multi-criteria assessment (MCA), i.e., VIP > 1, | Corr(t,X)|<br />

> 0.58, and the span of CIJKjk excluding zero, was used selecting<br />

the most significant and reliable variables [15, 16].<br />

Supporting information<br />

Statistical validation result of the OPLS‑DA model for the samples<br />

of H. gordonii and H. parviflora, as well as VIP plot, S-plot, and<br />

loading plot with CIJKjk from the model are available as Supporting<br />

Information. The PCA scores and loadings plots for the samples<br />

of H. gordonii and H. parviflora were also provided.


Results and Discussion<br />

!<br />

The genus Hoodia belongs to the Asclepiadaceae family. It has<br />

been found that many plants in this family commonly contain<br />

steroidal glycosides as major secondary metabolites [17]. Previous<br />

phytochemical investigations conducted by our group on<br />

H. gordonii led to the isolation and identification of more than<br />

twenty pregnane glycosides, including the purported active component<br />

P57 [13, 14,18]. These pregnane glycosides are characterized<br />

by an aglycone (either hoodigogenin A or calogenin) and<br />

sugar side chains structured by different 6-deoxy- and/or 2,6-dideoxy<br />

hexopyranose moieties (l " Fig. 1). Hoodigoside L was identified<br />

as a major pregnane glycoside present in the organic extract<br />

of H. gordonii [4]. l " Fig. 2 shows a representative 1 H NMR<br />

spectrum of H. gordonii extract, as well as the spectra of pure<br />

compound P57 and hoodigoside L. The assignment of signals in<br />

the 1 H NMR spectrum of H. gordonii extract was achieved (see<br />

l " Table 1) with the aid of 13 C NMR, 2D NMR (COSY, TOCSY, HSQC,<br />

and HMBC), and spiking experiments. The broad signals with a<br />

chemical shift in the range of δ 6.88 to 7.08 ppm were assigned<br />

to the olefinic protons from the tigloyl moieties of hoodigogenin<br />

and calogenin glycosides by using COSY and HSQC analyses. The<br />

assignment was further confirmed by the HMBC experiment,<br />

which showed the correlation to the carbonyl carbon (δC 168.1)<br />

of the tiglic acid moiety. The HSQC and HMBC experiments revealed<br />

that the two methyl groups on the tigloyl moiety corresponded<br />

to the two strong signals, one singlet and one doublet,<br />

with chemical shifts at δ 1.88 and 1.83 ppm, respectively. The<br />

tigloyl moiety is a characteristic structure segment of the unique<br />

pregnane glycosides present in H. gordonii. Its NMR signals can be<br />

used as spectral markers when using the NMR fingerprinting approach<br />

for authenticating Hoodia products. The term “spectralfeature-signatures”<br />

was used for such spectral markers in this<br />

study. Another significant singlet signal with strong intensity at<br />

δ 2.19 ppm was ascribed to the methyl group (C-21) of the unique<br />

ketone moiety of hoodigogenin glycosides. The olefinic proton<br />

(H-6) on the pregnane aglycone also presented a characteristic<br />

feature in the NMR fingerprint of H. gordonii. The signals of the<br />

H-6 protons from different pregnane glycosides in the extract<br />

were observed around 5.41 ppm. The pregnane glycosides have<br />

different sugar side chains consisting of various hexopyranoses<br />

(see l " Fig. 1), and the anomeric protons of these sugar moieties<br />

gave the characteristic signals in the region from δ 4.30 to δ<br />

4.77 ppm. Their assignments were confirmed by using 2D NMR<br />

analyses including COSY, HMBC, and HSQC. In addition, the methoxyl<br />

and methyl groups on the hexopyranose side-chain-moieties<br />

of the pregnane glycosides were ascribed to the characteristic<br />

strong singlet signals in the regions of δ 3.34 to 3.72 ppm<br />

and the doublet signals in the region of δ 1.12 to 1.37 ppm, respectively.<br />

Furthermore, the characteristic singlet signals with<br />

relatively strong intensity in the region of δ 0.98 to 1.06 ppm<br />

were attributed to the C-18 and C-19 methyl groups of pregnane<br />

aglycone. All the signals mentioned above can be served as “spectral-feature-signatures”<br />

for identifying or authenticating the<br />

presence of hoodigogenin, calogenin, and their glycosides. Further<br />

spectral analyses revealed that the olefinic protons from unsaturated<br />

long chains of lipids in the extracts gave signals with<br />

chemical shifts at about δ 5.35 ppm. Sucrose, α-glucose, and βglucose<br />

were identified on the basis of 13 C, 2D NMR (COSY,<br />

TOCSY, and HMBC), and spiking experiments. Their anomeric<br />

protons showed characteristic signals at δ 5.40 (d, J = 3.8 Hz), δ<br />

5.12 (d, J = 3.7 Hz), and δ 4.48 (d, J = 8.0 Hz) ppm, respectively.<br />

Original Papers<br />

Fig. 1 Structures of pregnane glycosides and hexopyranoses previously<br />

identified in H. gordonii.<br />

l " Fig. 3 shows the 1 H NMR spectra of four different Hoodia species,<br />

H. gordonii, H. parviflora, H. currorii, andH. ruschii. Some<br />

common features in their spectra were observed for these four<br />

species. For example, all of these Hoodia species demonstrate<br />

the presence of the “spectral-feature-signatures” of pregnane<br />

glycosides in their spectra, although the intensities vary. All extracts<br />

of these species contained lipids, giving the characteristic<br />

broad signals with chemical shifts at around 1.3 and 1.6 ppm.<br />

However, the NMR spectra of H. gordonii and H. ruschii showed<br />

distinct differences from each other at a range of 2.5 to 2.8 ppm.<br />

The spectrum of H. ruschii shows significant strong multiplet signals<br />

at δ 2.57 (dd, J = 16.0, 8.0 Hz), 2.77 (dd, J = 16.0, 4.0 Hz), and<br />

4.30 (dd, J = 4.0, 8.0 Hz), but the intensity of these signals was<br />

very low in the spectrum of H. gordonii. The investigation conducted<br />

by using 2D NMR analyses attributed these resonances to<br />

malic acid. The assignment and presence of malic acid were further<br />

confirmed by a spiking experiment. Another significant difference<br />

between H. gordonii and H. ruschii revealed from their<br />

NMR fingerprints was the difference of the signal intensities for<br />

α-glucose, β-glucose, and sucrose. With calculations from the integrals<br />

of the signals relative to the internal reference TSP (0.05%,<br />

w/v), it was found that the concentrations of α-glucose, β-glu-<br />

Zhao J et al. NMR Fingerprinting for… <strong>Planta</strong> Med 2011; 77: 851–857<br />

853


854<br />

Original Papers<br />

Table 1 Characteristics of 1 H NMR signals observed in H. gordonii extract.<br />

Compound Groupa Chemical shift<br />

δ (ppm)<br />

Pregnane glycosides Tigloyl<br />

=CH (br, d)<br />

6.88–7.08<br />

-CH3 (s)<br />

1.88<br />

-CH3 (d)<br />

Pregnane aglycone<br />

1.83<br />

=CH (H-6) (m)<br />

5.41<br />

-CH3 (H-18) (s)<br />

0.98–1.01<br />

-CH3 (H-19) (s)<br />

Hoodigogin<br />

1.04–1.06<br />

-CH3 (H-21) (s)<br />

Sugar moiety<br />

2.19<br />

-CH (anomeric) (d) 4.30–4.77<br />

-CH3 (d)<br />

1.12–1.37<br />

-OCH3 (s)<br />

3.34–3.72<br />

Sucrose -CH (anomeric) (d) 5.40<br />

α-Glucose -CH (anomeric) (d) 5.12<br />

β-Glucose -CH (anomeric) (d) 4.48<br />

Lipids Fatty acid chain<br />

-CH3 (chain terminal) (t) 0.89<br />

-CH2 (m)<br />

1.28<br />

-CH2 (m)<br />

1.59<br />

=CH (br)<br />

5.35<br />

a Letters in parentheses indicate the peak multiplicity property of proton NMR signal:<br />

s, singlet; d, doublet; t, triplet; m, multiplet; br, broad<br />

cose, and sucrose in H. gordonii were approximately 3.8, 2.3, and<br />

4.7 times, respectively, higher than in H. ruschii.<br />

Significant differences between H. gordonii and H. currorii in<br />

their NMR spectra were observed in the region of δ 2.20 to<br />

2.70 ppm and at 4.30 ppm. Malic acid was identified in H. currorii<br />

also, corresponding to the signals at δ 2.57, 2.77, and 4.30 ppm.<br />

The strong multiplet signals at around 2.33 and 2.45 ppm could<br />

Zhao J et al. NMR Fingerprinting for… <strong>Planta</strong> Med 2011; 77: 851–857<br />

Fig. 2 1 HNMR spectra of H. gordonii extract, P57,<br />

and hoodigoside L. Characteristic resonances assigned<br />

to: 1, olefinic proton on tigloyl moiety;<br />

2, olefinic proton (H-6) on pregnane aglycone;<br />

3, sucrose; 4, α-glucose; 5, β-glucose; 6, methoxyl<br />

on hexopyranose of pregnane glycoside; 7, methyl<br />

(C-21) on hoodigogenin; 8, methyl on tigloyl moiety;<br />

9, methyl on hexopyranose and methylene on<br />

fatty chains; 10, methyl (C-18/C-19) on pregnane<br />

aglycon.<br />

not be assigned to a known compound (or compounds) because<br />

of the complexity of the resonances. Another significant difference<br />

between H. gordonii and H. currorii in their NMR fingerprints<br />

that could be clearly observed was the difference of sucrose<br />

signals. The signal of sucrose at δ 5.40 ppm (J =3.8Hz) in<br />

H. currorii was undetectable. Furthermore, the quantitative analyses<br />

through the integrals of the anomeric signals revealed that<br />

the concentrations of α-glucose and β-glucose in H. gordonii were<br />

approximately 2.2 and 1.4 times, respectively, higher than in H.<br />

currorii.<br />

The 1 H NMR spectra of H. gordonii and H. parviflora illustrated<br />

close similarity regarding the “spectral-feature-signatures” of<br />

pregnane glycosides, although a different feature pattern in the<br />

range of δ 3.3 to 4.2 ppm (sugar range) was observed. The discrimination<br />

of H. gordonii from H. parviflora was further conducted<br />

by analysis of spectra using chemometric methods. Chemometrics<br />

is an effective tool for visualizing and deciphering<br />

the useful information hidden within NMR spectra [19]. Mutlivariate<br />

analysis is used to describe sample clustering and to detect<br />

the chemical compounds responsible for the separation of<br />

samples. To understand the significance of variables ( 1 H NMR signals)<br />

contributing to the differentiation between H. gordonii and<br />

H. parviflora, the data sets derived from the NMR spectra of samples<br />

of these two species were subjected to OPLS‑DA. OPLS‑DA is<br />

a supervised multivariate analysis tool that focuses on analyzing<br />

variance between the sample groups (interclass variation), making<br />

the interpretation straightforward [16].<br />

l " Fig. 4 shows the scores scatter plot from the OPLS‑DA model for<br />

H. gordonii and H. parviflora samples collected from different<br />

sources. The model presented a clear separation between the<br />

two species along the predictive component (tP) dimension. Besides,<br />

discernible subgroupings were observed for the samples<br />

within the same species group along the orthogonal component<br />

(t o) dimension. Such sub-separations would be explained as the<br />

variations of metabolites in individual samples collected from


different sources. Many factors such as the growing site, age of<br />

plant, and harvest season could cause the variation of metabolites<br />

in plant materials. The model had an overall goodness of fit,<br />

R 2 (Y), of 98.4% and an overall cross-validation coefficient, Q 2 (Y),<br />

of 98.0%. To test the possibility of correlation by chance, a “yscrambling”<br />

validation was carried out to assess the risk that the<br />

model was overfitted or spurious [20]. A substantial decrease in<br />

both values of parameters R 2 (Y) and Q 2 (Y) was observed after<br />

performing 200 rounds of random permutations, and the extrapolated<br />

value of the Q 2 regression line was − 0.298 (Fig. 1S in<br />

Supporting Information). Generally, the criteria for validity are<br />

Original Papers<br />

Fig. 3 1 H NMR spectra of Hoodia species. 1, H. gordonii;<br />

2. H. parviflora; 3, H. currorii; 4, H. ruschii.<br />

Fig. 4 OPLS‑DA scores scatter plot for H. gordonii<br />

samples (Δ, 2925; ○, 2926; ◊, 2927; ∇, 2821; ▫,<br />

3164) and H. parviflora samples (▲, 3162; ♦, 3163;<br />

▪, 3230).<br />

that all Q 2 values to the left are lower than the original points to<br />

the right or the regression line of the Q 2 points intersects the vertical<br />

axis below zero. The validation results demonstrated that<br />

the constructed model was statistically valid and that the value<br />

of predictability did not arise from overfitting.<br />

By using a combination analysis of VIP statistics, S-plot, and loading<br />

plot with a jack-knifed confidence interval as a tool [15, 16],<br />

the significant and reliable variables (SRVs) that made the greatest<br />

contribution to the discrimination of H. gordonii and H. parviflora<br />

in the constructed OPLS‑DA model were further investigated.<br />

The results revealed that the NMR spectral signals corre-<br />

Zhao J et al. NMR Fingerprinting for… <strong>Planta</strong> Med 2011; 77: 851–857<br />

855


856<br />

Original Papers<br />

sponding to these SRVs were ascribed to saccharides including<br />

sucrose (δ 5.40), α-glucose (δ 5.12), and β-glucose (δ 4.48), and<br />

lipids (with characteristic broad peaks at around δ 0.89, 1.28,<br />

1.59, and 2.01 ppm for fatty chain) (Fig. 2S in the Supporting Information).<br />

The concentrations of sucrose, α-glucose, and β-glucose<br />

in extracts of H. gordonii and H. parviflora were calculated<br />

from the integrals of their signals relative to that of the internal<br />

reference TSP (0.05% w/v), and the results showed that the concentrations<br />

of sucrose, α-glucose, and β-glucose in H. gordonii<br />

were approximately 2.1, 2.3, and 2.2 times, respectively, higher<br />

than in H. parviflora.<br />

H. gordonii is the only sought-after species for trade and the only<br />

source material for manufacturing Hoodia dietary supplement<br />

products so far. The adulteration problem related to Hoodia products<br />

has raised concerns about their quality and safety [4, 5]. NMR<br />

fingerprint analysis can be used as an efficient tool to provide information<br />

on the identity and quality of Hoodia products. In the<br />

present study, we investigated ten commercial Hoodia products,<br />

which were sold on the market and claimed to consist solely of<br />

100% pure H. gordonii.<br />

l " Fig. 5 shows the 1 H NMR spectra of H. gordonii and ten commercial<br />

Hoodia products. A close examination of the spectra for<br />

the tested products revealed that the products HCP-2, HCP-3,<br />

HCP-5, HCP-6, HCP-9, and HCP-10 had significant different spectral<br />

features in comparison with that of the H. gordonii reference.<br />

There was no presence of the “spectral-feature-signatures”,characteristic<br />

signals of the NMR spectrum of H. gordonii, in the spec-<br />

Zhao J et al. NMR Fingerprinting for… <strong>Planta</strong> Med 2011; 77: 851–857<br />

Fig. 5 1 H NMR spectra of H. gordonii and Hoodia<br />

products (HCP-1 to HCP-10).<br />

tra of these products. Therefore, these samples can be easily<br />

sorted as counterfeits according to their NMR fingerprints. The<br />

“spectral-feature-signatures” of H. gordonii could be detected in<br />

the NMR fingerprints of products HCP-1, HCP-4, HCP-7, and HCP-<br />

8. The spectrum of product HCP-1 matched very well with that of<br />

the H. gordonii reference, indicating it was a true product of<br />

H. gordonii. Product HCP-7 showed a spectral pattern that agreed<br />

with the reference spectrum of H. gordonii, but the signals at δ<br />

5.40, 5.12, 4.48 ppm, arising from sucrose, α-glucose, and β-glucose,<br />

respectively, showed relatively lower intensities compared<br />

to those of the H. gordonii standard. A much better match was observed<br />

when comparing the spectrum with that of H. parviflora.<br />

Therefore, product HCP-7 might have H. parviflora as its source<br />

material. Products HCP-4 and HCP-8 show clear “spectral-feature-signatures”<br />

of H. gordonii in their NMR fingerprints. However,<br />

the additional signals, showing at δ 2.26 (broad triplet),<br />

1.65 (broad), and 0.88 (broad triplet) ppm, as well as a stronger<br />

broad peak at around δ 1.29, suggested that these two products<br />

contained additional components. All ten products were analyzed<br />

using UPLC‑UV‑MS to determine the presence of P57 [4],<br />

and the results showed that the products HCP-1, HCP-4, HCP-7,<br />

and HCP-8 did contain P57 but products HCP-2, HCP-3, HCP-5,<br />

HCP-6, HCP-9, and HCP-10 did not, which was in agreement with<br />

the result of the NMR fingerprint analysis. Yet, the NMR approach<br />

enabled us to get a holistic view about the chemical composition<br />

of the products. In addition, the method was simple, rapid, and<br />

easy to conduct.


In summary, NMR fingerprint analysis can be applied as an efficient<br />

tool for identifying and discriminating Hoodia species and<br />

commercial products. The approach offered holistic views for<br />

the chemical composition of Hoodia plant materials and commercial<br />

products without bias. Since the composition is directly<br />

associated with the source of plant material and quality of product,<br />

the method can be used as an efficient tool for the assessment<br />

of the authenticity of Hoodia plant materials and quality of<br />

commercial products. In this study, the identification and differentiation<br />

of H. gordonii from H. currorii and H. ruschii were analyzed<br />

by comparing the characteristic features of the NMR fingerprints.<br />

The application of multivariate data analysis allowed for<br />

the visualization of the separation between H. gordonii and<br />

H. parviflora and deciphering of the information on signals and<br />

metabolites that made the greatest contribution to their differentiation.<br />

The chemical markers and spectral signals that most contributed<br />

to the discrimination of H. gordonii from the other three<br />

Hoodia species were revealed. The study demonstrated that the<br />

NMR fingerprinting approach could be a promising and efficient<br />

tool for the identification, differentiation, and quality assessment<br />

of botanicals.<br />

Acknowledgements<br />

!<br />

This research is supported in part by “Science Based Authentication<br />

of Dietary Supplements” and “Botanical Dietary Supplement<br />

Research” funded by the Food and Drug Administration grant<br />

numbers 5U01FD002071-09 and 1U01FD003871-01, and the<br />

United States Department of Agriculture, Agricultural Research<br />

Service, Specific Cooperative Agreement No. 58-6408-2-0009.<br />

The authors would like to thank the Missouri Botanical Garden,<br />

Missouri, USA, and AHP, USA for providing plant samples. The authors<br />

also would like to thank Prof. Jan (JH) Van der Westhuizen<br />

and Dr. PC Zietsman for providing the H. gordonii samples.<br />

References<br />

1 Mabberly DJ. The plant-book. A portable dictionary of the vascular<br />

plants. Cambridge: Cambridge University Press; 1997: 354<br />

2 van Heerden FR, Marthinus HR, Maharaj VJ, Vleggaar R, Senabe JV, Gunning<br />

PJ. An appetite suppressant from Hoodia species. Phytochemistry<br />

2007; 68: 2545–2553<br />

3 Tibe O, Modise DM, Mogotsi KK. Potential for domestication and commercialization<br />

of Hoodia and Opuntia species in Botswana. Afr J Biotechnol<br />

2008; 7: 1199–1203<br />

Original Papers<br />

4 Avula B, Wang YH, Pawar RS, Shukla YJ, Smillie TJ, Khan IA. A rapid<br />

method for chemical fingerprint analysis of Hoodia species, related<br />

genera, and dietary supplements using UPLC‑UV‑MS. J Pharm Biomed<br />

Anal 2008; 48: 722–731<br />

5 Robb-Nicholson C. By the way, doctor. Iʼve seen a lot of Internet ads for<br />

Hoodia, a natural supplement that suppresses your appetite. What do<br />

you know about it? Does it work, is it safe? Harv Womens Health<br />

Watch 2008; 15: 8<br />

6 Rumalla CS, Avula B, Shukla YJ, Wang YH, Pawar RS, Smillie TJ, Khan IA.<br />

Chemical fingerprint of Hoodia species, dietary supplements, and related<br />

genera by using HPTLC. J Sep Sci 2008; 31: 3959–3964<br />

7 Avula B, Wang YH, Pawar RS, Shukla YJ, Khan IA. Chemical fingerprinting<br />

of Hoodia species and related genera: chemical analysis of oxypregnane<br />

glycosides using high-performance liquid chromatography with<br />

UV detection in Hoodia gordonii. J AOAC Int 2007; 90: 1526–1531<br />

8 Teng R, Junankar PR, Bubb WA, Rae C, Mercier P, Kirk K. Metabolite<br />

profiling of the intraerythrocytic malaria parasite Plasmodium falciparum<br />

by 1H NMR spectroscopy. NMR Biomed 2009; 22: 292–302<br />

9 Powers R. NMR metabolomics and drug discovery. Magn Reson Chem<br />

2009; 47: S2–S11<br />

10 Verpoorte R, Choi Y, Kim H. NMR-based metabolomics at work in phytochemistry.<br />

Phytochem Rev 2007; 6: 3–14<br />

11 van der Kooy F, Maltese F, Choi YH, Kim HK, Verpoorte R. Quality control<br />

of herbal material and phytopharmaceuticals with MS and NMR based<br />

metabolic fingerprinting. <strong>Planta</strong> Med 2009; 75: 763–775<br />

12 Ward JL, Baker JM, Beale MH. Recent applications of NMR spectroscopy<br />

in plant metabolomics. FEBS J 2007; 274: 1126–1131<br />

13 Pawar RS, Shukla YJ, Khan IA. New calogenin glycosides from Hoodia<br />

gordonii. Steroids 2007; 72: 881–891<br />

14 Pawar RS, Shukla YJ, Khan SI, Avula B, Khan IA. New oxypregnane glycosides<br />

from appetite suppressant herbal supplement Hoodia gordonii.<br />

Steroids 2007; 72: 524–534<br />

15 Son HS, Hwang GS, Kim KM, Kim EY, van den Berg F, Park WM, Lee CH,<br />

Hong YS. 1 H NMR-based metabolomic approach for understanding the<br />

fermentation behaviors of wine yeast strains. Anal Chem 2009; 81:<br />

1137–1145<br />

16 Wiklund S, Johansson E, Sjostrom L, Mellerowicz EJ, Edlund U, Shockcor<br />

JP, Gottfries J, Moritz T, Trygg J. Visualization of GC/TOF‑MS-based metabolomics<br />

data for identification of biochemically interesting compounds<br />

using OPLS class models. Anal Chem 2008; 80: 115–122<br />

17 van Heerden FR. Hoodia gordonii: a natural appetite suppressant.<br />

J Ethnopharmacol 2008; 119: 434–437<br />

18 Shukla YJ, Pawar RS, Ding Y, Li XC, Ferreira D, Khan IA. Pregnane glycosides<br />

from Hoodia gordonii. Phytochemistry 2009; 70: 675–683<br />

19 Lindon JC, Nicholson JK. Analytical technologies for metabonomics and<br />

metabolomics, and multi-omic information recovery. Trends Analyt<br />

Chem 2008; 27: 194–204<br />

20 Kang J, Choi MY, Kang S, Kwon HN, Wen H, Lee CH, Park M, Wiklund S,<br />

Kim HJ, Kwon SW, Park S. Application of a 1 H nuclear magnetic resonance<br />

(NMR) metabolomics approach combined with orthogonal projections<br />

to latent structure-discriminant analysis as an efficient tool for<br />

discriminating between Korean and Chinese herbal medicines. J Agric<br />

Food Chem 2008; 56: 11 589–11 595<br />

Zhao J et al. NMR Fingerprinting for… <strong>Planta</strong> Med 2011; 77: 851–857<br />

857


858<br />

Original Papers<br />

Distribution, Synthesis, and Absorption<br />

of Kynurenic Acid in Plants<br />

Authors Michal P. Turski 1 , Monika Turska 1 , Wojciech Zgrajka 1 , Magdalena Bartnik 2 , Tomasz Kocki 3 , Waldemar A. Turski 1,3<br />

Affiliations<br />

Key words<br />

l " kynurenic acid<br />

l " medicinal plants<br />

l " synthesis<br />

l " absorption<br />

received May 15, 2009<br />

revised Nov. 8, 2010<br />

accepted Nov. 15, 2010<br />

Bibliography<br />

DOI http://dx.doi.org/<br />

10.1055/s-0030-1250604<br />

Published online December 14,<br />

2010<br />

<strong>Planta</strong> Med 2011; 77: 858–864<br />

© Georg Thieme Verlag KG<br />

Stuttgart · New York ·<br />

ISSN 0032‑0943<br />

Correspondence<br />

Waldemar A. Turski<br />

Department of Toxicology<br />

Institute<br />

of Agricultural Medicine<br />

Jaczewskiego 2<br />

20-950 Lublin<br />

Poland<br />

Phone: + 48 81718 45 43<br />

Fax: + 4881747 8646<br />

turskiwa@op.pl<br />

1 Department of Toxicology, Institute of Agricultural Medicine, Lublin, Poland<br />

2 Department of Pharmacognosy with Medicinal Plant Laboratory, <strong>Medica</strong>l University, Lublin, Poland<br />

3 Department of Experimental and Clinical Pharmacology, <strong>Medica</strong>l University, Lublin, Poland<br />

Abstract<br />

!<br />

Kynurenic acid (KYNA) is an endogenous antagonist<br />

of the ionotropic glutamate receptors and the<br />

α7 nicotinic acetylcholine receptor as well as an<br />

agonist of the G-protein-coupled receptor<br />

GPR35. In this study, KYNA distribution and synthesis<br />

in plants as well as its absorption was researched.<br />

KYNA level was determined by means<br />

of the high-performance liquid chromatography<br />

with fluorescence detection. KYNA was found in<br />

leaves, flowers, and roots of tested medicinal<br />

herbs: dandelion (Taraxacum officinale), common<br />

nettle (Urtica dioica), and greater celandine (Chelidonium<br />

majus). The highest concentration of this<br />

compound was detected in leaves of dandelion – a<br />

Introduction<br />

!<br />

Kynurenic acid (KYNA) is an oxidative pathway<br />

metabolite of tryptophan formed along the kynurenine<br />

pathway. Its presence was first demonstrated<br />

in urine by Liebig in 1853 [1]. KYNA is an<br />

endogenous antagonist of the ionotropic glutamate<br />

receptors and the α7 nicotinic acetylcholine<br />

receptor. Moreover, it exerts an anticonvulsant<br />

and neuroprotective activity [2–6]. KYNA is an agonist<br />

of the G-protein-coupled receptor GPR35,<br />

which is predominantly expressed in the gastrointestinal<br />

tissues [7]. Based on electrophysiological<br />

studies, KYNA exerts a modulatory effect on<br />

the neurotransmission in the brain [3,8]. The possible<br />

role of KYNA in the central nervous system<br />

was recently reviewed by Wonodi and Schwarcz,<br />

2010 [9], Vamos et al., 2009 [10], and Erhardt et<br />

al., 2009 [11].<br />

In recent years, the identification of KYNA and its<br />

potential role outside the brain became popular.<br />

KYNA was found in urine, serum, amniotic and<br />

synovial fluid, where its concentration varied<br />

from 0.02–0.8 µM [12–15].<br />

Turski MP et al. Distribution, Synthesis, and … <strong>Planta</strong> Med 2011; 77: 858–864<br />

mean value of 0.49 µg/g wet weight. It was shown<br />

that KYNA can be synthesized enzymatically in<br />

plants from its precursor, L-kynurenine, or absorbed<br />

by plants from the soil. Finally, the content<br />

of KYNA was investigated in 21 herbal tablets,<br />

herbal tea, herbs in sachets, and single herbs in<br />

bags. The highest content of KYNA in a maximum<br />

daily dose of herbal medicines appeared in<br />

St. Johnʼswort– 33.75 µg (tablets) or 32.60 µg (sachets).<br />

The pharmacological properties of KYNA<br />

and its presence in high concentrations in medicinal<br />

herbs may suggest that it possesses therapeutic<br />

potential, especially in the digestive system<br />

and should be considered a new valuable dietary<br />

supplement.<br />

More recently, it was demonstrated that KYNA is<br />

a constituent of the rat intestinal fluid and that its<br />

content increases along the small intestine,<br />

reaching a concentration of 16 µM in its distal<br />

part [13, 16]. Interestingly, KYNA in a concentration<br />

found in the digestive system may interact<br />

with the glycine site of the N-methyl-D-aspartate<br />

(NMDA) glutamate receptors, the α7 nicotinic receptor,<br />

and the GPR35 receptor [16]. Furthermore,<br />

it was shown that KYNA protects against<br />

the experimentally induced gastric ulcer [17, 18],<br />

attenuates the intestinal motility in experimental<br />

colonic obstruction in dogs [19] and decreases the<br />

motility and the inflammatory activation in the<br />

early phase of acute experimental colitis in rats<br />

[20]. Taken together, these findings suggest a potential<br />

role of KYNA in the regulation of the gastrointestinal<br />

tract.<br />

In our previous study, it was proved that KYNA is<br />

present in food and honeybee products, being<br />

found in all 37 tested samples. The highest concentration<br />

of KYNA was obtained from the samples<br />

of honeybee products: propolis, multiflorous<br />

honey, and bee pollen. A high concentration of


KYNA was also found in fresh broccoli and potatoes [18]. Therefore,<br />

the aim of this study was to investigate the content and distribution<br />

of KYNA in medicinal herbs, soil, surface water, and natural<br />

fertilizer, as well as to discover whether KYNA is synthesized<br />

in plants or absorbed by them.<br />

Materials and Methods<br />

!<br />

Standard and reagents<br />

Kynurenic acid (KYNA), the standard (98.5% purity, determined<br />

by HPLC) used for the quantitative analysis of KYNA presence in<br />

the plant extracts, was purchased from Sigma. All high-performance<br />

liquid chromatography (HPLC) reagents were purchased<br />

from J.T. Baker and were of the highest available purity: zinc acetate<br />

(analytical purity 100%), sodium acetate (analytical purity<br />

100%, assay by non-aqueous titration), water (HPLC gradient<br />

grade), acetonitrile (HPLC ultra gradient grade, purity determined<br />

by GC), and isopropyl alcohol (99.5%). L-kynurenine used<br />

as a sulfate salt (purity 99%, assay by titration), triethylammonium<br />

phosphate (HPLC grade), and aminooxyacetic acid (AOAA)<br />

were obtained from Sigma. Hydrochloric acid (J.T. Baker) and trichloroacetic<br />

acid (POCH) were of an analytical grade. For purification<br />

of the extracts obtained from the plant material and the<br />

environmental sources and for extraction of KYNA, cation exchange<br />

resin Dowex 50 WX4–400, purchased from Sigma, was<br />

used.<br />

Herbal medicines<br />

The following herbal tablets were used: camomile (Matricaria<br />

chamomilla; batch 01072007), flax (Linum usitatissimum; batch<br />

803), midland hawthorn (Crataegus oxyacantha; batch 803),<br />

heartsease (Viola tricolor; batch 804), lemon balm (Melissa officinalis;<br />

batch 804), small-leaved-lime (Tilia cordata; batch<br />

01012007), common nettle (Urtica dioica; batch 803), and<br />

St. Johnʼs wort(Hypericum perforatum; batch 02042007). All of<br />

those tablets were purchased from Zakłady Farmaceutyczne Colfarm<br />

S.A. Broccoli (Brassica oleracea var. botrytis; batch AL003)<br />

purchased from Unipharm sp. z o. o. and capsules with lupulin<br />

(Lupulinum; batch 010408) purchased from Herbapol Kraków<br />

S. A. were used. The only herbal tea used, nettle root (Urticae radix),<br />

was purchased from Dary Natury Mirosław Angielczyk. The<br />

following herbs in sachets were used: lime flower (Tiliae flos)<br />

and nettle leaf (Urticae folium), both purchased from Herbapol<br />

Białystok S. A. The following single herbs in bags were used: matricaria<br />

flower (Matricariae flos), elderberry flower (Sambuci<br />

flos), common mallow flower (Malvae flos), peppermint leaf<br />

(Menthae piperitae folium), mullein flower (Verbasci flos),<br />

St. Johnʼs wort (Hyperici herba) (all supplied by “Kawon-Hurt”<br />

Nowak Sp. J.), strawflower (Helichrysi inflorescentia; Zakład<br />

Konfekcjonowania Ziół FLOS Elżbieta i Jan Głąb), and meadowsweet<br />

herb (Ulmariae herba; Herbapol Białystok S.A.).<br />

Medicinal herbs: dandelion (Taraxacum officinale), greater celandine<br />

(Chelidonium majus), common nettle (Urtica dioica), and<br />

horse-chestnut (Aesculus hippocastanum) were collected from<br />

the natural state, divided into organs: leaves, flowers, and roots<br />

and analyzed when fresh. Herbs were identified by Dr. M. Bartnik,<br />

a pharmacognosist.<br />

Original Papers<br />

Water, soil, and natural fertilizer<br />

Water samples were collected from four lakes and one river in Lublin,<br />

Opole Lubelskie and Poniatowa, Poland. Forest soil was collected<br />

in Ludwin, Poland. Peat was collected in Piaseczno, Poland.<br />

Fresh natural fertilizer – dung – was obtained from a farm in Jakubowice,<br />

Poland.<br />

Cultivation of garden cress<br />

Garden cress (Lepidium sativum) seeds (Plantico) were seeded in<br />

the artificial soil formed from cellulose pads on petri dishes<br />

(50 mm diameter) or in the natural soil (Agroplanta – universal<br />

natural soil for plants, PPHU “Atlas-<strong>Planta</strong>”) in flowerpots<br />

(62 × 62 × 62 mm) – 100 seeds in each flowerpot or on each petri<br />

dish.<br />

Adding KYNA to the artificial soil<br />

The artificial soil formed from cellulose pads was divided into 3<br />

groups and placed on petri dishes. The first group was watered<br />

with 4 mL of tap water, the second group with 4 mL of KYNA solution<br />

(pH = 7.0) in the concentration of 1 mM, and the third<br />

group with 4 mL of KYNA solution (pH = 7.0) in the concentration<br />

of 10 mM on the first day. On the following days, the artificial soil<br />

was watered with tap water only.<br />

Adding KYNA to the natural soil<br />

The natural soil was divided into 3 groups. The first group was<br />

watered with 200 mL of tap water per 1 kg of soil (control group),<br />

the second group with 200 mL of KYNA solution (pH = 7.0) in the<br />

concentration of 1 mM per 1 kg of soil, and the third group with<br />

200 mL of KYNA solution (pH = 7.0) in the concentration of<br />

10 mM per 1 kg of soil on the first day. The natural soil was then<br />

thoroughly mixed. On the following days, the natural soil was<br />

watered with tap water only.<br />

KYNA synthesis in plants<br />

Dandelion (Taraxacum officinale) was collected and cleaned from<br />

the remaining soil. Fresh plants were divided into roots, leaves,<br />

and flowers. Samples were weighed and Tris solution was added<br />

to all of them (1 : 4 w/v). Samples were then homogenized (Sonopuls<br />

2070; Bandelin Electronic GMBH & Co. KG) and centrifuged<br />

(4000 rpm, 5 min). Samples from each part of a plant – roots,<br />

leaves, and flowers separately – were placed in culture wells in<br />

the volume of 0.9 mL each and incubated in the presence of Lkynurenine<br />

in the volume of 0.1 mL for 2 hours at room temperature.<br />

The final volume of a solution was 1 mL. To stop the reaction,<br />

the plates were placed on ice. 1 mL of supernatant was collected<br />

for KYNA determination. Other samples of the homogenate<br />

of dandelion leaves were placed in culture wells in the volume<br />

of 0.9 mL and divided into 2 groups: the first group was incubated<br />

in the presence of AOAA in the concentration of 50 µM<br />

and in the volume of 0.1 mL, while the second group was incubated<br />

in the presence of both L-kynurenine and AOAA in the concentration<br />

of 50 µM and in the volume of 0.1 mL for 2 hours at<br />

room temperature. The final volume of a solution was 1 mL. To<br />

stop the reaction, the plates were placed on ice. 1 mL of supernatant<br />

was collected for KYNA determination.<br />

Preparation of samples for KYNA determination<br />

Distilled water was added to herbal tablets (1 : 5 w/v) and they<br />

were left for 30 minutes. Then they were homogenized and centrifuged<br />

(4000 rpm, 5 min) and 1 mL of supernatant was collected<br />

for further experiments. Herbal tea, herbs in sachets, and<br />

Turski MP et al. Distribution, Synthesis, and … <strong>Planta</strong> Med 2011; 77: 858–864<br />

859


860<br />

Original Papers<br />

single herbs in bags were prepared according to producersʼ<br />

guidelines. All the extracts were made using methods common<br />

in pharmacy: decoction, maceration, and infusion. Subsequently,<br />

they were centrifuged (4000 rpm, 5 min), and 1 mL of supernatant<br />

was collected for further experiments. Fresh medicinal herbs<br />

were cleaned from the remaining soil and were divided into<br />

parts. Samples were weighed, and distilled water was added to<br />

them (1 : 5 w/v). They were then homogenized and centrifuged<br />

(4000 rpm, 5 min), and 1 mL of supernatant was collected for further<br />

experiments.<br />

15 mL of water samples obtained from the lakes and the river<br />

were centrifuged (4000 rpm, 5 min), and 10 mL of supernatant<br />

was collected for further experiments.<br />

Samples of soil and dung were weighed, and distilled water was<br />

added to them (1 : 5 w/v). They were then homogenized and centrifuged<br />

(4000 rpm, 5 min), and 1 mL of supernatant was collected<br />

for further experiments.<br />

Samples of garden cress (Lepidium sativum) growing in the natural<br />

soil with or without added KYNA solution and in the artificial<br />

soil with or without added KYNA solution were cut 5 mm<br />

above the ground after 4 days. Plants were then counted,<br />

weighed and thoroughly washed. Distilled water was added to<br />

them (1 : 5 w/v). They were then homogenized and centrifuged<br />

(4000 rpm, 5 min), and 1 mL of supernatant was collected for further<br />

experiments.<br />

Measurement of KYNA<br />

KYNA was isolated and determined according to the methods described<br />

previously [5, 22]. Samples were acidified with 50% trichloroacetic<br />

acid. Denaturated proteins were removed by centrifugation<br />

(4000 rpm, 5 min) and supernatant acidified with 0.1 N<br />

HCl was applied to the columns containing cation exchange resin<br />

Dowex 50 prewashed with 0.1 N HCl. Subsequently, the columns<br />

were washed with 1 mL of 0.1 N HCl and 1 mL of water. The fraction<br />

containing KYNA was eluted with 4 mL of water. The eluate<br />

was subjected to HPLC, and KYNA was detected fluorometrically<br />

(system I – Hewlett Packard 1050 HPLC system: ESA catecholamine<br />

HR-80, 3 µm, C18 reverse-phase column, mobile phase:<br />

250 mM zinc acetate, 25 mM sodium acetate, 5% acetonitrile,<br />

pH 6.2, flow rate of 1.0 mL/min; fluorescence detector: excitation<br />

244 nm, emission 398 nm). Mobile phase was filtered through a<br />

0.2-µm membrane filter (Alltech Associates) and degassed by<br />

ultrasonic equipment for 30 min. The injection volume was<br />

100 µL. Separation was achieved at an ambient temperature. The<br />

Chromeleon software was employed to control HPLC system I<br />

and process chromatographic data.<br />

At least 10% of randomly assigned samples were subjected to another<br />

HPLC system (system II) which consisted of a Varian Pro<br />

Star pump, ESA catecholamine HR-80, 3 µm, C18 reverse-phase<br />

column, mobile phase: 250 mM zinc acetate, 25 mM sodium acetate,<br />

5% acetonitrile, pH 6.2, flow rate of 1.0 mL/min; Dionex<br />

RF2000 fluorescence detector set at excitation 350 nm and emission<br />

404 nm. Mobile phase was filtered through a 0.2-µm membrane<br />

filter (Alltech Associates) and degassed by ultrasonic<br />

equipment for 30 min. The injection volume was 100 µL. Separation<br />

was achieved at an ambient temperature. The Chromeleon<br />

software was employed to control HPLC system II and process<br />

chromatographic data.<br />

Turski MP et al. Distribution, Synthesis, and … <strong>Planta</strong> Med 2011; 77: 858–864<br />

Identification and purity determination<br />

The Dionex HPLC system (DionexA) consisted of a gradient pump<br />

P580 LPG, manual injector (7725i Rheodyne) with a 20 µL loop,<br />

ODS-2 Hypersil, 5 µm, C18 column (Thermo-Fisher Scientific),<br />

elution solvent (A) acetonitrile, elution solvent (B) 30 mM triethylammonium<br />

phosphate; elution profile: 0–3min 100–90%<br />

B; 4–15 min 90–20% B; 16–22 min 0% B; 23–25 min 0–100% B,<br />

flow rate of 1 mL/min; UVD340S diode array UV detector set at<br />

334 nm. Separation was achieved at an ambient temperature.<br />

The Dionex Chromeleon software was employed to control HPLC<br />

system and process chromatographic data. For identification purposes,<br />

the eluate corresponding to the KYNA fraction obtained<br />

from HPLC system I was used.<br />

Validation of the method of KYNA determination<br />

Linearity: KYNA standard was prepared by dissolving the authentic<br />

substance (obtained from Sigma) in water for HPLC at<br />

the following concentration levels: 0.0000945, 0.000189,<br />

0.000378, 0.0004725, 0.000945, and 0.00189 µg/100 µL in HPLC<br />

system I and 0.0000189, 0.0000378, 0.0000756, 0.0001134,<br />

0.0001512, and 0.000189 µg/100 µL in HPLC system II. The sixpoint<br />

linearity curve was prepared in three replicates. The calibration<br />

curve was obtained by plotting the quotient of the peak<br />

areas against KYNA content. The linearity of the calibration curve<br />

was evaluated by the linear regression analysis. The following r 2<br />

values were obtained: 0.9989 and 0.9939 in HPLC system I and<br />

II, respectively. The precision expressed as a relative standard deviation<br />

(RSD%) was 4.8 and 6.3 in HPLC system I and II, respectively.<br />

Limit of detection: Limit of detection (LOD) was determined on<br />

the basis of the calibration curve. LOD of KYNA was<br />

0.0000201 µg/100 µL in HPLC system I and 0.0000149 µg/100 µL<br />

in HPLC system II. LOQ was 0.0000603 µg/100 µL and<br />

0.0000447 µg/100 µL in HPLC system I and II, respectively.<br />

Recovery: The accuracy of the Dowex extraction method was<br />

evaluated from the recovery studies. Standard KYNA solutions at<br />

6 different concentrations: 0.00378, 0.00756, 0.01512, 0.0189,<br />

0.0378, and 0.0756 µg/4 mL were subjected to the Dowex procedure<br />

and determined by HPLC (as described above). The recovery<br />

for the KYNA standard was 94.77% ± 1.62.<br />

Selectivity: The selectivity of the method was determined by a<br />

comparison of the chromatograms obtained from the different<br />

parts of plants with chromatograms obtained from standards, using<br />

two different mobile phases in HPLC system I: standard mobile<br />

phase as described above and second mobile phase consisting<br />

of: 250 mM zinc acetate, 25 mM sodium acetate, 2.25% acetonitrile,<br />

isopropyl alcohol 2%, pH 6.2, flow rate of 1.0 mL/min.<br />

Moreover, randomly assigned samples (at least 10%) were subjected<br />

to HPLC system II and determined fluorimetrically at an<br />

excitation wavelength of 350 nm instead of 244 nm in HPLC system<br />

I and at an emission wavelength of 398 nm in both systems.<br />

Matrix effect and internal standard: KYNA standard prepared by<br />

dissolving the authentic substance (obtained from Sigma) in<br />

water for HPLC was added to the samples of dandelion leaves before<br />

homogenization at 6 concentration levels: 0.00378, 0.00756,<br />

0.01512, 0.01890, 0.03780, 0.07560 µg/4 mL (n = 5). Then, samples<br />

were processed in HPLC system I as described above. The calibration<br />

curve was obtained by plotting the quotient of the peak<br />

areas (after subtraction of an endogenous KYNA content) against<br />

added KYNA content. The linearity of the calibration curve was<br />

evaluated by the linear regression analysis. Calculated r 2 value<br />

was 0.9971. LOD and LOQ determined on the basis of the calibra-


tion curve were 0.0001124 µg/100 µL and 0.0003372 µg/100 µL,<br />

respectively.<br />

The calculated recovery for the KYNA standard in samples of dandelion<br />

leaves was 70.24% ± 1.22. In order to compensate the matrix<br />

effect in all further experiments, a known amount of the<br />

standard solution of an authentic KYNA was added to samples<br />

before homogenization as an internal standard. Then, samples<br />

were processed as described above. At least 3 original samples<br />

with added KYNA solution as an internal standard were analyzed<br />

for each plant material. The mean value was calculated and used<br />

to determine the content of KYNA.<br />

Statistical analysis<br />

Data were presented as a mean value and a standard error of the<br />

mean (SEM). The statistical analysis was accomplished using oneway<br />

ANOVA followed by the post hoc Tukey test. A p value of less<br />

than 0.05 was considered significant.<br />

Results<br />

!<br />

The substance extracted from herbal tablets, herbal tea, herbs in<br />

sachets, single herbs in bags, fresh medicinal herbs, soil, dung,<br />

and garden cress was compared to the authentic KYNA in two<br />

chromatographic systems. A solution of the authentic KYNA was<br />

added to the samples of herbs which were then processed as described<br />

in the Materials and Methods section. Retention times<br />

and shapes of obtained peaks were compared. In all cases, both<br />

shapes and retention times to peaks of isolated and authentic KY-<br />

NA were identical. Moreover, the substance extracted from dandelion<br />

leaves and eluted at the retention time of authentic KYNA<br />

in HPLC system I was identified by comparing retention times<br />

and diode-array spectra (wavelength range 200–595 nm) with<br />

those of the authentic reference substance. Variation between retention<br />

times which was less than 1% and spectra analysis which<br />

showed accuracy exceeding 99% confirmed the identity and purity<br />

of the peak extracted from dandelion leaves.<br />

Distribution of KYNA in plants was measured in dandelion, common<br />

nettle, greater celandine, and horse-chestnut. KYNA was<br />

found in leaves, flowers, and roots of all three medicinal herbs,<br />

as well as in leaves and flowers of horse-chestnut. A high concentration<br />

of KYNA was found in leaves: horse-chestnut leaves –<br />

0.636 µg/g, dandelion leaves – 0.489 µg/g, nettle leaves – 0.445<br />

µg/g, and greater celandine leaves – 0.283 µg/g. A lower amount<br />

of KYNA was found in flowers: horse-chestnut flowers – 0.043<br />

µg/g, dandelion flowers – 0.041 µg/g, greater celandine flowers –<br />

0.069 µg/g, and common nettle flowers – 0.243 µg/g. The content<br />

of KYNA in roots was as follows: common nettle roots – 0.063 µg/<br />

g, greater celandine roots – 0.022 µg/g, and dandelion roots –<br />

0.011 µg/g (l " Table 1).<br />

KYNA synthesis in plants was investigated in the homogenates of<br />

dandelion leaves, flowers, and roots. KYNA was synthesized in all<br />

investigated parts of dandelion. The most efficient synthesis of<br />

KYNA occurred in the presence of L-kynurenine (compared to<br />

the control group without added L-kynurenine) in the homogenate<br />

of dandelion leaves (1.208 ± 0.020 µg/g/1 h). A less effective<br />

synthesis was observed in the homogenate of dandelion flowers<br />

(0.343 ± 0.003 µg/g/1 h), and the least effective synthesis was<br />

measured in the homogenate of dandelion roots (0.141 ±<br />

0.001 µg/g/1 h) (l " Fig. 1).<br />

Influence of AOAA on KYNA synthesis in plants was measured in<br />

the homogenate of dandelion leaves. AOAA in the concentration<br />

Table 1 Content of kynurenic acid in the different parts of fresh collected<br />

plant material.<br />

Medicinal<br />

plant<br />

KYNA content [µg/g wet weight]<br />

Original Papers<br />

Leaves Flowers Roots<br />

Greater celandine 0.283 ± 0.018 0.069 ± 0.008 0.022 ± 0.001<br />

Common nettle 0.445 ± 0.025 0.243 ± 0.024 0.063 ± 0.007<br />

Dandelion 0.489 ± 0.040 0.041 ± 0.004 0.011 ± 0.001<br />

Horse-chestnut 0.636 ± 0.051 0.043 ± 0.005 not determined<br />

Data are presented as a mean value of 6 determinations and a standard error of the<br />

mean (SEM). KYNA – kynurenic acid<br />

Fig. 1 Kynurenic acid production in the different parts of dandelion. Data<br />

are presented as mean ± SEM, n = 6; one-way ANOVA followed by the post<br />

hoc Tukey test, p < 0.05 versus appropriate group.<br />

Fig. 2 Influence of aminooxyacetic acid (AOAA) upon kynurenic acid production<br />

in the homogenate of dandelion leaves. Data are presented as<br />

mean ± SEM, n = 6; one-way ANOVA followed by the post hoc Tukey test,<br />

p < 0.05 versus L-kynurenine.<br />

of 50 µM lowered KYNA synthesis from its precursor – L-kynurenine<br />

by about 60% (l " Fig. 2).<br />

Turski MP et al. Distribution, Synthesis, and … <strong>Planta</strong> Med 2011; 77: 858–864<br />

861


862<br />

Original Papers<br />

Table 2 Content of kynurenic acid in surface water, forest soil, peat, and natural<br />

fertilizer.<br />

Environmental source KYNA content<br />

Surface water 0.000024 ± 0.000002 µg/mL<br />

Forest soil 0.002365 ± 0.000213 µg/g<br />

Peat 0.005670 ± 0.000395 µg/g<br />

Natural fertilizer (dung) 0.466070 ± 0.046223 µg/g<br />

Data are presented as a mean value of 3 determinations and a standard error of the<br />

mean (SEM); KYNA: kynurenic acid<br />

Table 3 Content of kynurenic acid in a maximum daily dose of herbal tablets.<br />

Medicinal Content<br />

Maximum KYNA content<br />

plant<br />

of medicinal daily dose in a maximum<br />

plant extract [tablets/day] daily dose<br />

[g/tablet]*<br />

[µg/day]<br />

Camomile 0.15 6 0.41<br />

Flax 0.25 6 0.53<br />

Midland<br />

hawthorn<br />

0.10 6 1.19<br />

Lupulin 0.25 8 4.42<br />

Heartsease 0.15 6 5.95<br />

Broccoli 0.50 2 9.51<br />

Common<br />

nettle<br />

0.15 6 12.15<br />

Small-leavedlime<br />

0.15 6 13.55<br />

Lemon balm 0.15 6 15.98<br />

St. Johnʼs wort 0.15 6 30.38<br />

Data are presented as means. Samples were determined in duplicates. * Content of<br />

medicinal plant extract was specified by the supplier<br />

KYNA content was measured in water, forest soil, peat, and dung.<br />

KYNA was found in water obtained from the lakes and a river in a<br />

mean concentration of 0.000 024 µg/mL. KYNA was also found in<br />

the samples of forest soil (0.0024 µg/g), peat (0.0057 µg/g), and<br />

natural fertilizer (dung; 0.47 µg/g) (l " Table 2).<br />

Absorption of KYNA by plants was investigated on the basis of the<br />

cultivation of garden cress. It was found that garden cress growing<br />

in the natural soil with added KYNA solution in the concentration<br />

of 10 mM contained 0.203 µg KYNA/g, which is about 6.5<br />

times more than garden cress growing in the natural soil without<br />

added KYNA solution (which contained 0.031 µg KYNA/g). The<br />

addition of KYNA solution in the concentration of 1 mM into the<br />

natural soil had no influence on the presence of KYNA in garden<br />

cress (0.031 µg/g) (l " Fig. 3).<br />

It was found that garden cress growing in the artificial soil with<br />

added KYNA solution in concentrations of 1 mM or 10 mM contained<br />

2.750 or 20.639 µg KYNA/g, respectively, which is 254<br />

and 1910 times more than garden cress growing in the artificial<br />

soil without added KYNA solution (0.011 µg KYNA/g) (l " Fig. 4).<br />

KYNA was found in all ten tested herbal tablets available on the<br />

market (the amount of KYNA is expressed in µg KYNA in a maximum<br />

recommended daily dose): in camomile – 0.41, flax – 0.53,<br />

midland hawthorn – 1.19, lupulin – 4.42, heartsease – 5.95, broccoli<br />

– 9.51, common nettle – 12.15, small-leaved-lime – 13.55,<br />

lemon balm – 15.98, and St. Johnʼswort– 30.38 (l " Table 3).<br />

The amount of KYNA in herbal tea, herbs in sachets, and single<br />

herbs in bags was expressed in µg KYNA per a maximum daily<br />

dose of the medicinal plant extract recommended by producers.<br />

Turski MP et al. Distribution, Synthesis, and … <strong>Planta</strong> Med 2011; 77: 858–864<br />

Fig. 3 Kynurenic acid absorption by roots of garden cress from the natural<br />

soil enriched with kynurenic acid. Data are presented as mean ± SEM, n = 6;<br />

one-way ANOVA followed by the post hoc Tukey test, p < 0.05 versus appropriate<br />

group.<br />

Fig. 4 Kynurenic acid absorption by roots of garden cress from the artificial<br />

soil containing kynurenic acid. Data are presented as mean ± SEM,<br />

n = 6; one-way ANOVA followed by the post hoc Tukey test, p < 0.05 versus<br />

appropriate group.<br />

KYNA was found in a herbal tea (common nettle root – 1.08 µg),<br />

in two types of herbs in sachets (lime flower – 4.24 µg and common<br />

nettle leaf – 32.50 µg), as well as in the following single<br />

herbs in bags: mullein flower – 2.44 µg, common mallow flower<br />

– 5.98 µg, matricaria flower – 10.17 µg, strawflower – 10.74 µg,<br />

meadowsweet herb – 13.27 µg, peppermint leaf – 19.50 µg, elderberry<br />

flower – 20.74 µg, and St. Johnʼswort– 32.60 µg (l " Table 4).<br />

Discussion<br />

!<br />

To the best of our knowledge, this is the first scientific approach<br />

towards the detailed analysis of KYNA content in medicinal<br />

herbs. Previously, KYNA was mentioned as a constituent of the<br />

plants Gingko biloba [23] and Ephedra transitoria [24]. However,<br />

it was neither unequivocally identified nor had its content been<br />

measured. In our recent research, it was proved that KYNA is<br />

present in food and honeybee products [21], being found in all


Table 4 Content of kynurenic acid in a maximum daily dose of herbal medicines.<br />

Herbal<br />

Method of preparation/ KYNA content<br />

medicine<br />

temperature/time* in a maximum<br />

daily dose* [µg/day]<br />

Nettle root Infusion/90 °C/15 min 1.08<br />

Mullein flower Infusion/90 °C/15 min 2.44<br />

Lime flower Infusion/90 °C/15 min 4.24<br />

Common mallow<br />

flower<br />

Infusion/90 °C/10 min 5.98<br />

Matricaria flower Infusion/90 °C/10 min 10.17<br />

Strawflower Decoction/100 °C/5 min 10.74<br />

Meadowsweet herb Infusion/90 °C/5 min 13.27<br />

Peppermint leaf Infusion/90 °C/10 min 19.50<br />

Elderberry flower Decoction/100 °C/3 min 20.74<br />

Nettle leaf Infusion/90 °C/5 min 32.50<br />

St. Johnʼs wort Infusion/90 °C/60 min 32.60<br />

Data are presented as means. Samples were determined in duplicates.<br />

* Method of preparation and a maximum daily dose were specified by the supplier<br />

37 tested samples. The highest concentration of KYNA was obtained<br />

from the samples of honeybee products, propolis<br />

(1.63 µg/g), multiflorous honey (0.87 µg/g), and bee pollen (0.64<br />

µg/g). A high concentration of KYNA was also found in fresh broccoli<br />

(0.42 µg/g) and potatoes (0.51 µg/g) [21]. An extremely high<br />

level of KYNA in honey (23–2,114 µg/g) was recently reported by<br />

Beretta et al., 2009 [25]. Since propolis, honey, and bee pollen are<br />

commonly used in folk medicine and apitherapy, and they contain<br />

a lot of KYNA as previous studies have indicated [21, 25], it<br />

was decided to investigate the content and distribution of KYNA<br />

in herbal medicines.<br />

Experiments were carried out on four medicinal plants – dandelion,<br />

common nettle, greater celandine, and horse-chestnut. It<br />

was found that KYNA is present in all examined parts of these<br />

plants; nevertheless, its concentration expressed in µg/g wet<br />

weight varies significantly between parts. A high concentration<br />

of KYNA was found in leaves of all chosen plants. A lower concentration<br />

of KYNA was found in flowers, and the lowest concentration<br />

of KYNA was found in roots.<br />

Since achieved results proved our hypothesis that KYNA is<br />

present in medicinal herbs to be correct, it was decided to check<br />

whether KYNA might be synthesized in plants and/or absorbed<br />

by plants. KYNA is formed along the tryptophan–kynurenine<br />

pathway. Conversion from tryptophan to kynurenine is achieved<br />

by tryptophan-2,3-dioxygenase and indole-2,3-dioxygenase, and<br />

KYNA is produced irreversibly from kynurenine by kynurenine<br />

aminotransferases [26]. In order to investigate whether KYNA<br />

might be produced in plants, its precursor – L-kynurenine – was<br />

added to the homogenates of dandelion leaves, flowers, and<br />

roots, and after 2 hours of incubation at room temperature, KYNA<br />

content in all parts mentioned above was measured. It was found<br />

that KYNA is formed in such conditions, and that the rate of KY-<br />

NA synthesis is highest in leaves, followed by flowers and roots.<br />

Comparison between the amount of KYNA and the rate of KYNA<br />

synthesis in dandelion parts suggests that there is a positive correlation<br />

between both values.<br />

Due to the fact that KYNA is formed from kynurenine by the activity<br />

of kynurenine aminotransferases which are blocked by<br />

AOAA, it was decided to check whether KYNA is synthesized<br />

along this pathway in plants. In order to do this, AOAA and kyn-<br />

Original Papers<br />

urenine were added to the homogenate of dandelion leaves. AOAA<br />

significantly reduced KYNA production in such conditions, which<br />

firmly proves that KYNA is enzymatically synthesized in plants.<br />

Further experiments were aimed at checking whether KYNA<br />

might be absorbed by plants from the soil. Firstly, KYNA presence<br />

in the soil was investigated. It was proved that the highest concentration<br />

of KYNA is found in peat, and a considerably lower<br />

concentration in forest soil. Those results suggest that KYNA is<br />

present in soils of an organic origin. An additional argument for<br />

such a thesis is the huge amount of KYNA found in the natural fertilizer<br />

– dung. Moreover, a low concentration of KYNA was found<br />

in surface water. Those results interchangeably suggest that KY-<br />

NA is present in the soil, and that such soil might be a source of<br />

KYNA for plants if they are able to absorb it from the ground. Further<br />

experiments carried out on garden cress supported the assumptions<br />

that KYNA might be absorbed by plants either from a<br />

liquid basis or from the natural soil. Thus, our experiments demonstrate<br />

that KYNA found in plants might be synthesized in a<br />

plant and/or absorbed by a plant. To the best of our knowledge,<br />

this is the first report on the fate of KYNA in plants. The physiological<br />

role of KYNA in plants remains to be elucidated.<br />

Medicinal herbs are marketed in different forms. Single herbs in<br />

bags and herbal sachets are the most popular among the global<br />

society. However, herbal tablets are growing in popularity because<br />

they do not require prior time-consuming self preparation,<br />

they are handy, easy to take, and virtually tasteless. A high concentration<br />

of KYNA in honeybee products [21] and the presence<br />

of KYNA in all tested medicinal herbs prompted us to investigate<br />

the content of KYNA in herbal preparations and herbal tablets<br />

which are available in almost all pharmacies and herbal shops.<br />

Herbal teas and single herbs in bags were prepared in accordance<br />

with the producersʼ guidelines, and KYNA content was measured<br />

in µg KYNA/maximum recommended daily dose. Although KYNA<br />

was found in all tested herbal preparations and herbal tablets, its<br />

concentration varied distinctly between different products; the<br />

highest concentration was measured in St. Johnʼs wort and nettle<br />

leaf. When considering only herbal tablets, the highest concentration<br />

of KYNA was found in St. Johnʼs wort, lemon balm, smallleaved-lime,<br />

and common nettle. The majority of herbal preparations<br />

and herbal tablets containing significant amounts of KYNA,<br />

such as peppermint leaf, lemon balm, small-leaved-lime, common<br />

nettle, and St. Johnʼs wort are used to reduce the symptoms<br />

and cure the diseases of the digestive system. They stimulate secretion<br />

of gastric juices and bile and reduce smooth musclesʼ<br />

spasm. The most recent scientific reports suggest that KYNA is<br />

found in high concentrations in the intestine, bile, and pancreatic<br />

juice [27]. It was proved that this compound protects against<br />

stomach mucous membrane ulceration [17]. Moreover, it was also<br />

demonstrated that KYNA counteracts pathological reactions<br />

caused by ileus in dogs [19] and colitis in rats [20] and suggested<br />

that it plays an important role in regulating the functions of the<br />

digestive system [19,27]. Bearing in mind all those scientific reports,<br />

our results showing a high KYNA concentration in herbal<br />

preparations and herbal tablets used mainly in the diseases of<br />

the digestive system suggest that this compound might be partly<br />

responsible for their healing properties.<br />

The obtained results and recently published scientific reports enable<br />

us to make a hypothesis of an existing KYNA circulation in<br />

the biosphere. KYNA might be synthesized along the enzymatic<br />

pathway in plants and animals, absorbed from food of both vegetable<br />

and animal origin and transported to the soil in animal excrements<br />

and plant debris. Plants might absorb it from the soil.<br />

Turski MP et al. Distribution, Synthesis, and … <strong>Planta</strong> Med 2011; 77: 858–864<br />

863


864<br />

Original Papers<br />

Much less is known about KYNA metabolism. In mammals, KYNA<br />

seems to be an end-product formed along the kynurenine pathway<br />

[28]. However, microbiotics such as Pseudomonas were<br />

found to metabolize this compound to glutamic acid, alanine,<br />

and acetic acid [29]. Thus, the catabolism of KYNA protects<br />

against its accumulation in the biosphere.<br />

Summing up, pharmacological properties of KYNA and its presence<br />

in high concentrations in medicinal herbs may suggest that<br />

it possesses therapeutic potential and should be considered a<br />

new valuable dietary supplement.<br />

Acknowledgements<br />

!<br />

M. P. Turski and M. Turska are students, and volunteers in the Department<br />

of Toxicology. This study was supported in part by the<br />

Foundation for Polish Science.<br />

References<br />

1 Liebig J. Über Kynurensäure. Justus Liebigs Ann Chem 1853; 86: 125–<br />

126<br />

2 Ganong AH, Cotman CW. Kynurenic acid and quinolinic acid act at Nmethyl-D-aspartate<br />

receptors in the rat hippocampus. J Pharmacol<br />

Exp Ther 1986; 236: 293–299<br />

3 Hilmas C, Pereira EF, Alkondon M, Rassoulpour A, Schwarcz R, Albuquerque<br />

EX. The brain metabolite kynurenic acid inhibits alpha7 nicotinic<br />

receptor activity and increases non-alpha7 nicotinic receptor expression:<br />

physiopathological implications. J Neurosci 2001; 21: 7463–7473<br />

4 Perkins MN, Stone TW. An iontophoretic investigation of the actions of<br />

convulsant kynurenines and their interaction with the endogenous excitant<br />

quinolinic acid. Brain Res 1982; 247: 184–187<br />

5 Turski WA, Nakamura M, Todd WP, Carpenter BK, Whetsell Jr WO,<br />

Schwarcz R. Identification and quantification of kynurenic acid in human<br />

brain tissue. Brain Res 1988; 454: 164–169<br />

6 Schwarcz R, Pellicciari R. Manipulation of brain kynurenines: glial targets,<br />

neuronal effects, and clinical opportunities. J Pharmacol Exp Ther<br />

2002; 303: 1–10<br />

7 Wang J, Simonavicius N, Wu X, Swaminath G, Reagan J, Tian H, Ling L.<br />

Kynurenic acid as a ligand for orphan G protein-coupled receptor<br />

GPR35. J Biol Chem 2006; 281: 22 021–22 028<br />

8 Scharfman HE, Goodman JH, Schwarcz R. Electrophysiological effects of<br />

exogenous and endogenous kynurenic acid in the rat brain: studies in<br />

vivo and in vitro. Amino Acids 2000; 19: 283–297<br />

9 Wonodi I, Schwarcz R. Cortical kynurenine pathway metabolism: a novel<br />

target for cognitive enhancement in schizophrenia. Schizophr Bull<br />

2010; 36: 211–218<br />

10 Vamos E, Pardutz A, Klivenyi P, Toldi J, Vecsei L. The role of kynurenines<br />

in disorders of the central nervous system: possibilities for neuroprotection.<br />

J Neurol Sci 2009; 283: 21–27<br />

11 Erhardt S, Olsson SK, Engberg G. Pharmacological manipulation of kynurenic<br />

acid: potential in the treatment of psychiatric disorders. CNS<br />

Drugs 2009; 23: 91–101<br />

Turski MP et al. Distribution, Synthesis, and … <strong>Planta</strong> Med 2011; 77: 858–864<br />

12 Kazda H, Taylor N, Healy D, Walker D. Maternal, umbilical, and amniotic<br />

fluid concentrations of tryptophan and kynurenine after labor or cesarean<br />

section. Pediatr Res 1998; 44: 368–373<br />

13 Milart P, Sikorski R. Kynurenic acid concentration in blood and urine<br />

during normal pregnancy. Ginekol Pol 1998; 69: 968–973<br />

14 Milart P, Urbanska EM, Turski WA, Paszkowski T, Sikorski R. Intrapartum<br />

levels of endogenous glutamate antagonist – kynurenic acid in amniotic<br />

fluid, umbilical and maternal blood. Neurosci Res Commun 1999;<br />

24: 173–178<br />

15 Parada-Turska J, Rzeski W, Zgrajka W, Majdan M, Kandefer-Szerszen M,<br />

Turski W. Kynurenic acid, an endogenous constituent of rheumatoid arthritis<br />

synovial fluid, inhibits proliferation of synoviocytes in vitro.<br />

Rheumatol Int 2006; 26: 422–426<br />

16 Kuc D, Zgrajka W, Parada-Turska J, Urbanik-Sypniewska T, Turski WA.<br />

Micromolar concentration of kynurenic acid in rat small intestine.<br />

Amino Acids 2008; 35: 503–505<br />

17 Glavin GB, Pinsky C. Kynurenic acid attenuates experimental ulcer formation<br />

and basal gastric acid secretion in rats. Res Commun Chem<br />

Pathol Pharmacol 1989; 64: 111–119<br />

18 Glavin GB, Pinsky C, Bose R. Gastrointestinal effects of contaminated<br />

mussels and putative antidotes thereof. Can Dis Wkly Rep 1990; 16<br />

(Suppl. 1E): 111–115<br />

19 Kaszaki J, Palasthy Z, Erczes D, Racz A, Torday C, Varga G, Vecsei L, Boros<br />

M. Kynurenic acid inhibits intestinal hypermotility and xanthine oxidase<br />

activity during experimental colon obstruction in dogs. Neurogastroenterol<br />

Motil 2008; 21: 53–62<br />

20 Varga G, Erces D, Fazekas B, Fulop M, Kovacs T, Kaszaki J, Fulop F, Vecsei L,<br />

Boros M. N-Methyl-D-aspartate receptor antagonism decreases motility<br />

and inflammatory activation in the early phase of acute experimental<br />

colitis in the rat. Neurogastroenterol Motil 2010; 22: 217–225<br />

21 Turski MP, Turska M, Zgrajka W, Kuc D, Turski WA. Presence of kynurenic<br />

acid in food and honeybee products. Amino Acids 2009; 36:<br />

75–80<br />

22 Shibata K. Fluorimetric micro-determination of kynurenic acid, an endogenous<br />

blocker of neurotoxicity, by high-performance liquid chromatography.<br />

J Chromatogr 1988; 430: 376–380<br />

23 Drieu K. Preparation and definition of Ginkgo biloba extract. Presse<br />

Med 1986; 15: 1455–1457<br />

24 al-Khalil S, Alkofahi A, al-Eisawi D, al-Shibib A. Transtorine, a new quinoline<br />

alkaloid from Ephedra transitoria. J Nat Prod 1998; 61: 262–263<br />

25 Beretta G, Artali R, Caneva E, Orlandini S, Centini M, Facino RM. Quinoline<br />

alkaloids in honey: further analytical (HPLC‑DAD‑ESI‑MS, multidimensional<br />

diffusion-ordered NMR spectroscopy), theoretical and<br />

chemometric studies. J Pharm Biomed Anal 2009; 50: 432–439<br />

26 Nemeth H, Toldi J, Vecsei L. Role of kynurenines in the central and peripheral<br />

nervous systems. Curr Neurovasc Res 2005; 2: 249–260<br />

27 Paluszkiewicz P, Zgrajka W, Saran T, Schabowski J, Valverde Piedra JL,<br />

Fedkiv O, Rengman S, Pierzynowski SG, Turski WA. High concentration<br />

of kynurenic acid in bile and pancreatic juice. Amino Acids 2009; 37:<br />

637–641<br />

28 Turski WA, Schwarcz R. On the disposition of intrahippocampally injected<br />

kynurenic acid in the rat. Exp Brain Res 1988; 71: 563–567<br />

29 Horibata K, Taniuchi H, Tashiro M, Kuno S, Hayaishi O. The metabolism<br />

of kynurenic acid. II. Tracer experiments on the mechanism of kynurenic<br />

acid degradation and glutamic acid synthesis by Pseudomonas<br />

extracts. J Biol Chem 1961; 236: 2991–2995


Association between Chemical and Genetic<br />

Variation of Wild and Cultivated Populations<br />

of Scrophularia ningpoensis Hemsl.<br />

Authors Shuting Yang 1,2 *, Chuan Chen 1 *, Yunpeng Zhao 1 ,WangXi 1 , Xiaolong Zhou 3 , Binlong Chen 3 , Chengxin Fu 2<br />

Affiliations<br />

Key words<br />

l " Scrophularia ningpoensis<br />

l " Scrophulariaceae<br />

l " Radix Scrophulariae<br />

l " chemical diversity<br />

l " genetic diversity<br />

l " ISSR fingerprinting<br />

l " HPLC fingerprinting<br />

received April 30, 2010<br />

revised October 4, 2010<br />

accepted Nov. 11, 2010<br />

Bibliography<br />

DOI http://dx.doi.org/<br />

10.1055/s-0030-1250601<br />

Published online December 14,<br />

2010<br />

<strong>Planta</strong> Med 2011; 77: 865–871<br />

© Georg Thieme Verlag KG<br />

Stuttgart · New York ·<br />

ISSN 0032‑0943<br />

Correspondence<br />

Dr. Yunpeng Zhao<br />

Department of Biology<br />

College of Life Sciences,<br />

Zhejiang University<br />

388 Yukangtang Road<br />

Hangzhou 310058<br />

P. R. China<br />

Phone: + 86 57188 20 6463<br />

Fax: + 8657186 4322 73<br />

ypzhao@zju.edu.cn<br />

Correspondence<br />

Prof. Chengxin Fu<br />

Laboratory of Systematic<br />

and Evolutionary Botany<br />

Institute of Plant Sciences,<br />

Zhejiang University<br />

388 Yukangtang Road<br />

Hangzhou 310058<br />

P. R. China<br />

Phone: + 86 57188 20 6607<br />

Fax: + 8657186 4322 73<br />

cxfu@zju.edu.cn<br />

1 The Key Laboratory of Conservation Biology for Endangered Wildlife of the Ministry of Education,<br />

College of Life Sciences, Zhejiang University, Hangzhou, P.R. China<br />

2 Laboratory of Systematic & Evolutionary Botany and Biodiversity, Institute of Plant Sciences<br />

and Conservation Center for Gene Resources of Endangered Wildlife, Zhejiang University, Hangzhou, P. R. China<br />

3 Panʼan Institute of Chinese Materia <strong>Medica</strong>, Panʼan, P.R. China<br />

Abstract<br />

!<br />

Scrophularia ningpoensis Hemsl. is an important<br />

Chinese medicinal herb with a domestication history<br />

of more than one thousand years. Although a<br />

number of studies have focused on either chemical<br />

or genetic variation, none have dealt with<br />

their association to discuss the formation of<br />

chemical diversity. We applied HPLC fingerprinting<br />

with identification of four predominant bioactive<br />

compounds using LC‑ESI‑MS to assess chemical<br />

variation among 6 cultivated and 5 wild populations<br />

of S. ningpoensis. Significant chemical differences<br />

were revealed between wild and cultivated<br />

populations in terms of chromatographic<br />

profiles, principal component analysis (PCA)<br />

plots, and bioactive compounds contents. Compared<br />

to cultivated populations, the chemical profiles<br />

varied considerably among wild populations,<br />

of which some were remarkably similar to cultivated<br />

populations. Inter simple sequence repeats<br />

(ISSR) fingerprinting indicated a genetic differentiation<br />

pattern parallel to chemical variation. Evidence<br />

strongly supported the association be-<br />

Introduction<br />

!<br />

Phytochemical diversity, on which the pharmaceutical<br />

quality of medicinal plants relies, results<br />

from plasticity of plant secondary metabolism<br />

which has evolved to respond to stresses and interactions<br />

with continuously changing environments<br />

[1]. Genetic diversity underlies the plasticity<br />

of secondary metabolism [2]. However, domestication<br />

has led to an overall reduction of genetic<br />

diversity of crop plants, including medicinal<br />

plants, despite increased productivity [3]. The<br />

narrowing genetic variability may weaken crop<br />

* These two authors contributed to this work equally.<br />

tween chemical and genetic variation of S. ningpoensis.<br />

Based on both sets of data, suggestions<br />

are proposed for the conservation of genetic diversity,<br />

crop improvement, and good agricultural<br />

practice. The present results will also facilitate<br />

our theoretical understanding of the selective<br />

and adaptive evolutionary processes of medicinal<br />

plant species impacted by domestication and a<br />

changing environment.<br />

Abbreviations<br />

!<br />

Original Papers<br />

FST: genetic differentiation estimated by<br />

ANOVA<br />

ΦST: inter-population genetic differentiation<br />

estimated by AMOVA<br />

ΦCT: among-group genetic differentiation<br />

estimated by AMOVA<br />

Supporting information available online at<br />

http://www.thieme-connect.de/ejournals/toc/<br />

plantamedica<br />

plasticity with respect to compositional quality<br />

as well as responses to biotic and abiotic stresses<br />

[1, 4]. Therefore, wild ancestors and landraces,<br />

which are likely to bear higher genetic diversity,<br />

attract an increasing interest for their potential<br />

role in improving chemical composition and other<br />

traits in crop plants [4].<br />

Radix Scrophulariae, the root of Scrophularia<br />

ningpoensis Hemsl. (Scrophulariaceae), is a famous<br />

Chinese traditional medicinal herb with a<br />

domestication history of over 1000 years. Iridoids,<br />

like harpagoside, phenylpropanoid glycosides,<br />

like angroside C and acteoside, and cinnamic acid<br />

were revealed to be its main bioactive components<br />

with anti-inflammatory, antimicrobial, and<br />

antitumor activities [5–9]. Several methods based<br />

Yang S et al. Association between Chemical… <strong>Planta</strong> Med 2011; 77: 865–871<br />

865


866<br />

Original Papers<br />

Table 1 Sample information of 16 populations of S. ningpoensis surveyed for ISSR variation and HPLC fingerprinting.<br />

Population<br />

Locality Sample<br />

Sample<br />

Vouchers<br />

Code<br />

Cultivated populations<br />

size for ISSR size for HPLC<br />

DP Dapan, Panan County, Zhejiang Province 15 3 C. Chen, 060601-060603<br />

YC Yaochuan, Panan County, Zhejiang Province 15 3 C. Chen, 060604-060606<br />

RC Renchuan, Panan County, Zhejiang Province 15 3 C. Chen, 060607-060609<br />

HB Enshi City, Hubei Province 10 3 C. Chen, 060614-060616<br />

SX Zhenping County, Shaanxi Province 10 3 C. Chen, 060617-060619<br />

CQ<br />

Wild populations<br />

Jinfo, Chongqing Municipality 19 3 C. Chen, 060620-060622<br />

TM Tianmu Mountain, Zhejiang Province 10 3 C. Chen, 060623-060625<br />

TW Dapan Mountain, Zhejiang Province<br />

(introduced to SH)<br />

10 3 C. Chen, 060626-060628<br />

AH Tiantangzhai, Anhui Province – 4 P. Li, 091120-091122<br />

HN Pingjiang County, Hunan Province – 3 P. Li, 091131-091133<br />

JX Jiujiang County, Jiangxi Province – 3 P. Li, 091134-091136<br />

on HPLC or combined with LC‑ESI‑MS were developed to qualify<br />

and quantify all or part of the foregoing four bioactive compounds<br />

for the purposes of quality assessment on Radix Scrophulariae<br />

[10–14]. HPLC fingerprinting with no or few identified<br />

chromatographic peaks combined with multivariate analysis<br />

was also applied for quality evaluation [15, 16]. The chemical differences<br />

of Radix Scrophulariae among various production regions<br />

were demonstrated to different extents. Compared to<br />

chemical assessment, there are relatively fewer studies on the genetic<br />

variation of S. ningpoensis. Zhao et al. developed ISSR fingerprinting<br />

for this species [17] and analyzed the genetic relationships<br />

among one wild and five cultivated populations [18]. Chen<br />

et al. analyzed the genetic differentiation among three cultivars<br />

using sequence-related amplified polymorphism (SRAP) [19].<br />

Although studies have focused on either chemical or genetic<br />

analyses, no previous work has combined both sets of data to deal<br />

with the association between the chemical and genetic variation<br />

of S. ningpoensis. Therefore, in the present paper we aim (i) to estimate<br />

the chemical variation pattern of wild and cultivated Radix<br />

Scrophulariae from different regions in China using HPLC‑DAD<br />

fingerprinting and LC‑ESI‑MS, (ii) to determine genetic diversity<br />

and differentiation of these S. ningpoensis populations using ISSR<br />

Yang S et al. Association between Chemical … <strong>Planta</strong> Med 2011; 77: 865–871<br />

Fig. 1 Distribution of studied populations of<br />

S. ningpoensis.<br />

markers, and (iii) to probe the possible relationship between<br />

chemical and genetic variation of S. ningpoensis. The results will<br />

facilitate an exploration of the genetic basis of chemical variation<br />

and the development of strategies for utilization and conservation<br />

of S. ningpoensis, as well as understanding evolution of plant<br />

secondary metabolism.<br />

Materials and Methods<br />

!<br />

Sampling<br />

Young leaves of S. ningpoensis were collected from May to June of<br />

2006 and dried in silica gel (l " Table 1; l " Fig. 1). Only two wild<br />

populations (TM and TW) were subjected to ISSR analyses due<br />

to the material availability. All the materials were authenticated<br />

by Professor Chengxin Fu, and voucher specimens were deposited<br />

at the Herbarium of Zhejiang University (HZU). Samples of<br />

Radix Scrophulariae were collected in December of 2006 from<br />

the same populations from which leaf materials were sampled<br />

(l " Table 1). The remaining wild materials were collected in November<br />

of 2009. All the sampled roots were gently washed and<br />

dried at 50°C followed by pulverization and 40-mesh sieving.


HPLC fingerprinting<br />

An accurately weighed root powder (1.0 g) was ultrasonically extracted<br />

with 50 mL 70% methanol for 30 min, and the extract was<br />

diluted to a volume of 50 mL following paper-filtration. The solutions<br />

were filtered through a 0.45 µm membrane prior to HPLC<br />

analysis. Chromatograms were generated on an Agilent 1100 series<br />

HPLC system equipped with a 5 µm HC‑C18 column<br />

(250 mm × 4.6 mm) using a gradient of acetonitrile (A) and 1%<br />

aqueous acetic acid (B). The gradient program was performed as<br />

follows: 0–5min, 15% A; 5–30 min, 15–50% A; 30–50 min, 50–<br />

65% A; 50–60 min, 65% A, with a flow rate of 0.8 mL ·min −1 , column<br />

temperature 35°C, and a detection wavelength of 280 nm.<br />

One analyte was randomly selected for 5 consecutive analyses to<br />

validate precision. Stability examination was accomplished with<br />

the same analyte in 48 h (n = 5). Peak area variation was estimated<br />

in terms of relative standard deviation (RSD).<br />

Identification of four main compounds<br />

in chromatograms<br />

The four main bioactive compounds of Radix Scrophulariae, including<br />

harpagoside, angroside C, acetoeside, and cinnamic acid,<br />

were identified in terms of retention time and mass spectrum by<br />

comparison with reference compounds. The ESI MS n spectra were<br />

acquired in both positive and negative ion modes in Thermo Finnigan<br />

LCQ DECA XP system equipped with an electrospray ionization<br />

source (Thermo LC/MS Division). The mass spectrometry detector<br />

(MSD) parameters were as follows: nebulizer sheath gas,<br />

N 2 (80 uit); nebulizer auxiliary gas, N2 (20 uit); capillary temperature,<br />

350 °C; spray voltage, 4500 V in negative ion ESI mode,<br />

5000 V in positive ion ESI mode; capillary voltage, − 13 V in (−)<br />

ESI, 25 V in (+) ESI; lens voltage, 18 V in (−) ESI, − 16 V in (+) ESI;<br />

isolation width for the MS n experiments, 1.0 m/z; collision gas,<br />

He; and collision energy, 35%.<br />

HPLC fingerprinting data analyses<br />

Characteristic peaks extracted from chromatogram data in Agilent<br />

Chemstation software were used as variables for principal<br />

component analysis (PCA). Difference significance of bioactive<br />

compound contents was tested using one-way ANOVA or independent<br />

t-test. The program SPSS 13.0 was employed for both<br />

analyses.<br />

DNA extraction and ISSR amplification<br />

Total genomic DNA was extracted using the modified CTAB<br />

method [20]. ISSR‑PCR amplifications were performed in a PTC-<br />

200 thermal cycler (MJ Research) programmed for an initial 5min<br />

denaturation at 94°C, followed by 45 cycles of 1 min denaturation<br />

at 94°C, 45 s annealing at 49.4–65°C (depending on different<br />

primers), and 1.5 min elongation at 72°C, as well as a final<br />

elongation step of 10 min at 72°C. Twelve primers (UBC primer<br />

set no. 9, Biotechnology Laboratory, University of British Columbia)<br />

were screened for fingerprinting all 100 individuals. The following<br />

primers were used (annealing temperature in parentheses):<br />

UBC809 (60.5 °C), UBC810 (53 °C), UBC811 (52.7 °C),<br />

UBC812 (50.8 °C), UBC827 (60.5 °C), UBC834 (49.4 °C), UBC855<br />

(62 °C), UBC859 (57 °C), UBC874 (65 °C), UBC881 (60.5 °C),<br />

UBC887 (50.8 °C), and UBC889 (50.8 °C).<br />

ISSR data analyses<br />

The following parameters of genetic diversity were calculated using<br />

POPGENE version 1.31 [21]: (i) the percentage of polymorphic<br />

fragments (PPF); (ii) Neiʼs expected heterozygosity (HPOP); and<br />

Original Papers<br />

(iii) Neiʼs coefficient of population differentiation (GST). Considering<br />

a possible bias of HPOP resulting from the assumption of the<br />

Hardy-Weinberg equilibrium, Bayesian gene diversity (HB) was<br />

also computed using HICKORY version 1.0 [22]. A dendrogram of<br />

the unweighted pair-group method with arithmetic means<br />

(UPGMA) was generated by TFPGA version 1.3 [23], and bootstrap<br />

values were computed by resampling with replacement over loci<br />

(1000 replicates). Hierarchical structuring of genetic variation<br />

and pairwise Φ ST distances among populations were also determined<br />

by an analysis of molecular variance (AMOVA) with<br />

WINAMOVA version 1.55 [24]. Significance levels of the variance<br />

components were based on 1000 permutations. Furthermore, an<br />

estimator of F ST under a free model of population sampling, θB,<br />

and GST‑B, a Bayesian analogue of Neiʼs GST, was produced using<br />

HICKORY version 1.0. Principal coordinates analysis (PCoA) was<br />

executed using MVSP version 1.3 [25].<br />

Supporting information<br />

Information on the peak areas of all analyzed samples, the chromatogram<br />

of wild and cultivated Radix Scrophulariae, the PCA<br />

analysis of Radix Scrophulariae and the PCoA analysis of ISSR<br />

phenotypes are available as Supporting Information.<br />

Results<br />

!<br />

HPLC validation presented satisfactory precision, reproducibility,<br />

stability, and recovery with RSD of peak areas less than 5%. In total,<br />

29 characteristic peaks were extracted as fingerprint markers<br />

from all 34 analyzed samples of Radix Scrophulariae (refer to<br />

Fig. 1S for representative chromatograms in Supporting Information).<br />

Harpagoside, angroside C, acteoside, and cinnamic acid<br />

were characterized as the four predominant common peaks.<br />

Eight peaks including the four compounds mentioned above<br />

were shared by all samples (monomorphic) with varying intensities,<br />

while the other 21 compounds were polymorphic<br />

(72.41%). Most wild samples exhibited two- to three-fold greater<br />

intensities of chromatographic peaks than cultivated samples<br />

(see Table 1S in Supporting Information). Compared to wild Radix<br />

Scrophulariae, slighter differences of peak amount and peak<br />

area existed to different extents among cultivated ones.<br />

In the PCA scatter plot of all samples generated by 29 peak areas,<br />

cultivated populations approximately fell into a big cluster, while<br />

the wild populations appeared more scattered, especially the distant<br />

TW population. However, wild populations of JX, TM (except<br />

one sample), and HN distributed considerably close to the cluster<br />

of cultivated populations (l " Fig. 2 a). Further PCA excluding wild<br />

samples produced four clustered groups in coincidence with cultivated<br />

populations (l " Fig. 2 b). We ran additional PCAs using the<br />

four main bioactive compounds as variables, and the projections<br />

showed similar trends (see Fig. 2S in Supporting Information), indicating<br />

their potency as markers for quality assessment of Radix<br />

Scrophulariae. The mean contents of the two compounds, angroside<br />

C and harpagoside, of wild populations were significantly<br />

higher than those of cultivated ones (l " Table 2). Generally, one<br />

of the wild populations, TW, produced the highest contents of<br />

bioactive compounds. There were also significant differences of<br />

angroside C and cinnamic acid contents among cultivated populations.<br />

Besides these two compounds, the wild populations<br />

showed further difference of acetoside content, implying more<br />

chemical variation among wild populations.<br />

Yang S et al. Association between Chemical… <strong>Planta</strong> Med 2011; 77: 865–871<br />

867


868<br />

Original Papers<br />

Yang S et al. Association between Chemical … <strong>Planta</strong> Med 2011; 77: 865–871<br />

Fig. 2 a Scatter plot of principal component analysis<br />

(PCA) for all samples of Radix Scrophulariae.<br />

PC1 and PC2 are the first principal components using<br />

29 chromatographic peak areas as variables.<br />

Fig. 2 b Scatter plot of principal component analysis<br />

(PCA) for the cultivated samples of Radix<br />

Scrophulariae. PC1 and PC2 are the first principal<br />

components using 29 chromatographic peak areas<br />

as variables.


Table 2 Content comparison of the four bioactive compounds among different populations of S. ningpoensis (µg/mg).<br />

Population<br />

Cultivated populations<br />

Acteoside Angroside C Harpagoside Cinammic acid<br />

DP 0.104 ± 0.002c 0.152 ± 0.017cd 1.823 ± 0.613cde 0.596 ± 0.127bc YC 0.122 ± 0.033bc 0.138 ± 0.020cd 1.557 ± 0.495de 0.702 ± 0.097b RC 0.086 ± 0.029c 0.138 ± 0.019cd 1.465 ± 0.440de 0.445 ± 0.109bcd HB 0.064 ± 0.017c 0.127 ± 0.022d 0.685 ± 0.389e 0.310 ± 0.073cd SX 0.063 ± 0.008c 0.251 ± 0.055b 2.236 ± 0.822cde 0.289 ± 0.040d CQ 0.059 ± 0.008c 0.159 ± 0.025bcd 1.521 ± 0.280de 0.287 ± 0.016d Mean<br />

Wild populations<br />

0.083 ± 0.029 0.161 ± 0.049 1.548 ± 0.657 0.438 ± 0.181<br />

TM 0.178 ± 0.081ab 0.369 ± 0.128a 5.732 ± 3.739b 0.475 ± 0.104bcd TW 0.226 ± 0.061a 0.418 ± 0.049a 3.506 ± 1.119bcd 1.918 ± 0.417a JX 0.077 ± 0.020c 0.227 ± 0.062bc 4.077 ± 0.632bc 0.272 ± 0.036d HN 0.071 ± 0.004c 0.195 ± 0.030bcd 5.369 ± 1.253b 0.361 ± 0.145cd AH 0.069 ± 0.028c 0.233 ± 0.027bc 11.434 ± 0.959a 0.204 ± 0.141d Mean 0.121 ± 0.078 0.285 ± 0.106 6.362 ± 3.506 0.618 ± 0.676<br />

Lowercases represent significant differences at the level of 95% revealed by post hoc multiple comparison<br />

The survey of 100 individuals from 8 populations of S. ningpoensis<br />

with 12 ISSR primers produced a total of 124 reproducible<br />

fragments, of which 108 were polymorphic (87.10%). The highest<br />

genetic diversity was observed in the wild population TM, with<br />

PPF, HPOP, and HB up to 48.39%, 0.166, and 0.208, respectively. In<br />

contrast, the level of genetic diversity was lowest in the cultivated<br />

population DP (PPF = 12.90%, HPOP = 0.052, HB = 0.073).<br />

Considering groups of populations, those from the wild populations<br />

(TM and TW) possessed on average higher levels of diversity<br />

(PPF = 41.94%, H POP = 0.150, HB = 0.186) than those from cultivated<br />

populations (PPF = 16.80%, HPOP = 0.064, HB = 0.80) (l " Table 3).<br />

Neiʼs estimator of population substructure (GST) indicated a<br />

strong differentiation among cultivated populations (GST =<br />

0.706) while a greatly lower value among wild ones (GST =<br />

0.289). The analysis of molecular variance (AMOVA) produced a<br />

parallel result of population differentiation within both groups<br />

(Φ ST = 0.801 vs. ΦST = 0.399) (l " Table 4). As the hierarchical estimates<br />

of variance components displayed, the variance component<br />

was large and significant among groups (48.04%, p < 0.001;<br />

l " Table 4). The Bayesian estimate of FST (θB) and Neiʼs GST (GST‑B)<br />

indicated a similar result: cultivated populations harbored higher<br />

levels of variance distributed among populations compared to<br />

wild populations (l " Table 4).<br />

UPGMA clustering based on the genetic distance matrix resulted<br />

in two clades in the studied populations of S. ningpoensis, cultivated<br />

and wild clades (l " Fig. 3). In the cultivated clade, population<br />

CQ was clearly separated from the other cultivated populations,<br />

while all populations from Zhejiang (DP, RC, and YC) composed<br />

a close clade parallel to HB and SX populations. PCoA indicated<br />

four main plots approximately consistent with UPGMA<br />

clustering (see Fig. 3S in Supporting Information).<br />

Discussion<br />

!<br />

In many cases, chemical diversity has been largely attributed to a<br />

variable environment. However, recent work demonstrated that<br />

chemical diversity was correlated with genetic variation in Anemopsis<br />

californica [26, 27], Pinus ponderosa [28], and Vitex rotundifolia<br />

[29]. In our present research, PCA (l " Fig. 2) and UPGMA<br />

(l " Fig. 3) showed that chemical and genetic differentiation<br />

among S. ningpoensis populations demonstrated nearly identical<br />

Original Papers<br />

Table 3 Genetic diversity indices of the ten populations of S. ningpoensis.<br />

Population<br />

Cultivated populations<br />

PPF (%) HPOP HB DP 12.90 0.052 0.073<br />

YC 20.97 0.086 0.094<br />

RC 13.71 0.042 0.058<br />

HB 22.58 0.085 0.103<br />

SX 16.94 0.068 0.092<br />

CQ 13.71 0.048 0.060<br />

Mean<br />

Wild populations<br />

16.80 ± 4.13 0.064 ± 0.019 0.080 ± 0.019<br />

TM 48.39 0.166 0.208<br />

TW 35.48 0.134 0.164<br />

Mean 41.94 ± 9.13 0.150 ± 0.023 0.186 ± 0.031<br />

PPF, percentage of polymorphic fragments; HPOP, Neiʼsexpected heterozygosity; HB, expected Bayesian heterozygosity<br />

patterns. Both chemical and genetic diversity of wild populations<br />

were significantly higher than that of cultivated ones (l " Table 2;<br />

l " Table 3; Table 1S in Supporting Information). Regarding the<br />

currently unanalyzed wild populations of S. ningpoensis, our further<br />

ITS-based maximum parsimony (MP) tree revealed a substantial<br />

differentiation within population TM with one clade parallel<br />

to clade AH and another relatively remote clade close to cultivated<br />

populations. Populations JX and HN considerably resembled<br />

all the cultivated populations in the ITS tree (C. Chen et<br />

al., unpublished manuscript). All the above genetic relationships<br />

indicated by the ITS tree unexpectedly coincided with the chemical<br />

differentiation represented in l " Fig. 2. The correlation between<br />

chemical and genetic variation was further supported by<br />

the detection of a specific chloroplast DNA haplotype in population<br />

TW, which is phytochemically distinct, and by the existence<br />

of a shared haplotype between most cultivated populations and a<br />

wild population, JX, which is chemically the closest to the cultivated<br />

ones (C. Chen et al., unpublished manuscript). Therefore,<br />

chemical variation was strongly indicated to be associated with<br />

genetic variation in S. ningpoensis.<br />

Compared to cultivated populations, wild populations of S. ningpoensis<br />

produced a couple of unique chromatographic peaks as<br />

well as much higher contents of most metabolites, especially in<br />

Yang S et al. Association between Chemical… <strong>Planta</strong> Med 2011; 77: 865–871<br />

869


870<br />

Original Papers<br />

Table 4 Analysis of molecular variance (AMOVA) and Bayesian F ST estimators for ISSR data of wild and cultivated S. ningpoensis.<br />

AMOVA Bayesian estimators<br />

Source of variation<br />

Wild populations<br />

d.f. Variance percent Φ-statistic P value θB ±SD GST‑B ±SD<br />

Variation among populations 1 39.92% ΦST = 0.399 < 0.001 0.397 ± 0.041 0.236 ± 0.021<br />

Variation within populations<br />

Cultivated populations<br />

18 60.08%<br />

Variation among populations 5 80.13% ΦST = 0.801 < 0.001 0.720 ± 0.018 0.691 ± 0.016<br />

Variation within populations<br />

Hierarchical analysis for all studied populations<br />

70 19.87%<br />

Among groups of populations 1 48.04% ΦCT = 0.480 < 0.001<br />

Among populations within the groups 6 36.80%<br />

Within populations 88 15.16%<br />

ΦST: inter-population genetic differentiation; ΦCT: genetic differentiation among groups; θB: FST analogues under a free model of population sampling; GST‑B: Bayesian analogue of<br />

Neiʼs GST; d. f.: degree of freedom<br />

terms of angroside C and harpagoside (l " Table 2; Fig. 1S and<br />

Table 1S in Supporting Information). Secondary metabolites<br />

function in response to stresses and changing environments<br />

[1]. High genetic diversity was proposed to benefit the longterm<br />

persistence of plant species through increasing their<br />

adaptivity to ever changing environments [30]. The wild populations<br />

of S. ningpoensis (like population TW), harboring a much<br />

higher level of genetic and chemical variation, would be highly<br />

valuable for breeding programs promoting metabolite production<br />

and stress resistance. Considering that variation was largely apportioned<br />

within populations (60.08%, see l " Table 4), a sampling<br />

strategy using more individuals from one population was suggested<br />

for both breeding program and germplasm conservation.<br />

In contrast, the six cultivated populations of S. ningpoensis exhibited<br />

a remarkably low level of average ISSR diversity within populations<br />

(l " Table 3) and high differentiation (l " Table 4; l " Fig. 3).<br />

These results were consistent with the previous report of S. ningpoensis<br />

[18] as well as many other crop plants [31, 32]. This could<br />

be explained by the long-term artificial selection, the mode of<br />

clonal propagation, and limited among-population exchange of<br />

genetic materials in cultivated populations of S. ningpoensis. The<br />

cultivated populations with remarkable genetic differentiation<br />

(landraces) displaying a high among-population portion of genetic<br />

variation (80.13%, see l " Table 4) should be included for ex situ<br />

Yang S et al. Association between Chemical … <strong>Planta</strong> Med 2011; 77: 865–871<br />

Fig. 3 UPGMA dendrogram illustrating the genetic<br />

relationships among eight populations of S. ningpoensis.<br />

Numbers on branches indicate bootstrap<br />

values from 1000 replicates.<br />

conservation. Although clonally propagated in cultivated populations,<br />

S. ningpoensis reproduces sexually in wild populations.<br />

Therefore, to maintain and improve genetic diversity, sexual<br />

propagation should be encouraged to a certain extent within<br />

and among cultivated populations, even between cultivated and<br />

wild populations of S. ningpoensis.<br />

The following conclusions can be drawn from the present results.<br />

(i) There is a strong association between chemical and genetic<br />

variation of wild and cultivated populations of S. ningpoensis. (ii)<br />

The wild and genetically differentiated cultivated populations<br />

call for priority to in situ or ex situ conservation and utilization.<br />

(iii) Sexual propagation should be partly carried out in practice<br />

to maintain and improve genetic diversity. The present research<br />

considerably promotes our theoretical understanding of medicinal<br />

plant species regarding the selective and adaptive evolutionary<br />

processes under the influence of both domestication and<br />

their habitats. Our findings are also beneficial to genetic conservation,<br />

crop improvement, and quality control of S. ningpoensis.


Acknowledgements<br />

!<br />

This work was financially supported by the National Basic Research<br />

Program of China (Grant No. 2007CB411600), the National<br />

Science & Technology Pillar Program during the Eleventh Five-<br />

Year Plan Period (Grant No. 2006BAI21B07), Zhejiang Provincial<br />

Natural Science Foundation of China (Grant No. Y3080087), and<br />

the Fundamental Research Funds for the Central Universities.<br />

The authors are grateful to Pan Li for collecting part of wild populations,<br />

the local producers for their kindness of facilitating<br />

sampling, and Xinhang Jiang and Dr. Zhican Wang for LC‑MS<br />

analysis. We also thank Dr. Yingxiong Qiu for his assistance in genetic<br />

data analyses and crucial suggestions and Dr. Jimmy Triplette<br />

for manuscript improvement.<br />

References<br />

1 Fernie AR. The future of metabolic phytochemistry: larger numbers of<br />

metabolites, higher resolution, greater understanding. Phytochemistry<br />

2007; 68: 2861–2880<br />

2 Hartmann T. From waste products to ecochemicals: fifty years research<br />

of plant secondary metabolism. Phytochemistry 2007; 68: 2831–2846<br />

3 Hoisington D, Khairallah M, Reeves T, Ribaut JM, Skovmand B, Taba S,<br />

Warburton M. Plant genetic resources: what can they contribute toward<br />

increased crop productivity? Proc Natl Acad Sci USA 1999; 96:<br />

5937–5943<br />

4 Fernie AR, Tadmor Y, Zamir D. Natural genetic variation for improving<br />

crop quality. Curr Opin Plant Biol 2006; 9: 196–202<br />

5 Diaz AM, Abad MJ, Fernandez L, Silvan AM, De Santos J, Bermejo P. Phenylpropanoid<br />

glycosides from Scrophularia scorodonia: in vitro anti-inflammatory<br />

activity. Life Sci 2004; 74: 2515–2526<br />

6 Galindez JD, Lanza AMD, Matellano LF. Biologically active substances<br />

from the genus Scrophularia. Pharm Biol 2002; 40: 45–59<br />

7 Garcia D, Fernandez A, Saenz T, Ahumada C. Antiinflammatory effects of<br />

different extracts and harpagoside isolated from Scrophularia frutescens<br />

L. Farmaco 1996; 51: 443–446<br />

8 Liu L, Hudgins WR, Shack S, Yin MQ, Samid D. Cinnamic acid: a natural<br />

product with potential use in cancer intervention. Int J Cancer 1995;<br />

65: 345–350<br />

9 Miyazawa M, Okuno Y, Nakamura S, Kameoka H. Suppression of SOSinducing<br />

activity of chemical mutagens by cinnamic acid derivatives<br />

from Scrophulia ningpoensis in the Salmonella typhimurium TA1535/<br />

pSK1002 umu Test. J Agric Food Chem 1998; 46: 904–910<br />

10 Zhang HX, Song JY, Zhao SR, Zhao HQ, Gong KM. Simultaneous determination<br />

of the contents of the cinnamic acid and harpagoside in Radix<br />

Scrophulariae by RP-HPLC. J Shenyang Pharm Univ 2006; 23: 507–510<br />

11 Liu CW, Bi ZM, Zhu YF, Li P. Simultaneous determination of four kinds of<br />

bioactive components in Radix Scrophulariae by HPLC. China Pharm<br />

J 2007; 42: 1614–1616<br />

12 Li P, Zhang Y, Xiao L, Jin X, Yang K. Simultaneous determination of harpagoside<br />

and cinnamic acid in rat plasma by high-performance liquid<br />

chromatography: application to a pharmacokinetic study. Anal Bioanal<br />

Chem 2007; 389: 2259–2264<br />

13 Wang SJ, Ruan JX, Zhao YH. Simultaneous determination of harpagoside<br />

and cinnamic acid in rat plasma by liquid chromatography electrospray<br />

ionization mass spectrometry and its application to pharmacokinetic<br />

studies. Biomed Chromatogr 2008; 22: 50–57<br />

Original Papers<br />

14 Zhu YF, Bi ZM, Liu CW, Ren MT, Wu FH, Li P. Endothelial cell extraction<br />

and HPLC‑ESI/TOF MS analysis for predicting potential bioactive components<br />

of Radix Scrophulariae. J China Pharm Univ 2008; 39: 228–<br />

231<br />

15 Bai ZC. Fingerprints of Radix Scrophulariae by HPLC. Chin Mater Med<br />

2006; 29: 1295–1299<br />

16 Ma DF, Zhou N, Bai ZC, Fang LJ, Sun CJ. Identification of the habitat of<br />

Radix Scrophulariae with chromatographic fingerprinting. Lishizhen<br />

Med Mater Med Res 2009; 20: 789–793<br />

17 Zhao ZX, Liang ZS, Jiang ZM, Xue YF, Yang YZ. Establishment and optimization<br />

of ISSR reaction system for Scrophularia ningpoensis Hemsl.<br />

Pharm Biotechnol 2007; 14: 318–323<br />

18 Zhao ZX. The study on germplast resources diversity of Scrophularian<br />

ningpoensis Hemsl. [dissertation]. Yangling: Northwest Agricultural<br />

Forestry University; 2008<br />

19 Chen DX, Li LY, Peng R, Wu Y, Cai Y. Analysis of genetic difference among<br />

Scrophularia ningpoensis cultivars by SRAP. China J Chin Mater Med<br />

2009; 34: 138–142<br />

20 Doyle JJ. DNA protocols for plants – CTAB total DNA isolation. In: Hewitt<br />

GM, Johnston A, editors. Molecular techniques in taxonomy. Heidelberg:<br />

Springer-Verlag; 1991: 283–293<br />

21 Yeh FC, Yang RC, Boyle TB, Ye ZH, Mao JX. POPGENE, the user-friendly<br />

shareware for population genetic analysis. Available at http://www.<br />

ualberta.ca/~fyeh. Accessed October 3, 2009<br />

22 Holsinger KE, Lewis PO. Hickory: a package for analysis of population<br />

genetic data V1.0. Available at http://www.eeb.uconn.edu. Accessed<br />

October 3, 2009<br />

23 Miller MP. AMOVA-PREP 1.01: A program for the preparation of AMO-<br />

VA input files from dominant-markers raw data. Flagstaff: Department<br />

of Biological Sciences, Northern Arizona University; 1998<br />

24 Excoffier L, Smouse PE, Quattro JM. Analysis of molecular variance inferred<br />

from metric distances among DNA haplotypes: applications to<br />

human mitochondrial DNA restriction data. Genetics 1992; 131: 479–<br />

491<br />

25 Kovach WL. MVSP – A multivariate statistical package for windows,<br />

version 3.1. Wales: Kovach Computing Services; 1999<br />

26 Medina-Holguin AL, Holguin FO, Micheletto S, Goehle S, Simon JA, OʼConnell<br />

MA. Chemotypic variation of essential oils in the medicinal plant,<br />

Anemopsis californica. Phytochemistry 2008; 69: 919–927<br />

27 Medina-Holguin AL, Martin C, Micheletto S, Holguín FO, Rodriguez J,<br />

OʼConnel MA. Environmental influences on essential oils in roots of<br />

Anemopsis californica. Hortscience 2007; 42: 1578–1583<br />

28 Gerson EA, Kelsy RG, Clair JB. Genetic variation of piperidine alkaloids in<br />

Pinus ponderosa: a common garden study. Ann Bot 2009; 103: 447–<br />

457<br />

29 Hu Y, Zhang QY, Xin HL, Qin LP, Lu BR, Rahman K, Zheng H. Association<br />

between chemical and genetic variation of Vitex rotundifolia populations<br />

from different locations in China: its implication for quality control<br />

of medicinal plants. Biomed Chromatogr 2007; 21: 967–975<br />

30 Vrijenhoek RC. Animal population genetics and disturbance: the effects<br />

of local extinctions and recolonizations on heterozygosity and fitness.<br />

In: Pickett STA, White PS, editors. The ecology of natural disturbance<br />

and path dynamics. London, MA: Academic Press; 1985: 265–285<br />

31 Abbo S, Berger J, Turner NC. Evolution of cultivated chickpea: four bottlenecks<br />

limit diversity and constrain adaptation. Funct Plant Biol<br />

2003; 30: 1081–1087<br />

32 Doebley J. Isozymic evidence and the evolution of crop plants. In: Soltis<br />

DE, Soltis PS, editors. Isozymes in plant biology. Portland: Dioscorides<br />

Press; 1989: 165–191<br />

Yang S et al. Association between Chemical… <strong>Planta</strong> Med 2011; 77: 865–871<br />

871

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

Saved successfully!

Ooh no, something went wrong!