<<

Hindawi Publishing Corporation Evidence-Based Complementary and Alternative Medicine Volume 2013, Article ID 340215, 51 pages http://dx.doi.org/10.1155/2013/340215

Review Article Bidens pilosa L. (Asteraceae): Botanical Properties, Traditional Uses, Phytochemistry, and Pharmacology

Arlene P. Bartolome,1,2 Irene M. Villaseñor,1 and Wen-Chin Yang2,3,4,5,6

1 Institute of Chemistry, University of the Philippines, Diliman, Quezon City, Philippines 2 Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan 3 Institute of Pharmacology, Yang-Ming University, Taipei 112, Taiwan 4 Department of Life Sciences, National Chung-Hsing University, Taichung 402, Taiwan 5 Institute of Zoology, National Taiwan University, Taipei 106, Taiwan 6 Agricultural Biotechnology Research Center, Academia Sinica, No. 128, Academia Sinica Road, Section 2, Nankang, Taipei 11529, Taiwan

Correspondence should be addressed to Wen-Chin Yang; [email protected]

Received 31 January 2013; Accepted 29 April 2013

AcademicEditor:GailB.Mahady

Copyright © 2013 Arlene P. Bartolome et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

There are 230 to 240 known Bidens species. Among them, Bidens pilosa is a representative perennial herb, globally distributed across temperate and tropical regions. B. pilosa has been traditionally used in foods and medicines without obvious adverse effects. Despite significant progress in phytochemical and biological analyses of B. pilosa over the past few years, comprehensive and critical reviews of this plant are anachronistic or relatively limited in scope. The present review aims to summarize up-to-date information on the phytochemistry, pharmacology, and toxicology of B. pilosa from the literature. In addition to botanical studies and records of thetraditionaluseofB. pilosa in over 40 diseases, scientific studies investigating the potential medicinal uses of this species and its constituent phytochemicals for a variety of disorders are presented and discussed. The structure, bioactivity, and likely mechanisms of action of B. pilosa anditsphytochemicalsareemphasized.Althoughsomeprogresshasbeenmade,furtherrigorouseffortsare required to investigate the individual compounds isolated from B. pilosa to understand and validate its traditional uses and develop clinical applications. The present review provides preliminary information and gives guidance for further basic and clinical research into this plant.

1. Introduction systems, Chinese herbal medicine, and Ayurvedic medicine, respectively, that continue to be widely practiced today [2]. The United Nations World Health Organization estimates Therefore, since antiquity, medicinal herbs have played a that as many as 5.6 billion people, 80% of the world popu- prominentroleinhumanhealth. 1 lation, utilize herbal medicine for primary health care [1]. B. pilosa is an easy-to-grow herb that is widely distributed Plants have formed the foundation of complicated traditional all over the world. It is considered to be a rich source of medicine systems for thousands of years. Medicinal herbs are food and medicine for humans and animals [3, 4]. There is appliedtotreatawiderangeofdiseasecategories.Thefirst increasing global interest in the use of B. pilosa as shown written documentation of the use of medicinal herbs dates bythemanystudiesconductedontheplantinrecentyears. from the 26th century BCE in Mesopotamia, and the first The folkloric use of B. pilosa has been recorded in America, record of the use of medicinal herbs by the Egyptians and Africa, Asia, and Oceania [5]. To explore the potential clinical Greeks dates from 18th century BCE and the 5th century application of B. pilosa, it is important to link its traditional BCE, respectively. Starting around the 11th century BCE, use with rigorous evidence-based scientific study. The present the Chinese and Indians started to develop herbal medicine review focuses on recent studies on the botany, traditional 2 Evidence-Based Complementary and Alternative Medicine

Table 1: Taxonomy of B. pilosa [6]. (“all bountiful grass”) in Chinese because of their prosperous growth. Kingdom Plantae Subkingdom Tracheobionta Superdivision Spermatophyta Division Magnoliophyta 1.2. Traditional Uses. B. pilosa is used as an herb and as an ingredient in teas or herbal medicines. Its shoots and Class Magnoliopsida leaves, dried or fresh, are utilized in sauces and teas [14, Subclass Asteridae 15].Inthe1970s,theUnitedNationsFoodandAgriculture Order Asterales Organization (FAO) promoted the cultivation of B. pilosa in Family Asteraceae Africa because it is easy to grow, edible, palatable, and safe Genus Bidens [7]. The nutritional value of B. pilosa is shown in Table 2. Species Bidens Pilosa L. All parts of B. pilosa plant, the whole plant, the aerial parts (leaves, flowers, seeds, and stems), and/or the roots, fresh or dried, are used as ingredients in folk medicines. It is usage, phytochemistry, pharmacology, and toxicology of B. frequently prepared as a dry powder, decoction, maceration pilosa. The information provided here highlights the pos- or tincture [16]. Generally, this plant is applied as dry sible usefulness of B. pilosa and its isolated compounds powder or tincture when used externally, and as a powder, and offers insights into possible future research directions. maceration, or decoction when used as an internal remedy Studies of B. pilosa are divided into three groups: (1) the [15]. botany, ethnomedical uses, plant chemistry, pharmacology, As summarized in Table 3, B. pilosa,eitherasawhole and biosafety of B. pilosa; (2) scientific studies that validate plant or different parts, has been reported to be useful in the ethnomedical uses of B. pilosa; and (3) the therapeutic and the treatment of more than 40 disorders such as inflamma- future research potential of B. pilosa. tion, immunological disorders, digestive disorders, infectious diseases, cancers, metabolic syndrome, wounds, and many others [26–29]. B. pilosa is usually ingested; however, it can 1.1. Botany. B. pilosa was first collected and named by Carl also be utilized externally. For instance, fresh B. pilosa is used Linnaeus in 1753 [3]. Taxonomically, it is assigned to the to treat snake bites and wounds [23], and in Trinidad and Bidens genus(Asteraceae)asshowninTable1.Thisgenusis Tobago the aqueous solution of the leaves of B. pilosa is used estimated to include 230 to 240 species worldwide [3, 4]. B. to bathe babies and children [22]. pilosa has several varieties such as B. pilosa var. radiata,var. B. pilosa is sometimes used alone; it is also used as minor,var.pilosa, and var. bisetosa. Alongside examination of an ingredient in medicinal mixtures together with other morphological traits, authentication of B. pilosa can be aided medicinal plants such as Aloe vera, Plectranthus mollis, by chemotaxonomy and molecular characterization [8]. Valeriana officinalis, and Cissus sicyoides among others [17, 18, B. pilosa is an erect, perennial herb widely distributed 20, 25]. Whether the commonly used mixtures of B. pilosa, across temperate and tropical regions. B. pilosa is either or the mixtures of the compounds found in B. pilosa afford glabrous or hairy, with green opposite leaves that are serrate, synergistic effects is not yet clear and needs to be verified lobed, or dissected. It has white or yellow flowers, and long by further studies. However, one study has suggested that narrow ribbed black achenes (seeds). It grows to an average B. pilosa varieties share similar phytochemical compositions height of 60 cm and a maximum of 150 cm in favorable and may be substituted for each other [8]. environments [9](Figure1). B. pilosa prefers full sun and moderately dry soil. However, it can grow in arid and barren land from low to high elevations. With the advantage of being fast-growing, in the 1970s, the Food and Agricultural 2. Phytochemicals Organization actively promoted the cultivation of B. pilosa in Africa [10]. B. pilosa propagates via seeds. A single plant Interest in basic research and application of B. pilosa has can produce 3000–6000 seeds. Dry mature seeds from B. increased since its first identification in 1753. This is mainly pilosa canbegerminatedin3to4daysinmoistsoilor due to its wide application in medicines, foods, and drinks. after being soaking in water. Seeds are viable for at least 3 Around 116 publications have documented the exploitation years [11]. Minimal agricultural techniques are required for andmedicaluseofB. pilosa.Todate,201compounds B. pilosa cultivation. Due to its invasive tendencies, B. pilosa comprising 70 aliphatics, 60 flavonoids, 25 terpenoids, 19 is generally considered to be a weed [12]. phenylpropanoids, 13 aromatics, 8 porphyrins, and 6 other B. pilosa is thought to have originated in South America compounds, have been identified from this plant as compiled andsubsequentlyspreadallovertheworld[13]. Bidens previously [30]. The structures of these compounds are pre- species and their varieties bear vernacular names based on sented in Tables 4, 5, 6, 7, 8, 9,and10,respectively.However, their characteristics. For example, Bidens species are known the association between B. pilosa phytochemicals and their by such names as Spanish needles, beggar’s ticks, devil’s bioactivities is not yet fully established and should become needles, cobbler’s pegs, broom stick, pitchforks, and farmers’ a future research focus. In the present review, we explore friendsinEnglishandsomeotherlanguagesbecauseoftheir possible associations (Table 11), describe the importance of sticky achenes [9] and are sometimes known as xian feng cao the known compounds in relation to their biological activity Evidence-Based Complementary and Alternative Medicine 3

Table 2: Nutritional facts about B. pilosa, courtesy of the United Nations Food and Agriculture Organization [7].

Carotene Plant Energy Moisture Protein Fat Carbohydrate Fiber Ash Calcium Phosphorus Iron Thiamine equivalent (100 g) (kcal) (%) (g) (g) (g) (g) (g) (mg) (mg) (𝜇g) (mg) (𝜇g) Raw 43 85.1 3.8 0.5 8.4 3.9 2.2 340 67 — 1 800 — Dried 33 88.6 2.8 0.6 6 1.3 2 111 39 2.3 — — “—” denotes not detectable.

(a) (b)

(c)

Figure 1: B. pilosa (a)anditsflowers(b)andachenes(c). and discuss their likely mechanisms of action (Table 3). Com- 3. Pharmacological Properties pelling evidence suggests that the various diverse bioactivities reported for B. pilosa reflect its phytochemical complexity. As outlined in Table 3, B. pilosa is traditionally used to B. pilosa is an extraordinary source of phytochemicals, treat a wide variety of ailments. Different preparations of particularly flavonoids and polyynes. Plant flavonoids are its whole plant and/or parts have been purported to treat commonly reported to possess anticancer, antiinflammatory, over 40 categories of illnesses. Scientific studies, although not antioxidant, and other bioactivities. However, the bioactivi- extensive, have demonstrated that B. pilosa extracts and/or ties of only seven of the 60 flavonoids present in B. pilosa have compounds have antitumor [43, 69, 77–83], antiinflamma- been studied. The bioactivities of the remaining 53 flavonoids tory [27, 75, 79, 84–88], antidiabetic and antihyperglycemic are poorly understood and deserve further investigation. [8, 24, 49, 51, 53], antioxidant [70, 89, 90], immunomodu- Other classes of compounds found in B. pilosa arealsoinneed latory [50, 69], antimalarial [47, 77], antibacterial [47, 91, of further examination, as shown in Table 11. 92], antifungal [92, 93], antihypertensive, vasodilatory [28, 94], and antiulcerative [26] activities. In this section, the 4 Evidence-Based Complementary and Alternative Medicine

Table 3: Ethnomedical information about B. pilosa.

Disorder Plant partb Mode of use Region/country References India Stomachache LE Decoction [17–19] Africa Africa Colics WP Decoction [18] China Juice or decoction; Catarrh WP Cuba [20] taken orally LE and Fresh leaves or Africa Diarrhea [18, 21] WP Decoction Uganda Constipation WP Decoction Africa [18] Dysentery/Bacillary Africa WP Decoction [18] dysentery China Choleretic WP Decoction Middle America [18] China Anti-inflammatory WP Not stated [20, 22] Cuba Decoction or Cuba Asthma WP maceration; taken [20, 23] China orally RT and Hong Kong Antirheumatic Juice and decoction [18, 22] WP Zulu, Africa Acute appendicitis WP Decoction Hong Kong [23] Africa Enteritis WP Decoction [18] China Pruritus WP Decoction Hong Kong [23] Africa Conjunctivitis WP Decoction [18] China Africa Otitis WP Decoction [18] China Africa Pharyngitis WP Decoction [18] China Gastritis WP Juice; taken orally Cuba [20] Decoction; taken Cuba Diabetes WP [20, 24] orally Taiwan Headache WP Decoction Bafia, Cameroon [25] Diuretic WP Decoction Middle America [18] Decoction; taken Hypotensive WP Bafia. Cameroon25 [ ] orally China LE and Colds Fresh or decoction Middle America [18, 21, 23] WP Uganda China LE and Yellow Fever Fresh or decoction Middle America [18, 21–23] WP Uganda China LE and Influenza Fresh or decoction Middle America [18, 21, 23] WP Uganda Acute infectious WP Decoction Hong Kong [23] hepatitis Intestinal worms WP Decoction Africa [18] RT and Africa Malaria Juice [18, 23] WP China Evidence-Based Complementary and Alternative Medicine 5

Table 3: Continued. Disorder Plant partb Mode of use Region/country References LE and Uganda Eye Infection Fresh or juice [18, 21] WP Middle America Decoction for Trinidad and Antimicrobial AP drinking; [22] Tobago bathing/external use Decoction or Pulmonary Cuba WP maceration; taken [20, 23] tuberculosis China orally Bacterial infections Trinidad and in WP Decoction [22] Tobago gastrointestinal tracts Decoction; taken Renal infection LE Cuba [20] orally China LE and Sore throat Fresh or decoction Middle America [18, 21, 23] WP Uganda Decoction; taken Cuba Cough WP [20, 23] orally China Coolness of the Decoction; taken WP Cuba [20] uterus orally Menstrual Decoction; taken WP Cuba [20] irregularities orally Dysmenorrhea WP Decoction Bafia, Cameroon25 [ ] Hyperemesis gravidarum WP Decoction Africa [18] (morning sickness) Hemorrhoids WP Decoction Hong Kong [23] China LE and Nose bleeds Fresh or decoction Middle America [18, 21, 23] WP Uganda Cuba LE and Maceration or juice; Stomach ulcers Middle America [18, 20, 21, 23] WP taken orally Uganda Trinidad and Tobago Fresh plant or Africa Cuts,burns,andskin LE and decoction; topical China [18, 22, 26] problems WP application/bathing Cameroon Brazil Venezuela China Africa Wounds WP Crushed herb [23] Central America Hawaii Snake bites WP Pulverized herb China [23] bLE:leaves,ST:stem,FW:flower,WP:wholeplant,RT:roots,AP:aerialparts.

primary pharmacological properties of B. pilosa extracts and guided isolation and fractionation methods to discover new phytochemicals are presented and discussed. compounds from B. pilosa. For example, Kviecinski and colleagues tested hydroalcoholic crude extracts, chloroform, ethyl acetate, and methanol fractions for anti-tumor activity 3.1. AntiCancer Activity. Folkloric reports revealed the pos- [82]. The cytotoxicity of the extracts was assessed using 2 sible antitumor efficacy of B. pilosa, and several scientific in brine shrimp, hemolytic, MTT, and neutral red uptake vitro studies have supported the claim that B. pilosa extracts (NRU) assays. In vivo studies were performed using Ehrlich and isolated compounds possess anti-cancer activities against ascites carcinoma in isogenic BALB/c mice. Among them, a variety of cancer cells. Several studies have used bioassay 6 Evidence-Based Complementary and Alternative Medicine

Table 4: Aliphatic natural products isolated from B. pilosa [30].

Plant part S. N. IUPAC names Common names Structure References (country) 1 Heneicosane CH3(CH2)19CH3 Aerial (Tanzania) [18] 2 Docosane CH3(CH2)20CH3 Aerial (Tanzania) [18] 3 Tricosane CH3(CH2)21CH3 Aerial (Tanzania) [18] 4 Tetracosane CH 3(CH2)22CH3 Aerial (Tanzania) [18] 5 Pentacosane CH3(CH2)23CH3 Aerial (Tanzania) [18] 6 Hexacosane CH3(CH2)24CH3 Aerial (Tanzania) [18] 7 Heptacosane CH3(CH2)25CH3 Aerial (Tanzania) [18] Aerial (Tanzania); not 8 Octacosone CH (CH ) CH [18, 31] 3 2 26 3 found (Taiwan) Aerial (Tanzania); not 9 Nonacosane CH (CH ) CH [18, 31] 3 2 27 3 found (Taiwan) Aerial (Tanzania); not 10 Triacontane CH (CH ) CH [18, 31] 3 2 28 3 found (Taiwan) Aerial (Tanzania); not 11 Hentriacontane CH (CH ) CH [18, 31] 3 2 29 3 found (Taiwan) Aerial (Tanzania); not 12 Dotriacontane CH (CH ) CH [18, 31] 3 2 30 3 found (Taiwan) Aerial (Tanzania); not 13 Tritriacontane CH (CH ) CH [18, 31] 3 2 31 3 found (Taiwan) 14 2-Butoxy-ethanol CH3(CH2)3OCH2CH2OH Whole (Taiwan) [32] 15 Tetracosan-1-ol CH3(CH2)22CH2OH Aerial (Tanzania) [18] 16 Hexacosan-1-ol CH3(CH2)24CH2OH Aerial (Tanzania) [18] 17 1-Octacosanol CH3(CH2)26CH2OH Aerial (Tanzania) [18] Not found 18 1-Hentriacontanol CH (CH ) CH OH [31] 3 2 29 2 (Taiwan) 19 Tetradecanoic acid Myristic acid CH3(CH2)12CO2H Aerial (Tanzania) [18] 20 Hexadecanoic acid Palmitic acid CH3(CH2)14CO2H Aerial (Tanzania) [18] 21 Octadecanoic acid Stearic acid CH3(CH2)16CO2H Aerial (Tanzania) [18] 22 Eicosanoic acid Arachidic acid CH3(CH2)18CO2H Aerial (Tanzania) [18] Leaves 23 Docosanoic acid Behenic acid CH (CH ) CO H [33] 3 2 20 2 (Philippines) O OH 24 2-butenedioic acid HO Aerial (China) [34, 35] O O

(Z)-9-Octadecenoic OH 25 Oleic acid Aerial (Tanzania) [18] acid Evidence-Based Complementary and Alternative Medicine 7

Table 4: Continued. Plant part S. N. IUPAC names Common names Structure References (country) O (E)-9-Octadecenoic Leaves 26 Elaidic acid [33] acid HO (Philippines)

O Aerial (Z,Z)-9,12- Linolic acid or Linoleic 27 (Tanzania); [18, 36] Octadecadienoic acid acid OH whole (Taiwan)

O (Z,Z,Z)-9,12,15- 28 Octadecatrienoic 𝛼-Linoleic acid OH Whole (Taiwan) [32] acid

O (Z,Z)-9,12- 29 Octadecadienoic acid, Ethyl linoleate O Whole (Taiwan) [32] ethyl ester

(Z,Z,Z)-9,12,15- 30 Octadecatrienoic Methyl linolenate O Whole (Taiwan) [32] acid, methyl ester O

(Z,Z,Z)-9,12,15- 31 Octadecatrienoic Ethyl linolenate O Whole (Taiwan) [32] acid, ethyl ester O

(Z)-9-Octadecenoic 32 acid, 2-butoxyethyl 2-Butoxyethyl oleate O Whole (Taiwan) [32] O ester O

O 2-Butoxyethyl O 33 Whole (Taiwan) [32] linoleate O

(Z,Z,Z)-9,12,15- 34 O Octadecatrienoic 2-Butoxyethyl linoleate O Whole (Taiwan) [32] acid, butoxyethyl ester O

1,7E,9E,15E- Heptadeca-2E,8E,10E, Not found 35 Heptadecatetraene- [37] 16-tetraen-4,6-diyne (China) 11,13-diyne 8 Evidence-Based Complementary and Alternative Medicine

Table 4: Continued. Plant part S. N. IUPAC names Common names Structure References (country)

1,11-Tridecadiene- Roots (not 36 [38] 3,5,7,9-tetrayne stated)

Leaves (not 1-Tridecene-3,5,7,9,11- stated) and 37 Pentayneene [38, 39] pentayne not found (Egypt)

5-Tridecene-7,9,11- not found 38 [39] triyne-3-ol (Egypt) HO OH 2,10,12-Tridecatriene- Part not specified 39 [40] 4,6,8-triyn-1-ol (Not stated)

OH Roots (Not 1,11-Tridecadiene- 2,12-Tridecadiene- stated); not 40 3,5,7,9- [38, 39] 4,6,8,10-tetrayn-1-ol found tetrayn-13-ol (Egypt) O 1,11-Tridecadiene- 2,12-Tridecadiene- 41 3,5,7,9- Roots (Germany) [41] 4,6,8,10-tetraynal tetrayn-13-al

2,12-Tridecadiene- 1,11-Tridecadiene- O Roots (not 42 4,6,8,10-tetrayn-1-ol, 3,5,7,9- [38] stated) 1-acetate tetrayn-13-acetate O OH (5E)-1,5- 43 Tridecadiene-7,9- Aerial (China) [42] diyn-3,4,13-triol OH HO

(6E,12E)-3-oxo- OH 44 tetradeca-6,12-dien- Aerial (China) [42] 8,10-diyn-1-ol O HO

1,2-Dihydroxy-5(E)- (E)-5-Tridecene- 45 tridecene- Whole (Taiwan) [43] 7,9,11-triyne-1,2-diol 7,9,11-triyne OH

OH 1,3-Dihydroxy-6(E)- (E)-6-Tetradecene- 46 tetradecene- Whole (Taiwan) [43–45] 8,10,12-triyne-1,3-diol 8,10,12-triyne OH

OH (2R,3E,11E)-3,11- Not found (Egypt 47 Tridecadiene-5,7,9- Safynol [37, 39] and China) triyne-1,2-diol HO Evidence-Based Complementary and Alternative Medicine 9

Table 4: Continued. Plant part S. N. IUPAC names Common names Structure References (country) HO 5,7,9,11- OH 1,2-Dihydroxytrideca- 48 Tridecatetrayne-1,2- Whole (Taiwan) [43, 44, 46] 5,7,9,11-tetrayne diol

HO

(R)-3,5,7,9,11- (R)-1,2- 49 Tridecapentayne-1,2- Dihydroxytrideca- Aerial (Japan) [47] diol 3,5,7,9,11-pentayne OH HO OH H O H (4E)-1- 2-𝛽-D- H ˙ O (Hydroxymethyl)-4- Glucopyranosyloxy- H OH Aerial (USA); [8, 45, 48– 50 dodecene- 1-hydroxy-5(E)- whole (Taiwan); OH H 51] 6,8,10-triyn-1-yl-𝛽-D- tridecene- leaves (Taiwan) glucopyranoside 7,9,11-triyne HO

HOOH H O (4E)-1-(2- H 3-𝛽-D- H O Aerial (USA and Hydroxyethyl)-4- H˙ Glucopyranosyloxy- OH China); whole [8, 35, 42, 51 dodecene- 1-hydroxy-6(E)- OH H (Taiwan); leaves 45, 48–51] 6,8,10-triyn-1-yl-𝛽-D- tridecene-8,10,12-triyne (Taiwan) glucopyranoside OH

OH OH HO 𝛽-D- 3-Hydroxy-6- H Glucopyranosyloxy- O tetradecene-8,10,12- H 52 3-hydroxy- Whole (Mexico) [52] triynyl-𝛽-D- O H 6E-tetradecence- OH H glucopyranoside 8,10,12-triyne H OH HOOH 1-(Hydroxymethyl)- Cytopiloyne, H O 4,6,8,10- 2-𝛽-D- H Whole (Taiwan); 53 dodecatetrayne- Glucopyranosyloxy- H O [8, 50, 53] H leaves (Taiwan) 1-yl-𝛽-D- 1-hydroxytrideca- OH OH H glucopyranoside 5,7,9,11-tetrayne HO

OH 2-O-D- O Glucosyltrideca-11E- H 54 Leaves (Brazil) [27] en-3,5,7,9-tetrayn-1,2- HO O diol HO H H H OH H OH 10 Evidence-Based Complementary and Alternative Medicine

Table 4: Continued. Plant part S. N. IUPAC names Common names Structure References (country)

OH (R)-1- O (Hydroxymethyl)- 2- 𝛽 -D- H 2,4,6,8,10- Glucopyranosyloxy- Aerial (China 55 HO O [35, 47] dodecapentayn- 1-hydroxytrideca- H H and Japan) 1-yl-a-Dˆ - 3,5,7,9,11-pentayne HO glucopyranoside H OH H OH O O OH 1- O [[(Carboxyacetyl)oxy] H HO O 56 methyl]4,6,8,10- O Aerial (Japan) [54] dodecatetraynyl- HO H H a-Dˆ -glucopyranoside H OH H OH O O OH (4E)-1- O H [[(Carboxyacetyl)oxy] HO O 57 -methyl]-4-dodecene- O Aerial (Japan) [54] 6,8,10-triynyl-a-Dˆ - HO H H glucopyranoside H OH H OH O

O OH (4E)-1- O O [[(Carboxyacetyl) H oxy]-ethyl]-4- 58 HO Aerial (Japan) [54] dodecene- O 6,8,10-triynyl- HO H H a-Dˆ -glucopyranoside H OH H OH OH

(5E)-5-Heptene-1,3- 1-Phenylhepta-1,3- 59 Whole (Taiwan) [32] diyn-1-yl-benzene diyn-5en OH Roots (not 7-Phenyl-2(E)- 60 stated); Aerial [38, 42] heptene-4,6-diyn-1-ol (China)

O 7-Phenyl-2(E)- Roots (not 61 heptene-4,6-diyn-1- O [38, 55, 56] stated; Brazil) ol-acetate

OH

7-Phenyl-4,6- Whole (Taiwan); 62 Pilosol A [42, 57] heptadiyn-2-ol aerial (China) Evidence-Based Complementary and Alternative Medicine 11

Table 4: Continued. Plant part S. N. IUPAC names Common names Structure References (country) OH

7-Phenylhepta-4,6- 63 Aerial (China) [42] diyn-1,2-diol HO

Leaves (not stated); leaves of tissue culture 64 1,3,5-Heptatriyn-1-yl- 1-Phenylhepta-1,3,5- (not stated); [18, 34, 38, benzene triyne aerial (Tanzania; 42, 56–58] China); Whole (Taiwan); roots (Brazil) Leaves (not 7-Phenyl-2,4,6- 65 stated); aerial [38, 42] heptatriyn-1-ol HO (China)

7-Phenyl-2,4,6- O Leaves (not 66 heptatriyn-1-ol- [38] O stated) acetate S 5-(2-Phenylethynyl)- OH 67 2-thiophene Aerial (China) [42] methanol S O H OH OH H 5-(2-Phenylethynyl)- O H 68 2𝛽-glucosylmethyl- H Aerial (China) [42] thiophene O H HOOH OH H 𝛽 3- -D- O H Glucopyranosyl-1- HO Leaves 69 hydroxy-6(E)- HO [28] H (Cameroon) tetradecene-8,10,12- O OH H H triyne OH

1-Phenyl-1,3-diyn-5- O 70 en-7-ol-acetate Leaves (Brazil) [27] O

S.N. denotes serial number. the chloroform fraction was the most toxic with a half value of 6.5 ± 0.01 𝜇g/mL. Moreover, the positive control, maximal inhibitory concentration (IC50)of97 ± 7.2 and 83 ± taxol, showed higher activity than phenyl-1,3,5-heptatriyne 5.2 𝜇g/mL in NRU and MTT, respectively [82]. Kumari and [77]. Furthermore, in vitro comet assays were performed to colleagues also reported the anti-cancer and anti-malarial evaluate the toxicity of n-hexane, chloroform, and methanol activities of B. pilosa leaves [77]. Based on a cytotoxicity- extracts of B. pilosa and its ethyl acetate, acetone, and water directed fractionation strategy, they identified phenyl-1,3,5- fractions on Hela and KB cells. The ethyl acetate fraction from heptatriene with IC50 values of 8±0.01, 0.49±0.45, 0.7±0.01, the methanol extract exhibited the highest activity with half and 10 ± 0.01 𝜇g/mL against human oral, liver, colon, and maximal cytotoxic concentrations (CTC50)of965.2𝜇g/mL breast cancer cell lines, respectively. However, phenyl-1,3,5- and 586.2 𝜇g/mL against Hela and KB cells, respectively. heptatriyne showed lower activity against breast cancer cell Despite the moderate toxicity, these findings suggest that lines than the chloroform leaf extract which had an IC50 these B. pilosa extracts/fractions could be useful for future 12 Evidence-Based Complementary and Alternative Medicine ] ] 60 ] ] ] , 59 , 35 35 59 59 , 35 [ 35 References [ Plant part (Country) aerial (China) Aerial (China) [ Aerial (China) [ Leaves (Japan) [ Leaves (Japan and Leaves (Not stated); China); aerial (China) OH OH OH OH OH OH OH OH OH ]. OH O O O 30 [ O O O O O B. pilosa O O HO O HO O O OH H H H OH O OH O OH HO O H H H O H H H H HO HO H OH O OH H H H H HO HO HO HO HO HO Table 5: Flavonoids isolated from - - - 󸀠 D D - - - - 󸀠 -acetyl- ,4 󸀠 󸀠 𝛽 𝛽 O ,4 - - ,4 󸀠 󸀠 O O -(6- -tetrahydroxy; )-6,7,3 Aurone, Aurone, Aurone, Aurone, 󸀠 6,7,3 O )-7- )-6- -6,7,3 Sulfuretin Z maritimein ,4 Z maritimetin Z ( -glucopyranosyl) tetrahydroxy tetrahydroxy 󸀠 tetrahydroxy; ( ( glucopyranosyl- glucopyranosyl- D )-6- - Z 𝛽 ( 6,7,3 - -2- ] D - 𝛽 - - ] ] -7- -6- ] ] -Acetyl- -7-hydroxy-3(2H)- O ] methylene methylene methylene methylene (6- benzofuranone benzofuranone benzofuranone benzofuranone benzofuranone hydroxy-3(2H)- -glucopyranosyloxy)- [ 6-hydroxy-3(2H)- 6-hydroxy-3(2H)- -glucopyranosyloxy)-7- D 6- (3,4-Dihydroxyphenyl)- (3,4-Dihydroxyphenyl)- (3,4-Dihydroxyphenyl)- (3,4-Dihydroxyphenyl)- 6,7-dihydroxy-3(2H)- - D (3,4-dihydroxyphenyl)- [ [ [ [ glucopyranosyl)oxy - [ 𝛽 ( 𝛽 2- 2- 2- 2- ( methylene S. N.71 IUPAC names Common names Structure 75 74 72 73 Evidence-Based Complementary and Alternative Medicine 13 ] ] ] 61 , ] ] 61 35 , , 35 59 60 , 34 34 [ 34 [ References Plant part (Country) aerial (China) aerial (China) Aerial (China) [ Leaves (Japan) [ Leaves (China) [ Leaves (not stated); Leaves (not stated); OH OH OH OH OH OH OH OH OH OH O O O O O O O O O O HO HO O HO O HO OH O HO O O H H H OH O O H O O H OH O OH O H OH H O H O H H H O H H O H O O H H O O H H H H H O H O O H Table 5: Continued. O O HO O HO H O H O O O O O O HO HO HO - - - D - 󸀠󸀠 󸀠󸀠 - - - O 󸀠󸀠 𝛽 - ,6 ,6 - O- 󸀠󸀠 󸀠󸀠 D ,6 D - - 𝛽 - 󸀠󸀠 󸀠 ,4 ,4 )-6- 𝛽 𝛽 - 󸀠󸀠 󸀠󸀠 ,4 -(6- Z 󸀠 -(4 D O - -(3 -acetyl- -(2 O 𝛽 -tetrahydroxy -tetrahydroxy -tetrahydroxy -tetrahydroxy; O O O 󸀠 󸀠 󸀠 Aurone, Aurone, Aurone, Aurone, 󸀠 6,7,3 coumaroyl- )-6- ,4 ,4 ,4 triacetyl- )-6- ,4 󸀠 󸀠 󸀠 Z tetrahydroxy bidenoside A 󸀠 p- diacetyl- triacetyl- )-6- ( )-6- Z ( -glucropyranosyl)- Aurone, ( Z Z glucropyranosyl)- glucropyranosyl)- glucropyranosyl)- glucropyranosyl)- ( ( D 6,7,3 6,7,3 6,7,3 (3,6-di- 6,7,3 - - - - - ] ] - - D D D - - ] - - ] - 𝛽 -2- 𝛽 ] 𝛽 𝛽 𝛽 - oxy ] ] -7- ] 3-(4- [ -acetyl- - -acetyl- -Acetyl- -7-hydroxy- -7-hydroxy- -7-hydroxy- -Acetyl- O ] ] ] O -7-hydroxy-6- O O O ] 6- [[ hydroxy-6- methylene 6- hydroxyphenyl)- (3,6-di- (4,6-di- -glucopyranosyl (3,4-dihydroxyphenyl)- (3,4,6-tri- [ (3,4-Dihydroxyphenyl)- (3,4-Dihydroxyphenyl)- (3,4-Dihydroxyphenyl)- [ 1-oxo-2-propenyl glucopyranosyl)oxy -glucopyranosyl)-oxy 3(2H)-benzofuranone 3(2H)-benzofuranone 3(2H)-benzofuranone 3(2H)-benzofuranone 3(2H)-benzofuranone [ [ glucopyranosyl)-oxy (3,4-dihydroxyphenyl)- [ [ [ methylene methylene methylene (2,4,6-tri- glucopyranosyl)oxy D [ D [ methylene 6- 6- 2- 2- 2- 2- S. N. IUPAC names76 Common names Structure 78 80 79 77 14 Evidence-Based Complementary and Alternative Medicine ] ] ] ] ] 63 63 , – 35 35 60 61 59 [ [ References Plant part (Country) leaves (Japan) Aerial (China) [ Aerial (China) [ Leaves (China) [ flowers (Germany) Leaves (Germany); Flowers (Germany); OH OH OH OH OH OH OH OH OH OH O O O OH O OH O O HO OH O H O OH OH HO O HO H H H OH O OH O H H H HO HO H OH H H Table 5: Continued. H HO H OH H OH H HO H HO HO HO HO HO - 󸀠 ,4 󸀠 ,3,2 -D- 𝛼 glucoside glucoside 𝛽 - O -D- -D- - Butein Okanin Okanin Okanin 󸀠 𝛽 𝛽 2 tetrahydroxy- O- O- glucopyranosyl - - 󸀠 󸀠 Chalcone, 3 4 - ] -2- ] -2- ] -D- -D- -D- 𝛽 𝛽 𝛽 3-( 2-( 4-( [ [ [ 1- 1- 1- propen-1-one propen-1-one hydroxy-3-(3- 2-propen-1-one 2-propen-1-one 2-propen-1-one hydroxyphenyl)-2- glucopyranosyloxy)- glucopyranosyloxy)- 4-hydroxyphenyl Glucopyranosyloxy)- 2,3-dihydroxyphenyl 2,4-dihydroxyphenyl 3-(3,4-dihydroxyphenyl)- 1-(2,4-Dihydroxyphenyl)- 3-(3,4-Dihydroxyphenyl)- 3-(3,4-Dihydroxyphenyl)- 3-(3,4-Dihydroxyphenyl)- 1-(2,3,4-trihydroxyphenyl)- S. N.81 IUPAC names Common names Structure 85 82 83 84 Evidence-Based Complementary and Alternative Medicine 15 ] ] ] ] 63 34 34 64 References Plant part (Country) Aerial (China) [ Aerial (China) [ Leaves (Germany) [ Flowers (Germany) [ OH OH OH OH OH OH OH OH O O O O OH OH OH OH O O O O HO HO HO H H HO H H OH O O OH O O O OH O H H H H O O O H H H O H H H H O O O Table 5: Continued. H O H H H O H O O O O HO HO HO O O O HO - -D 𝛽 -diacetyl)- O- - 󸀠󸀠 Okanin 󸀠 ,6 4 󸀠󸀠 glucopyranoside (4 - ] - -3- 󸀠󸀠 ] - - (6 -D -D 𝛽 𝛽 -D- -Acetyl- 𝛽 O- O- O - - 󸀠 󸀠 O- -triacetyl)- -triacetyl)- - 󸀠󸀠 󸀠󸀠 󸀠 ,6 ,6 glucoside glucoside 󸀠󸀠 󸀠󸀠 (4,6-di- propen-1-one [ ,4 ,4 -acetylglucoside) 󸀠󸀠 󸀠󸀠 4- Okanin 4 Okanin 4 O -glucopyranosyl)-oxy [ (3 (2 Okanin 4 1- -D 2,3-dihydroxyphenyl (3,4-dihydroxyphenyl)-2- 𝛽 S. N. IUPAC names86 Common names Structure 87 88 89 16 Evidence-Based Complementary and Alternative Medicine ] ] ] 65 65 64 References Plant part (Country) Leaves (Germany) [ Leaves (Germany) [ Leaves (Germany) [ OH OH OH OH OH OH O O O OH OH OH OH OH OH O O O HO HO HO H H H OH OH O O O O O H H H H H H Table 5: Continued. H H O H O O H H H O O O HO O O HO HO HO O O - trans - 󸀠󸀠 -(6 -D Okanin glucoside 𝛽 -coumaroyl)- p O- - 󸀠 4 - ] -D- -acetyl- 𝛽 󸀠󸀠 -phenyl - ] ] (4 -diacetyl- 󸀠󸀠 oxy ,4 -D- ] coumaroyl)- coumaroyl)- 󸀠󸀠 1- 𝛽 (2 O- Okanin - glucoside glucoside 󸀠 -D- 3-(4-hydroxyphenyl)- 2-propen-1one 2,3-Dihydroxy-4- 𝛽 [ [ trans-p- trans-p- - - - 1- O- 󸀠󸀠 󸀠󸀠 O oxo-2-propenyl - 6 6 󸀠 3-(3,4-dihydroxyphenyl)- 6- 4 [[ Okanin 4 glucopyranosyl S. N. IUPAC names90 Common names Structure 92 91 Evidence-Based Complementary and Alternative Medicine 17 ] ] ] 63 63 65 References Plant part (Country) Leaves (Germany) [ Flower (Germany) [ Flower (Germany) [ OH OH OH OH OH OH O O O OH OH OH O OH HO H OH O O H H O O H HO H OH H H OH OH O O H O O H H H H HO H HO H H OH H HO O Table 5: Continued. H H O OH H H H H O O H O HO H HO OH O HO H HO O HO HO - 󸀠󸀠 - O- - -D glucoside 󸀠 -diacetyl-6 𝛽 4 󸀠󸀠 -D- ,4 6)- 𝛽 coumaroyl)- 󸀠󸀠 Okanin Okanin → O- (3 glucoside glucopyranosyl- (1 Okanin glucopyranoside -D- -di- -D- 󸀠 𝛽 trans-p- ,4 [𝛽 󸀠 O- 3 - 󸀠 4 S. N. IUPAC names93 Common names Structure 95 94 18 Evidence-Based Complementary and Alternative Medicine ] ] ] ] ] 62 , 34 42 60 60 35 [ References (China) Plant part (Country) Aerial (China) [ Aerial (China) [ Leaves (China) [ Leaves (Germany); aerial O O OH OH OH OH OH OH O O O O OH OH O O Not found Leaves (China) [ OH O H O H O OH O O OH H Ac H H OH O O O H HO H H H OH H H O HO H H O H H O H HO Table 5: Continued. HO H O H HO O Ac O H Ac HO O Ac Ac Ac H HO HO Ac - iso -D 𝛽 O- - Okanin 󸀠 3 Okanin, 4-methyl ether- glucopyranoside - - 󸀠󸀠󸀠 - ] -D ,6 𝛽 -4H- 󸀠󸀠󸀠 ] ,4 󸀠󸀠 - ,6 -trimethoxy- -D 󸀠 󸀠󸀠 -acetyl- 𝛽 ,6 (4 󸀠 O - 3-( ,4 𝛽 [ 󸀠 dihydro 1- -glucopyranosyl- O- tetraacetyl)- 3-(3-hydroxy)- 2-propen-1-one glucopyranoside -di- O-D 4-methoxyphenyl)- 1-benzopyran-4-one 󸀠 Glucopyranosyloxy)- (2,4,6-tri- 2,4-dihydroxyphenyl dihydro-7,8-dihydroxy- 4- 2,3-dihydro-8-hydroxy- Okanin 4-methyl ether- [ ,4 4H-1-benzopyran-4-one glucopyranosyl)oxy 󸀠 2-(3,4-Dihydroxyphenyl)- 7 3 2-(3,4-Dihydroxyphenyl)-2,3- Chalcone, 2 S. N. IUPAC names96 Common names Structure 99 97 98 100 Evidence-Based Complementary and Alternative Medicine 19 , ] ] 42 , ] ] ] 66 67 , , 35 68 66 66 , 42 66 34 References [ Vietnam) Plant part (Country) Aerial (Uganda) [ Aerial (Tanzania) [ Aerial (Tanzania) [ Aerial (Tanzania; China; Aerial (Tanzania; China) [ OH OH OH OH OH OH O O O O O O O O OH O OH O O O O O O O O OH OH H H OH O OH O H H ˙ H H HO HO H OH H OH Table 5: Continued. H H HO HO HO HO -D- 𝛽 O- Luteolin Apigenin Apigenin -Methylhosludin -glucopyranoside glucopyranoside O O Luteolin 7- 5- 7- 5-hydroxy-2- -glucopyranosyloxy)- -Glucopyranosyloxy)- 2-phenyl-4H-1- 5hydroxy-4H-1- 5,7-Dihydroxy-2- 5,7-Dimethoxy-6- benzopyran-4-one benzopyran-4-one benzopyran-4-one (4-hydroxyphenyl)- (4-hydroxyphenyl)- -D 5,7-dihydroxy-4H-1- -D (5-methoxy-6-methyl- 𝛽 4-oxo-4H-pyran-3-yl)- 𝛽 4H-1-benzopyran-4-one 4H-1-benzopyran-4-one 2-(3,4-Dihydroxyphenyl)- 2-(3,4-Dihydroxyphenyl)- 7-( 7-( S. N. IUPAC names101 Common names Structure 102 103 105 104 20 Evidence-Based Complementary and Alternative Medicine ] ] ] ] ] 71 – 35 67 42 69 69 [ References (Taiwan) Plant part (Country) Aerial (China) [ Aerial (China) [ Whole (Taiwan) [ Aerial (Japan); whole OH O OH HO HO H O OH OH H O O OH OH ˙ H OH H O OH O O O O H O O O OH O OH Not found Aerial (Vietnam) [ O O O O O O H OH O H OH OH H O HO H H O H OH H H HO H OH Table 5: Continued. HO H HO HO HO - O ; Centaurein Axillaroside Centaureidin -glucopyranoside -3- -D 𝛽 -coumaroyl- E-p (3-hydroxy-4- Kaempferol 3- -glucopyranosyloxy)- -Glucopyranosyloxy)- -Glucopyranosyloxy)- 5,7-dihydroxy-2- -rhamnopyranoside) 5,7-Dihydroxy-2- dimethoxy-4H-1- dimethoxy-4H-1- benzopyran-4-one benzopyran-4-one L (4-hydroxyphenyl)- -D -D -D methoxyphenyl)-3,6- methoxyphenyl)-3,6- - 𝛽 𝛽 𝛽 (2,3-di- 4H-1-benzopyran-4-one 4H-1-benzopyran-4-one 𝛼 2-(3,4-Dihydroxyphenyl)- 5-hydroxy-3,6-dimethoxy- 5-hydroxy-2-(3-hydroxy-4- 7-( 7-( 3-( S. N. IUPAC names106 Common names Structure 107 108 109 110 Evidence-Based Complementary and Alternative Medicine 21 ] ] ] ] ] 72 , 73 73 67 42 37 References Plant part (Country) Aerial (China) [ Not found (Japan) [ Not found (Japan) [ Not found (China) [ OH OH O O OH O H OH O H HO H H OH O O HO O OH H O O O O O O O OH OH O O O O OH Not found Aerial (Vietnam) [ OH O O O H H H OH O OH O O OH O O H H H H H H H OH H Table 5: Continued. H OH H H H HO HO HO HO HO HO - 𝛼 - ] O [ - iso -D 𝛽 Luteoside (1-2)- Eupatorin, glucopyranoside -rhamnopyranosyl- L 3- - ] -L- 𝛼 dihydroxy- (6-deoxy- [ glucopyranosyloxy)- 5-Hydroxy-2- (3-hydroxy-4- -Glucopyranosyloxy)- chromen-4-one 7- dimehtoxy-4H-1- methoxyphenyl)- 8-methoxy-4H-1- benzopyran-4-one benzopyran-4-one benzopyran-4-one 6,7-dimethoxy-4H- -glucopyranosyloxy)-3,5- -D -D- methoxyphenyl)-3,8- methoxyphenyl)-4H-1- Mannopyranosyl)oxy 𝛽 𝛽 -D 5hydroxy-2-(4-hydroxy-3- 5-hydroxy-2-(4-hydroxy-3- 3- 𝛽 2-(3,4-Dimethoxyphenyl)-7- 7-( ( S. N. IUPAC names111 Common names Structure 112 113 114 115 22 Evidence-Based Complementary and Alternative Medicine ] ] 74 70 ] , , ] ] 54 42 42 66 60 , , 35 42 References [ [ Plant part (Country) Aerial (China) [ Aerial (China) [ whole (Taiwan) Aerial (Tanzania) [ Aerial (China); leaves Aerial (Japan; China); (China); whole (China) OH O OH O OH OH O OH OH OH H OH OH H O O HO H H O OH O HO H O O O O O O O O O OH OH O O OH OH O O O O OH H H OH OH O O H H O H H HO HO OH OH H H HO H H Table 5: Continued. HO HO HO HO - 󸀠 O- - glucoside 󸀠 - 𝛽 3,6,3 O- -glucoside trimethyl -trimethyl ether 𝛽 󸀠 Jacein; Quercetin trimethyl ether-6- Quercetagetin 3,7,3 3,6,3 ether-7- - -D 𝛽 O- 4-one 5-hydroxy-2- Glucopyranosyloxy)- (4-hydroxy-3- (4-hydroxy-3- chromen-4-one 2-(4-hydroxy-3- 6-((2S,3S,4S,5S)- 3,5,7-trihydroxy- Glucopyranoside 5,7-Dihydroxy-2- methoxyphenyl)- dimethoxy-4H-1- dimethoxy-4H-1- 4H-1-benzopyran- Luteolin 3- benzopyran-4-one benzopyran-4-one 3,7-dimethoxy-4H- -D- methoxyphenyl)-3,6- methoxyphenyl)-3,6- 𝛽 6-(hydroxymethyl)-2H- 2-(3,4-Dihydroxyphenyl)- pyran-2-yloxy)-5-hydroxy- 7-( Tetrahydro-3,4,5-trihydroxy- S. N. IUPAC names116 Common names Structure 118 119 117 120 Evidence-Based Complementary and Alternative Medicine 23 , , ] ] ] 66 66 75 , , 71 71 ] , , , 59 60 75 74 , , 70 , 66 [ 35 37 71 References [ [ (China) (Taiwan) Plant part (Country) Aerial (Japan); Whole Japan); Leaves (Japan) Leaves (China); Whole Aerial (Tanzania; Japan) [ Aerial (Tanzania; China; Aerial (Tanzania; Japan); H OH OH H OH H OH OH H OH H OH OH H H OH H H H OH OH OH OH O OH OH H H OH OH OH OH O H H HO HO O H H H H H H H O HO O O HO HO HO O H H H HO H O O O O O O O O O O O O OH OH OH OH HO HO HO Table 5: Continued. HO - - - -D -D 𝛽 𝛽 𝛽 O- O- O- Quercetin -robinobioside O -glucopyranoside Quercetin 3- 3- D Quercetin 3- Quercetin 3- galactoside; hyperin; glucuronopyranoside - ] oxy ] -(6- O 6- acid 4-one [[ -mannopyranosyl)- 3- 4-oxo-4H-1- -L -glucopyranosyloxy)- 5,7-dihydroxy- 5,7-dihydroxy- 5,7-dihydroxy- 𝛼 -galactopyranosyloxy)- dihydroxy-4H-1- benzopyran-3-yl- 4H-1-benzopyran- benzopyran-4-one -glucopyranosiduronic -D -galactopyranosyl -D 𝛽 -D 4H-1-benzopyran-4-one 𝛽 -D 2-(3,4-Dihydroxyphenyl)- 2-(3,4-Dihydroxyphenyl)- 2-(3,4-Dihydroxyphenyl)- 𝛽 𝛽 3-( 2-(3,4-dihydroxyphenyl)-5,7- 3-( Deoxy- S. N. IUPAC names121 Common names Structure 123 122 124 24 Evidence-Based Complementary and Alternative Medicine ] ] ] ] 76 71 35 76 , , , , 56 35 34 56 , 5 References Plant part (Country) Roots (Brazil) [ Roots (Brazil) [ Aerial (China) [ Aerial (Japan; China) [ OH O OH O OH O OH O O H OH H O OH O OH HO H O O H H O O O O O O OH OH O O OH O OH H O H H OH H O HO OH O H H O H HO H OH H O H O O O H H O H HO H H HO H OH H H H H HO Table 5: Continued. H H H HO HO HO H H HO HO HO HO HO - 𝛼 - - - - O 󸀠 󸀠 󸀠 - -D- 𝛽 -D 𝛽 -rutinoside 6)- O O- → dimethyl 7- (1 Isoquercitrin dimethyl ether Quercetin 3,3 Quercetin 3,3 Quercetin 3,4 glucopyranoside glucopyranoside -rhamnopyranosyl- ether-7- L dimethyl ether 7- - ] - - -5- -L -L -D- ] oxy 𝛼 𝛼 ] 𝛽 oxy ] 4-one -(6-Deoxy- -(6-Deoxy- O O 5-hydroxy-2- Glucopyranosyloxy)- (4-hydroxy-3- 6- 6- glucopyranosyl methoxyphenyl)- 3-methoxy-4H-1- 3-methoxy-4H-1- benzopyran-4-one benzopyran-4-one mannopyranosyl)- 3-methoxyphenyl)- [[ [[ -glucofuranosyloxy)-5,7- -D- 7- 7- 𝛽 -D- glucopyranosyl mannopyranosyl)- 4H-1-benzopyran-4-one -D 5-hydroxy-2-(4-hydroxy- hydroxy-2-(3-hydroxy-4- 𝛽 𝛽 2-(3,4-Dihydroxyphenyl)-3- 7-( methoxyphenyl)-3-methoxy- dihydroxy-4H-1-benzopyran- ( S. N. IUPAC names125 Common names Structure 128 126 127 Evidence-Based Complementary and Alternative Medicine 25 ] ] 18 70 References Plant part (Country) Whole (Taiwan) [ Aerial (not stated) [ OH OH C 3 OH OH OH H OH O OH O OH OH OH O O O R C 3 O O H C HO 3 H OH O O OH HO Table 5: Continued. HO -D- 𝛽 - O -rutinoside O galactopyranoside Quercetin 3- Quercetin 3- S. N. IUPAC names129 Common names Structure 130 26 Evidence-Based Complementary and Alternative Medicine

Table 6: Terpenoids isolated from B. pilosa [30].

Plant part S. N. IUPAC names Common names Structure References (Country)

3,7,11,11-Tetramethylbicyclo Leaves 131 [8.1.0]undeca- Bicyclogermacrene [95] (Brazil) 2,6-diene

4,11,11-Trimethyl-8- Leaves 132 methylenebicyclo[7.2.0]undec- E-Caryophyllene [95] (Brazil) 4-ene

1-Methyl-5-methylene-8- Leaves 133 (1-methylethyl)-1,6- Germacrene D [95] (Brazil) cyclodecadiene

4-(1,5-Dimethyl-4- Leaves 134 hexen-1-ylidene)-1- Z-𝛾-Bisabolene [95] (Brazil) methyl-cyclohexene

Decahydro-1,1,4-trimethyl-7- Leaves 135 methylene-1H-cycloprop[e]- 𝛽-Gurjunene [95] (Brazil) azulene

2,6,6,9-Tetramethyl-1,4,8- 𝛼-Humulene; Leaves 136 [95] cycloundecatriene 𝛼-caryophyllene (Brazil)

1,2,3,4,4a,5,6,8a-Octahydro- Leaves 137 1-isopropyl-7-methyl-4- 𝛿-Muurolene [95] (Brazil) methylenenaphthalene

1,2,3,4,4a,5,6,8a- Octahydro-4a,8-dimethyl- Selina-3,7(11)- Leaves 138 [95] 2-(1-methylethylidene)- diene (Brazil) naphthalene

HO (2E,7R,11R)-3,7,11,15- Whole 139 Tetramethyl-2- Phytol [57] (Taiwan) hexadecen-1-ol Evidence-Based Complementary and Alternative Medicine 27

Table 6: Continued. Plant part S. N. IUPAC names Common names Structure References (Country) O 3,7,11,15-Tetramethyl-2- Whole 140 Phytanic acid [57] hexadecenoic acid OH (Taiwan)

3,7,11,15-Tetramethyl-2- Phythyl Leaves 141 hexadecenyl ester-heptanoic [33] heptanoate O (Not stated) acid O (3S,10R,13R)-17- ((2R,5R)-5-Ethyl-6- methylheptan-2-yl)- 2,3,4,7,8,9,10,11,12,13,14,15,16,17- Aerial 142 Campestrol [18] tetradecahydro- (Tanzania) 10,13-dimethyl-1H- cyclopenta[a]phenanthren- HO 3-ol Not found 143 Not found Phytosterin-B Not found (Taiwan; [39, 96] Egypt)

Aerial (Tanzania); 144 Stigmast-5-en-3-ol 𝛽-sitosterol [18, 31, 57] H whole (Taiwan) H H HO

13,14,15,16,17- Tetradecahydro-10,13- dimethyl-1H- 𝛽-Sitosterol H Not found 145 cyclopenta[a]phenanthren- HOH [39] glucoside (Egypt) 3-yloxy)-tetrahydro- HO HO O H H 6-(hydroxymethyl)- HO H OH 2H-pyran-3,4,5-triol H H

5𝛼-Stigmasta-7- Whole 146 [57] en-3𝛽-ol (Taiwan)

HO

Whole 147 5𝛼-Stigmasta-7,22t-dien-3𝛽-ol [57] (Taiwan)

HO

Not found (Taiwan); aerial (Tanzania); [18, 31, 33, 148 Stigmasta-5,22-dien-3-ol Stigmasterol H leaves (not 57] stated); H H whole HO (Taiwan) 28 Evidence-Based Complementary and Alternative Medicine

Table 6: Continued. Plant part S. N. IUPAC names Common names Structure References (Country)

H H

Not found 149 Lup-20(29)-en-3-ol Lupeol [39] (Egypt) H HO H

H H

Not found 150 Lup-20(29)-en-3-ol, acetate Lupeol acetate [39] O (Egypt) H O H

H Not found 151 Olean-12-en-3-ol 𝛽-amyrin [39] (Egypt) H HO

5,9,13-Trimethyl- Aerial 152 Friedelan-3𝛽-ol [18] 24,25,26-trinoroleanan-3-ol (Tanzania)

HO

5,9,13-Trimethyl- Friedelin; Aerial 153 [18] 24,25,26-trinoroleanan-3-one friedelan-3-one (Tanzania)

O

Aerial (Tanzania); 2,6,10,15,19,23- Leaves (Not 154 Hexamethyl-2,6,10,14,18,22- Squalene [18, 33, 57] stated); tetracosahexaene Whole (Taiwan)

Leaves (Not 155 𝛽,𝛽-Carotene 𝛽-Carotene [97] stated) Evidence-Based Complementary and Alternative Medicine 29

studies [83]. Hot water extracts of B. pilosa var. minor Sheriff (50% growth inhibition in the NCI tumor line panel), was were also assessed for its antileukemic effects on leukemic 0.24 𝜇M[81]. These data mark centaureidin as a promising cell lines L1210, U937, K562, Raji, and P3HR1 using XTT- antimitotic agent for tumor therapy. based colorimetric assays. The extract inhibited the five cell In addition to anti-tumor flavones, polyynes found in B. 4 lines with IC50 values ranging from 145 𝜇g/mL to 586 𝜇g/mL. pilosa have also been shown to possess anti-tumor proper- L1210, K562, Raji, and P3HR1 were more sensitive to B. pilosa ties. Based on a bioactivity-directed isolation approach, Wu extract with IC50 values below 200 𝜇g/mL [98]. andcolleaguesidentifiedtwopolyyneaglyconesfromthe Consistent with the antitumor activities of B. pilosa ethyl acetate fraction of B. pilosa [44]. 1,2-Dihydroxytrideca- extracts and fractions, some of its phytochemicals also 5,7,9,11-tetrayne (48) and 1,3-Dihydroxy-6(E)- tetradecene- showed anticancer activity as outlined in Table 11.Among 8,10,12-triyne (46) exhibited significant anticell prolifera- them, luteolin (103), a well-studied flavonoid with multiple tion activity in primary human umbilical vein endothelium bioactivities, was more effective against tumor cell prolifera- cells (HUVEC) with IC50 values of 12.5 𝜇Mand1.73𝜇M, tion than its derivatives with IC50 values ranging from 3 𝜇M respectively. They also decreased angiogenesis and pro- to 50 𝜇Mincells,and5to10mg/kginanimals.Luteolinwas moted apoptosis in human endothelial cells. Their anti- also found to fight cancer as a food additive at concentrations angiogenic and cytotoxic effects correlated with activation 3 of 50 to 200 ppm [78] andpreventskincancer[78] and cancer of the CDK inhibitors and caspase-7 [44]. In addition, 1,2- invasion [103]. Lee and colleagues reported that luteolin Dihyroxy-5(E)-tridecene-7,9,11-triyne (45)showedantian- prevents cancer by inhibiting cell adhesion and invasion giogenic effects in HUVECs with an50 IC value of 12.4 𝜇Mas [103]. Significant inhibition concentration was reported to be evidenced by a decrease in the tube formation and migration 5 𝜇M and complete inhibition concentration was reported to of HUVECs [43]. The IC50 value of compound 45 in the be 40 𝜇M. Moreover, luteolin was reported to inhibit hepa- inhibition of basic fibroblast growth factor-induced HUVEC tocyte growth factor (HGF)-induced cell scattering as well growth was 28.2 𝜇M. However, it had higher IC50 values as cytoskeleton changes such as filopodia and lamellipodia than those for lung carcinoma cells and keratinocytes. This which was determined using phase-contrast and fluorescence compound could also inhibit cell proliferation of HUVECs, microscopy. Furthermore, luteolin also inhibited the HGF- lung carcinoma A549 cells and HACAT keratinocytes. The induced phosphorylation of c-Met, ERK 1/2, and Akt as mechanism by which compound 45 inhibits HUVEC growth well as the MAPK/ERK and P13 K-Akt pathways [78, 103]. and angiogenesis is complicated and includes decreasing the Other mechanisms underlying the anticancer activities of expression of cell cycle regulators (CDK4, cyclins D1 and A, luteolin are the inhibition of topoisomerase I and II, which retinoblastoma (Rb), and vascular endothelial growth factor inhibits cell replication and DNA repair thus promoting receptor 1), caspase-mediated activation of CDK inhibitors apoptosis, regulation of PI-3-Kinase/Akt/MAPK/ERK/JNK, p21 (Cip1) and p27 (Kip), upregulation of Fas ligand expres- activation of apoptosis in the mitochondrial pathway by sion, downregulation of Bcl-2 expression, and activation of activating caspase 9 and caspase 3, which were found in caspase-7 and poly (ADP-ribose) polymerase [43]. malignant cells but not in normal human peripheral blood mononuclear cells, death receptor-induced apoptosis, and a cell-cycle arrest mechanism, inhibition of fatty acid synthase which is upregulated in many cancer cells, and sensitization to chemotherapy whereby luteolin increases the susceptibility 3.2. Anti-Inflammatory Activity. B. pilosa is commonly used of cancer cells to chemotherapy [78]. Butein (82)isanother to treat inflammatory disorders. The anti-inflammatory phy- flavonoid that showed a cytotoxic effect on human colon tochemicals present in B. pilosa arelistedinTable11. 5 adenocarcinoma cell proliferation with a reported IC50 value Cyclooxygenase-2 (COX-2) is a physiologically important of 1.75 𝜇M. Butein at 2 𝜇M affected the incorporation of enzyme that converts arachidonic acid to prostaglandin 14 [ C]-labeled leucine, thymidine, and uridine which can (PGE2). Its expression is induced by a wide variety of external cause the inhibition of DNA, RNA, and protein synthesis of stimuli indicating its involvement in inflammatory diseases, human colon cancer cells. Moreover, butein also exhibited anditisusedasaninflammatorymarker[88]. Yoshida and noncompetitive inhibition of 1-chloro-2,4-dinitrobenzene colleagues studied the effects of the aqueous extracts of B. (CDNB) in glutathione S-transferase (GST) activity. Tumor pilosa aerial parts in the production of COX-2 and PGE2 as resistance was correlated with high levels of GST, thus, butein well as on the activation of mitogen activated protein kinases inhibited proliferation of cancer cells [80]. Another flavonoid (MAPKs) in normal human dermal fibroblasts (HDFs) in present in B. pilosa, centaureidin (109), also showed anti- response to inflammatory cytokine, IL-1𝛽.Thisworkshowed cancer activity in B lymphoma cells. Centaureidin, isolated that IL-1𝛽 activated MAPKs such as ERK1/2, p38, and JNK from Polymnia fruticosa, inhibited tubulin polymerization in to different extents and induced COX-2 expression. The vitro and induced mitotic figure formation in CA46 Burkitt COX-2expressioninHDFswasregulatedmainlybyp38 lymphoma cells. Using turbidimetric assay, the IC50 value of following IL-1𝛽 stimulation. Consistently, the p38 inhibitor centaureidin in inhibition of mitosis was 3 𝜇M[104]. Cyto- SB203580 blocked this expression. Using this cell platform, toxicity of centaureidin was further analyzed using American B. pilosa extracts were tested for inhibition of inflammation. National Cancer Institute (NCI) 60 human tumor cell lines. The extract dose-dependently suppressed the activation of The cytotoxicity potency of centaureidin, expressed as GI50 p38 and JNK and moderately suppressed ERK1/2, as well as suppressing COX-2 expression and PGE2 production 30 Evidence-Based Complementary and Alternative Medicine ] ] 84 ] ] ] ] , 99 , 99 99 99 45 100 , 75 43 [ Leaves (India) [ Whole (Japan) [ Whole (Japan) [ whole (Taiwan) Not found (Taiwan); Leaves and roots (Japan) [ Whole and aerial (Japan) [ OH OH O OH OH OH OH O OH O O ]. 30 [ O B. pilosa O O O O O O O HO HO HO O Table 7: Phenylpropanoids isolated from Caffeic acid Ferulic acid Caffeate, ethyl -Coumaric acid p pentan-2,4-dione 3-Propyl-3-(2,4,5- trimethoxy)benzyloxy- acid 2-propenoic acid propenoic acid, ethyl ester 3-(3,4-Dihydroxyphenyl)-2- methoxy]-2,4-pentanedione 3-(4-Hydroxy-3-methoxyphenyl)- 2-Methoxy-4-(2-propen-1-yl)-phenol Eugenol 3-Propyl-3[(2,4,5-trimethoxyphenyl)- 3-(3,4-Dihydroxyphenyl)-2-propenoic 3-(4-Hydroxyphenyl)-2-propenoic acid S. N.156 IUPAC names Common names Structure Plant part (country) References 157 159 158 160 161 Evidence-Based Complementary and Alternative Medicine 31 ] 75 ] ] ] ] ] , 71 73 101 101 101 101 , 70 [ (Taiwan) Leaves (Japan) [ Leaves (Japan) [ Leaves (Japan) [ Leaves (Japan) [ Not found (Japan) [ Aerial (Japan); whole O O OH OH OH OH OH OH OH OH OH O OH O O O O O O HO O O O O O OH OH O O HO O O OH O O O O HO HO O HO HO HO HO HO HO HO HO HO Table 7: Continued. -methyl -methyl -methyl -methyl- C C C C -Erythronic acid, Chlorogenic acid -caffeoyl-2- -caffeoyl-2- -caffeoyl-2- d -Caffeoyl-2- -Erythronate, methyl -Erythronate, methyl O O O -erythrono-1,4-lactone O d d D 3- 2- 2- 3- - ] -2,4- ] oxy ] oxy ] hydroxy-4- methyl ester methyl ester propenyl]oxy]- propenyl dihydroxyphenyl)- propenyl ester-2-propenoic acid 2-methyl-butanoic acid methyl-5-oxo-3-furanyl propen-1-yl]-oxy]-1,4,5- 4-(Acetyloxy)-3-[[3-(3,4- 3-(3,4-Dihydroxyphenyl)-1-oxo-2- 3-(3,4-Dihydroxyphenyl)-1-oxo-2- [[ [[ dihydroxy-2-methyl-butanoic acid, 3- 3-[[3-(3,4-Dihydroxyphenyl)-1-oxo-2- 1-oxo-2-propen-1-yl-]oxy]-2-hydroxy- 2-[[3-(3,4-Dihydroxyphenyl)-1-oxo-2- 2- 3,4-dihydroxy-2-methyl-butanoic acid trihydroxy-cyclohexanecarboxylic acid 3,4-dihydroxy-2-methyl-butanoic acid, 3-(3,4-Dihydroxyphenyl)-tetrahydro-4- S. N.162 IUPAC names Common names Structure Plant part (country) References 166 163 164 165 167 32 Evidence-Based Complementary and Alternative Medicine ] ] 71 71 , , ] 70 70 , , 71 49 49 , , 45 45 [ [ (Taiwan) (Taiwan) Aerial (Japan) [ Aerial (Japan); whole Aerial (Japan); Whole OH OH OH OH OH OH OH O O OH O O OH OH O O O HO HO OH HO O HO O O O O HO O HO HO HO Table 7: Continued. HO acid acid -caffeoylquinic -caffeoylquinic O O -Caffeoylquinic acid O 3,5-Di- 3,4-Di- 4- acid )-3-(3,4-Dihydroxyphenyl)- )-3-(3,4-Dihydroxyphenyl)- propen-1-yl]oxy]- E E cyclohexanecarboxylic acid 1-oxo-2-propen-1-yl]-oxy]-1,5- 4-[[3-(3,4-Dihydroxyphenyl)-1-oxo-2- dihydroxy-cyclohexanecarboxylic acid 1,3,5-trihydroxy-cyclohexanecarboxylic 3,4-bis[[2( 3,4-bis[[2( 1-oxo-2-propen-1-yl]-oxy]-1,4-dihydroxy- S. N. IUPAC names168 Common names Structure Plant part (country) References 170 169 Evidence-Based Complementary and Alternative Medicine 33 ] ] 70 , ] ] 59 , 39 59 49 , 34 [ 45 (China) Leaves (Japan) [ Whole (Taiwan) [ Not found (Egypt) [ Leaves (Japan); aerial O OH OH O O O H H O O O OH O H H H O O H O H O H H O OH OH O H O O HO HO HO HO O HO HO HO O HO HO Table 7: Continued. HO HO - - O O - p - O acid -caffeoylquinic -caoumaroyl- p O -acetyl-6- - caoumaroyl- O O -Coumaric acid, 4- -Coumaric acid, 4- glucopyranosyl) glucopyranosyl) p p (2- - - (6- 4,5-Di- -D -D 𝛽 𝛽 - - D -D - 𝛽 𝛽 -[3-(4- O -Acetyl-6- O -[3-(4-Hydroxyphenyl)- O hydroxyphenyl)- glucopyranosyl]- glucopyranosyl]- )-3-(3,4-Dihydroxyphenyl)-1- E 1-oxo-2-propen-1-yl]- 1-oxo-2-propen-1-yl]- cyclohexanecarboxylic acid oxy]phenyl]2-propenoic acid 3-[4-[[2- 6,7-Dihydroxy-2-chromenone Esculetin, cichorigenin oxy]-phenyl]-2-propenoic acid 3-[4-[[-6- oxo-2-propen-1-yl]oxo]-1,5-dihydroxy- 3,4-bis[[(2 S. N. IUPAC names171 Common names Structure Plant part (country) References 174 173 172 34 Evidence-Based Complementary and Alternative Medicine

Table 8: Aromatic compounds isolated from B. pilosa [30].

Plant part S. N. IUPAC names Common names Structure References (country) HO Whole 175 1,2-Benzenediol Pyrocatechin [99] HO (Japan)

OH Whole 176 4-Ethyl-1,2-benzenediol Pyrocatechol [99] (Japan) OH

O

Dimethoxyphenol; HO 177 Roots (Japan) [99] 2,6-dimethoxyphenol O

OH 4-Ethenyl-2-methoxy- Whole 178 p-Vinylguaiacol [99] phenol (Japan) O

2-Hydroxy-6- 6-Methyl- Whole 179 [99] methylbenzaldehyde salicylaldehyde O (Japan) OH HO Whole 180 Benzene-ethanol 2-Phenyl-ethanol [57] (Japan) HO 4-Hydroxy-3-methoxy- Aerial 181 Vanillin O [99] benzaldehyde O (Japan) OH

3-Hydroxy-4-methoxy- Leaves 182 Vanillin, iso O [99] benzaldehyde (Japan) O O p-Hydroxybenzoic OH Whole 183 4-Hydroxybenzoic acid [99] acid HO (Japan) HO O Stem and 184 2-Hydroxybenzoic acid Salicylic acid [99] roots (Japan) HO OH HO 3,4-Dihydroxybenzoic Whole 185 Protocatechuic acid OH [99] acid (Japan) O OH O 4-Hydroxy- Aerial 186 3methoxybenzoic Vanillic acid O (Uganda); [68, 99] acid Roots (Japan) OH Evidence-Based Complementary and Alternative Medicine 35

Table 8: Continued. Plant part S. N. IUPAC names Common names Structure References (country) HO OH 3,4,5-Trihydroxybenzoic HO Whole 187 Gallic acid [102] acid O (China) HO

[88]. This work supports the use of B. pilosa as an anti- they concluded that this inhibition was not due to the inflammatory agent; however, no compounds responsible for cytotoxicity of ethyl caffeate. The IC50 value of ethyl caffeate the anti-inflammatory activity of B. pilosa were identified. in the inhibition of NO production was 5.5 𝜇g/mL, slightly A further study also reported the anti-inflammatory lower than curcumin (positive control) which has an activity as well as the antiallergic activity of B. pilosa [75]. In IC50 value of 6.5 𝜇g/mL. They demonstrated that ethyl this study, dried powder of the aerial part of B. pilosa,which caffeate exerted anti-inflammatory activity via the reduced hadbeenpretreatedwiththeenzymecellulosine,wasused transcription and translation of iNOS (inducible nitric oxide for further tests. The results showed that oral administration synthase) in RAW 246.7 cells. In addition, this compound of the cellulosine-treated B. pilosa lowered the level of serum also suppressed COX-2 expression in RAW 246.7 cells IgEinmice10daysafterimmunizationwithDNP(2,4- and MCF-7 cells. The in vivo anti-inflammatory effect dintrophenyl)-Ascaris as an antigen. This treatment also of ethyl caffeate was verified by testing in TPA-treated reduced dye exudation in skin induced by passive cutaneous mouseskin.Likecelecoxib,thepositivecontrol,ethyl anaphylaxis and production of inflammatory mediators, his- caffeate significantly abolished COX-2 expression in a tamine, and substance P in rats [75]. Phytochemical analysis dose-dependent manner. Ethyl caffeate at 1 mg/200 𝜇L/site showed that cellulosine treatment increased the percentage of (24 mM) inhibited COX-2 expression at a level comparable caffeic acid and flavonoids. This study suggests that B. pilosa to celecoxib at 1 mg/200 𝜇L/site (13 mM). Remarkably, ethyl and its phenolics have anti-inflammatory functions. caffeate at 48 mM (2 mg/200 𝜇L/site) was more effective Phenolics and polyynes are major anti-inflammatory than celecoxib at 131 mM (10 mg/200 𝜇L/site). Moreover, 6 phytochemicals present in B. pilosa (Table 11). Unsurpris- this compound inhibited the activation of nuclear factor-𝜅B ingly, phenolics such as luteolin (103)andethylcaffeate (NF-𝜅B) by LPS via the prevention of NF-𝜅Bbindingto (161)thataremajorconstituentsofB. pilosa have also DNA [84]. In addition, 3 ethyl caffeate analogs (ethyl 3,4- been reported to possess anti-inflammatory activity. Lute- dihydroxyhydrocinnamate, ethyl cinnamate, and catechol) olin was reported to exhibit anti-inflammatory activity in also showed different degrees of NF-𝜅BbindingtoDNAas macrophages. Xagorari and colleagues showed that luteolin listed in Table 12. Ethyl cinnamate lacks a catechol moiety inhibited the release of inflammatory cytokines, TNF-𝛼 and which results in ineffective inhibition of NF-𝜅Bbindingto interleukin-6, in RAW 264.7 cells following LPS stimulation DNA [84]. [87]. It inhibited TNF-𝛼 production with an IC50 value Pereira and colleagues assessed the anti- 7 of 1 𝜇M. The underlying anti-inflammatory mechanism of inflammatory and immunomodulatory activities of luteolin was reported to be the inactivation of Akt and B. pilosa methanol extract as well as one polyyne, NF-𝜅Bactivation[87]. In addition, luteolin was reported 2-O-𝛽-glucosyltrideca-11(E)-en-3, 5,7,9-tetrayn-1,2-diol to confer anti-inflammatory activity through inhibition of (54) in T lymphocytes and a zymosan-induced arthritis LPS-stimulated iNOS expression in BV-2 microglial cells. mouse model [27]. They first examined the in vitro effect of 8 It inhibited LPS-activated microglia in a dose-dependent the B. pilosa extract and compound 54 on cell proliferation of manner with an IC50 value of 6.9 𝜇M. Moreover, immunoblot human T cells stimulated with 5 𝜇g/mL phytohemagglutinin andRT-PCRdataprovedthatluteolinsuppressedI𝜅B-𝛼 (PHA) or 100 nM 12-O-tetradecanoyl phorbol-13-acetate degradation and iNOS expression in LPS-activated microglia (TPA) plus 15 𝜇M ionomycin and on cell proliferation of [85]. Kim and colleagues stated that luteolin may have mouse T cells stimulated with 5 𝜇g/mL concanavalin A beneficial effects on inflammatory neural diseases through (Con A). The data demonstrated that both methanol extract inhibition of iNOS expression [85]. A related study revealed and compound 54 suppressed T-cell proliferation in a that luteolin decreased the transcriptional activity of NF-𝜅B dose-dependent manner. The estimated IC50 values of the B. RelA via partial inhibition of TNF-mediated NF-𝜅BDNA pilosa extract against human T cells stimulated with 5 𝜇g/mL binding activity. Luteolin also inhibited Akt phosphorylation PHA and 100 nM TPA plus 15 𝜇M ionomycin were 12.5 and and induced degradation of a transcription factor, interferon 25 𝜇g/mL, respectively. In comparison with the methanol regulatory factor (IRF) [86]. extract, compound 54 showed 10-fold more inhibition of Chiang and colleagues showed that ethyl caffeate human T-cell proliferation with an estimated IC50 value of (161) significantly inhibited NO production in mouse 1.5 𝜇g/mL. Accordingly, the B. pilosa extract and compound macrophages, RAW 264.7 cells [84]. Based on MTT assays, 54 dose-dependently suppressed mouse T-cell proliferation 36 Evidence-Based Complementary and Alternative Medicine

Table 9: Porphyrins isolated from B. pilosa [30].

Plant part S. N. IUPAC names Common names Structure References (country) (2E,7R,11R)-3,7,11,15- O Tetramethyl-2-hexadecen-1-yl ester-(15S,16S)-10-ethenyl-5- ethyl-1,16,18,20-tetrahydro- NH N 6,11,15,22-tetramethyl-18,20- Leaves 188 dioxo-15H-9,12-imino-21,2- AristophyllC [100] N HN (Taiwan) metheno-4,7:17,14- dinitrilopyrano[4,3- b]azacyclononadecine-16- O O O O propanoic OPhytyl acid (2E,7R,11R)-3,7,11,15- Tetramethyl-2-hexadecen-1-yl ester-(2S,18S,19S,20bR)-13- NH N ethenyl-8-ethyl- 2a,18,19, 20b-tetrahydro -20b- Leaves 189 (methoxycarbonyl)-9,14,18,24- Bidenphytin A N HN [100] (Taiwan) tetramethyl-4H-12,15-imino-3,5- metheno-7,10:20,17-dinitrilo-1,2- 󸀠 󸀠 O dioxeto-[3 ,4 :3,4]-cyclo- O O OPhytyl pent[1, 2]azacyclononadecine- O O 19-propanoic acid (2E,7R,11R)-3,7,11,15- Tetramethyl-2-hexadecen-1-yl ester-(2S,18S,19S,20bR)-13- ethenyl-8-ethyl- 2a,18,19, 20b-tetrahydro -2a- NH N hydroxy-20b- (methoxycarbonyl)- N Leaves 190 Bidenphytin B HN [100] 9,14,18,24-tetramethyl-4H-12,15- (Taiwan) imino-3,5-metheno-7,10:20,17- OH dinitrilo-1,2- 󸀠 󸀠 O dioxeto[3 ,4 :3,4]- O O OPhytyl O cyclo-pent[1,2-b]- O azacyclononadecine- 19-propanoic acid

NH N (2E,7R,11R)-3,7,11,15- Tetramethyl-2-hexadecen-1-yl ester-(3R,4S,21R)-14-ethyl-21- (132R)-132- N HN Leaves 191 [100] hydroxy-21-(methoxycarbonyl)- Hydroxypheophytin A (Taiwan) 4,8,9,13,18-pentamethyl-20-oxo- O HO 3-phorbinepropanoic acid O OPhytyl O O O

(2E,7R,11R)-3,7,11,15- NH N Tetramethyl-2-hexadecen-1-yl ester-(3R,4S,21S)-14-ethyl-21- (132S)-132- Leaves 192 N HN [100] hydroxy-21-(methoxycarbonyl)- Hydroxypheophytin A (Taiwan) 4,8,9,13,18-pentamethyl-20-oxo- 3-phorbinepropanoic acid O HO O OPhytyl O O Evidence-Based Complementary and Alternative Medicine 37

Table 9: Continued. Plant part S. N. IUPAC names Common names Structure References (country)

(2E,7R,11R)-3,7,11,15- NH N Tetramethyl-2-hexadecen-1-yl ester-(3R,4S,21R)-14-ethyl-13- (132R)-132- N HN Leaves 193 formyl-21-hydroxy-21- [100] Hydroxypheophytin B (Taiwan) (methoxycarbonyl)-4,8,9,18- tetramethyl-20-oxo-3- O HO phorbinepropanoic acid O O OPhytyl O

O

(2E,7R,11R)-3,7,11,15- NH N Tetramethyl-2-hexadecen-1-yl ester-(3R,4S,21S)-14-ethyl-13- (132S)-132- Leaves 194 formyl-21-hydroxy-21- N HN [100] Hydroxypheophytin B (Taiwan) (methoxycarbonyl)-4,8,9,18- tetramethyl-20-oxo-3- phorbinepropanoic acid O HO O O OPhytyl O

(2E,7R,11R)-3,7,11,15- NH N Tetramethyl-2-hexadecen-1-yl ester-(3S,4S,21R)-9-ethenyl-14- HN Leaves 195 Pheophytin A N [100] ethyl-21-(methoxycarbonyl)- O (Taiwan) 4,8,13,18-tetramethyl-20-oxo-3- phorbinepropanoic acid PhytylO O O O

with estimated IC50 values of 30 and 2.5 𝜇g/mL, respectively. 122]. Many studies have indicated that B. pilosa could treat Taken together, the data indicate that the B. pilosa extract type 1 diabetes (T1D) and type 2 diabetes (T2D) in animals. and compound 54 act on human and mouse T cells. To Etiologically speaking, T1D is caused by autoimmune- test the in vivo effect of the B. pilosa extract and compound mediated destruction of pancreatic 𝛽 cells, leading to insulin 54, a zymosan-induced arthritis mouse model was used. deficiency, hyperglycemia, and complications. Currently, This model was established from B10.A/SgSnJ mice with an there is no cure for T1D. Polarization of Th cell differentiation 9 injection of zymosan (0.15 mg). The zymosan-injected mice controls the development of T1D. Suppression of Th1 cell received an intraperitoneal injection of the B. pilosa extract differentiation and promotion of Th2 cell differentiation (1,5,or10mg)atonedoseadayfor5days.Popliteallymph ameliorate T1D [123]. One study showed that the butanol node (PLN) weight was monitored to check the development fraction of B. pilosa inhibited T-cell proliferation, decreased of arthritis. The results revealed that 10 mg of the methanol Th1 cells and cytokines, and increased Th2 cells and extract of B. pilosa extract could significantly diminish cytokines, leading to prevention of T1D in nonobese inflammation as evidenced by PLN weight27 [ ]. This work diabetic (NOD) mice [49]. Based on a bioactivity-directed suggests that B. pilosa (and compound 54) can suppress isolation strategy, 3 polyynes, 2-𝛽-D-Glucopyranosyloxy immune response and inflammation. -1-hydroxytrideca-5,7,9,11-tetrayne (53), also known as cytopiloyne, 3-𝛽-D-Glucopyranosyl-1-hydroxy-6(E)- tetradecene-8,10,12-triyne (69), 2-𝛽-D- Glucopyranosyloxy- 3.3. Antidiabetic Activity. Anti-diabetic agents are primarily 1-hydroxy-5(E)-tridecene-7,9,11-triyne (50) were identified developed from plants and other natural resources [8, 24, 49, from B. pilosa [49, 53]. The IC50 value of the butanol fraction 51, 53]. B. pilosa isoneof1,200plantspeciesthathavebeen was 200 𝜇g/mL. This inhibition was reported to be partially investigated for antidiabetic activity [120, 121]. B. pilosa is used attributed to cytotoxicity because the butanol fraction at as an anti-diabetic herb in America, Africa, and Asia [51, 120, 38 Evidence-Based Complementary and Alternative Medicine

Table 10: Other compounds isolated from B. pilosa [30].

Plant part S.N. IUPAC names Common names Structure References (country) N N 3,7-Dihydro-1,3,7- N O Aerial 196 trimethyl-1H-purine- Caffeine [68] (Uganda) 2,6-dione N O O 1-((2R,4S,5R)- HO Tetrahydro-4-hydroxy- NH 5-(hydroxymethyl) Not found 197 Thymidine N O [37] furan-2-yl)-5- (China) methylpyrimidine- HO O 2,4(1H,3H)-dione S Roots 198 1-(2-Thienyl)-ethanone 2-Acetyl-thiophene [41] (Germany) O OH HO OH (2R,3S,4S,5S)-2- O (Heptan-2-yloxy)- HO tetrahydro-6- Heptanyl H O ((tetrahydro-3,4- 2-O-𝛽-xylofuranosyl- HO Whole 199 HO O [70] dihydroxy-5- (1 → 6)-𝛽- H OH (Taiwan) (hydroxymethyl)furan- glucopyranoside H H 2-yloxy)methyl)- 2H-pyran-3,4,5-triol

2-[(3R,7R,11R)-3- Hydroxy-3,7,11,15- O O tetramethylhexadecyl]- 𝛼-Tocopheryl Whole 200 [32] 3,5,6-trimethyl- quinone (Taiwan) 2,5-cyclohexadiene-1,4- dione HO

󸀠󸀠 󸀠󸀠 7-O-(4 ,6 -Diacetyl)- Leaves 201 𝛽-D- Not found [60] (China) glucopyranoside

180 𝜇g/mL could cause 50% death of Th1 cells. Moreover, were measured since Th1 cytokine IFN𝛾; and Th2 cytokine this study suggested that the butanol fraction may prevent IL-4 favor the production of IgG2a and IgE, respectively. As diabetes in NOD mice in vivo via downregulation of Th1 expected, high levels of IgE and some decline in the levels of cells or upregulation Th2 cells that have effects which are IgG2a were observed in the serum. Profiling of the butanol antagonistic of those of Th1 cells49 [ ]. This was proven by extract revealed five compounds, 3-𝛽-D-Glucopyranosyl- intraperitoneal injection of the butanol fraction at a dose 1-hydroxy-6(E)-tetradecene-8,10,12-triyne (69), 2-𝛽-D- of 3 mg/kg BW, 3 times a week, to NOD mice from 4 to 27 Glucopyranosyloxy-1-hydroxy-5(E)-tridecene-7,9,11-triyne weeks. This dosage resulted in lower incidence of diabetes (50), 4,5-Di-O-caffeoyl- quinic acid, 3,5-Di-O-caffeoylquinic (33%). At a dose of 10 mg/kg, the butanol fraction of B. acid, and 3,4-Di-O-caffeoylquinic acid. Only the first two pilosa totally eliminated (0%) the initiation of the disease. compounds showed similar effects on the prevention of To further support this result, assessment of IgG2a and IgE diabetes in NOD mice as the B. pilosa butanol fraction. production was performed in the serum of NOD mice. As Moreover, compound 50 showed greater activity than in vivo results obtained from intracellular cytokine staining, compound 69 in terms of enhancement (by 34% compared experiments were not very conclusive levels of IgG2a and IgE to 8%) of differentiation of Th0 to Th2 at 15 𝜇g/mL (both Evidence-Based Complementary and Alternative Medicine 39

Table 11: Chemical constituents of B. pilosa and their biological activities.

Molecular S.N. Name Classification Biological activities formula Anti-listerial [69, 91] 109 Centaureidin [91] Flavonoid C H O 18 16 8 Cytotoxic [10] Anti-listerial [69, 91] 110 Centaurein [91] Flavonoid C24H26O13 Cytotoxic [10] Anti-viral [105] Anti-viral [107, 108] Cytotoxic [77] 103 Luteolin [106] Flavonoid C H O 15 10 6 Anti-inflammatory [109] Anti-allergic [109] Anti-leishmanial [111] 82 Butein [110] Flavonoid C H O 15 12 5 Cytotoxic [78] 48 1,2-Dihydroxytrideca-5,7,9,11-tetrayne [112] Polyyne C13H12O2 Anti-angiogeneic [112] 46 1,3-Dihyroxy-6(E)-tetradecene-8,10,12-triyne [112] Polyyne C14H16O2 Anti-angiogeneic [112] Anti-angiogeneic [113] 45 1,2-Dihyroxy-5(E)-tridecene-7,9,11-triyne [113] Polyyne C H O 13 14 2 Anti-proliferative [113] Anti-microbial [115] Anti-malarial [3] 64 1-Phenylhepta-1,3,5-triyne [114] Polyyne C H 13 8 Cytotoxic [3] Antifungal [15] Anti-viral (100) 27 Linoleic acid [79] Fatty acid C H O 18 32 2 Cytotoxic [17] 161 Ethyl caffeate [84] Phenylpropanoid C11H12O4 Anti-inflammatory [84] Immunosuppressive and 54 2-O-𝛽-Glucosyltrideca-11(E)-en-3,5,7,9-tetrayn-1,2-diol [26] Polyyne C H O 19 20 7 Anti-inflammatory [26] Anti-diabetic [116] 53 2-𝛽-D-Glucopyranosyloxy-1-hydroxytrideca-5,7,9,11-tetrayne [50] Polyyne C H O 19 22 7 Anti-inflammatory [50] Anti-diabetic [28] 69 3-𝛽-D-Glucopyranosyl-1-hydroxy-6(E)-tetradecene-8,10,12-triyne [28] Polyyne C H O 20 26 7 Anti-inflammatory [94] Anti-diabetic [28] Anti-inflammatory [94] 50 2-𝛽-D-Glucopyranosyloxy-1-hydroxy-5(E)-tridecene-7,9,11-triyne [28] Polyyne C H O 19 24 7 Anti-malarial and antibacterial [47] 129 𝛽 Quercetin 3-O- -D-galactopyranoside [18] Flavonoid C21H20O12 Anti-inflammatory [117] Anti-viral [57] 170 3,5-Di-O-caffeoylquinic acid94 [ ] Phenylpropanoid C H O 25 24 12 Antioxidant [70] Anti-viral [57] 171 4,5-Di-O-caffeoylquinic acid94 [ ] Phenylpropanoid C H O 25 24 12 Antioxidant [70] Anti-viral [57] 169 3,4-Di-O-caffeoylquinic acid94 [ ] Phenylpropanoid C H O 25 24 12 Antioxidant [70] 󸀠 Quercetin 3,3 -dimethyl ether 126 Flavonoid C H O Anti-malarial [119] 7-O-𝛼-L-rhamnopyranosyl-(1 → 6)-𝛽-D-glucopyranoside [118] 21 20 11 125 󸀠 𝛽 Quercetin 3,3 -dimethyl ether-7-O- -D-glucopyranoside [118] Flavonoid C21H20O12 Anti-malarial [119] 70 1-Phenyl-1,3-diyn-5-en-7-ol-acetate [27] Polyyne C15H12O2 Anti-malarial [27] 199 𝛽 → 𝛽 Heptanyl 2-O- -xylofuranosyl-(1 6)- -glucopyranoside [70] Miscellaneous C18H44O10 Antioxidant [70] 124 3-O-Rabinobioside [70] Saccharide C27H38O15 Antioxidant [70] 130 Quercetin 3-O-rutinoside [70] Flavonoid C27H38O15 Antioxidant [70] 167 Chlorogenic acid [70] Phenolic C16H26O9 Antioxidant [70] 119 Jacein [70] Flavonoid C24H26O13 Antioxidant [70] Anti-malarial and 49 (R)-1,2-dihydroxytrideca-3,5,7,9,11-pentayne [47] Polyyne C H O 13 8 2 Antibacterial [47] 40 Evidence-Based Complementary and Alternative Medicine

Table 12: Structure and activity relationship studies of ethyl caffeate Table 15: Radical scavenging activity of secondary metabolites from using in vitro NF-𝜅B/DNA binding assays [84]. B. pilosa [70]. 𝜅 𝜇 Concentration NF- B/ Metabolite (Table 11)/control DPPH assay, IC50 ( g/mL) Compound 𝜇 ( M) DNA binding 199 Not determined 100% 124 Ethyl caffeate 50 5.3 inhibition 130 6.8 100% Ethyl 3,4-dihydroxyhydrocinnamate 100 167 10.5 inhibition 169 3.3 100% Catechol 400 171 3.8 inhibition 119 Not determined No Ethyl cinnamate 400 110 inhibition Not determined Quercetin 2.56 Caffeic acid 8.90 Table 13: Apoptosis in cocultures of T cells and pancreatic 𝛽 cells [53]. Table 16: Antioxidant activity of the essential oils and water extracts % apoptosis and from B. pilosa [92] Cell/medium necrosis Extract IC (𝜇g/mL) CD4+ T cells/control medium <4 50 Leaf essential oils 47 CD4+ T cells/with PBS-treated 𝛽 cells 2 of NOD-SCID mice Flower essential oils 50 CD4+ Tcells/withcytopiloyne-treated𝛽 cells 18 Leaf extract 61 CD4+ Tcells/withcytopiloyne-treated𝛽 cells in Flower extract 172 7 thepresenceof𝛼-FasL antibody CD8+ T cells/control medium 4 + with T1D incidence, cytopiloyne delayed and reduced the CD4 T cells/with PBS-treated 𝛽 cells + 4 of NOD-SCID mice invasion of CD4 Tcellsintothepancreaticislets[53]. In vitro study showed that cytopiloyne (53)inhibitedthe CD8+ Tcells/withcytopiloyne-treated𝛽 cells 4 + differentiation of¨ naıve Th (Th0) cells (i.e., CD4 Tcells)into CD8+ Tcells/withcytopiloyne-treated𝛽 cells in 4 Th1 cells and promoted differentiation of Th0 cells into Th2 thepresenceof𝛼-FasL antibody cells [50]. The in vitro data are consistent with the in vivo results indicating that cytopiloyne reduced Th1 differentiation Table 14: Radical scavenging activities of B. pilosa extracts [70]. and increased Th2 differentiation as shown by intracellular cytokine staining and FACS analysis [53]. In line with the NBT/hypoxanthine 𝛾 DPPH assay, skewing of Th differentiation, the level of serum IFN- and Extracts/control 𝜇 superoxide assay, IgG2c decreased while that of serum IL-4 and serum IgE IC50 ( g/mL) 𝜇 IC50 ( g/mL) increased compared to the negative controls (PBS-treated Quercetin 1.98 1.5 mice). Cytopiloyne also enhanced the expression of GATA- Ascorbic acid 6.34 Not determined 3, a master gene for Th2 cell differentiation, but not the 𝛼-tocopherol 8.97 Not determined expression of T-bet, a master gene for Th1 cell differentiation, further supporting its role in skewing Th differentiation53 [ ]. Ethyl acetate + 13.83 59.7 Also importantly, cytopiloyne partially depleted CD4 extract + rather than CD8 T cells in NOD mice [53]. As shown in Butanol extract 16.69 11.4 + Table 13, coculture assays showed that the depletion of CD4 > > Water extract 100 100 T cells was mediated through the induction of Fas ligand expression on pancreatic islet cells by cytopiloyne, leading to + apoptosis of infiltrating CD4 T cells in the pancreas via the compounds) and inhibition (by 40% compared to 10%) of Fas and Fas ligand pathways. However, cytopiloyne did not induce the expression of TNF-𝛼 in pancreatic islet cells and, differentiation to Th1 at the same concentration [49]. + Among the three polyynes found in B. pilosa,cytopiloyne thus, had no effect on CD8 Tcells[53]. (53) had the most potent anti-T1D activity [53]. To test the in In addition, Chang and colleagues showed that cytopi- vivo effect of cytopiloyne, NOD mice received intraperitoneal loyne dose-dependently inhibited T-cell proliferation stimu- 3 or intramuscular injection of cytopiloyne at 25 𝜇g/kg BW, lated by IL-2 plus Con A or anti-CD3 antibody, using [ H] 3 times per week. Twelve-week-old NOD mice started to thymidine incorporation assay [53]. develop T1D, and 70% of NOD mice aged 23 weeks and Overall, the mechanism of action of cytopiloyne and, over developed T1D. Remarkably, 12- to 30-week-old NOD probably, its derivatives in T1D includes inhibition of T-cell mice treated with cytopiloyne showed normal levels of blood proliferation, skewing of Th cell differentiation, and partial glucose (<200 mg/dL) and insulin (1-2 ng/mL). Consistent depletion of Th cells. Due to the anti-diabetic mechanisms Evidence-Based Complementary and Alternative Medicine 41

Table 17: Antibacterial activity of essential oils and flower extracts from B. pilosa [92].

Mean zone of inhibition (mm) Strain Leaf essential oil Flower essential oil Leaf extract Flower extract Micrococcus flavus 12.7 ± 0.3 8.7 ± 0.3 10.2 ± 0.2 10.8 ± 0.3 Bacillus subtilis 17.3 ± 1.9 11.7 ± 0.2 10.9 ± 0.2 10.3 ± 0.2 Bacillus cereus 19.0 ± 1.4 11.2 ± 0.3 11.8 ± 0.4 18.5 ± 1.0 Bacillus pumilus 12.3 ± 0.7 10.8 ± 0.2 10.5 ± 0.4 7.7 ± 0.2 Escherichia coli 13.7 ± 0.4 20.3 ± 0.7 10.2 ± 1.1 14.0 ± 1.3 Pseudomonas ovalis 12.5 ± 0.8 13.7 ± 1.5 10.2 ± 0.6 12.5 ± 0.6

Table 18: Antibacterial activity of root extracts from B. pilosa [93]. insulin action, or both [124]. A study by Ubillas et al. showed that the aqueous ethanol extract of the aerial part MIC (mg/mL) Strain 50 of B. pilosa at 1 g/kg body weight (BW) lowered blood Methanol extract Acetone extract glucose in db/db mice, a T2D mouse model [51]. Based Bacillus cereus 10 — on a bioactivity-guided identification, compounds 69 and Escherichia coli 5550 were identified. Further, the mixture of the compounds Klebsilla pneumonia 510(69 : 50) in a 2 : 3 ratio significantly decreased blood glucose Micrococcus kristinae 10 — concentration and reduced food intake on the second day Pseudomonas aeruginosa 10 10 of treatment when administered at doses of 250 mg/kg twice Staphylococcus aureus 510a day to C5BL/Ks-db/db mice. When tested at 500 mg/kg, Sraphylococcus epidermidis 55a more substantial drop in blood glucose level as well as the stronger anorexic effect (food intake reduced from Serratia marcescens 10 — 5.8 g/mouse/day to 2.5 g/mouse/day) was observed [51]. In Shigelea flexneri 10 — this study, it was suggested that the blood glucose lower- Streptococcus faecalis 10 — ing effect of B. pilosa was caused, in part, by the hunger suppressing effect of its polyynes [51]. However, the hunger Table 19: Antibacterial activity of B. pilosa of compound 29 [47]. suppressing effect of the ethanol extract of B. pilosa was 𝜇 not found in the studies described below. In another study Strain MIC50 ( g/mL) [24], water extracts of B. pilosa (BPWE) were used in Escherichia coli NIHJ 1 diabetic db/db mice, aged 6–8 weeks, with postprandial Escherichia coli ATCC25922 1 blood glucose levels of 350 to 400 mg/dL. Like oral anti- Klebsiella pneumoniae ATCC700603 128 diabetic glimepiride, which stimulates insulin release, one Serratia marcescens ATCC13880 16 single dose of BPWE reduced blood glucose levels from Pseudomonas aeruginosa ATCC27853 8 374 to 144 mg/dL. The antihyperglycemic effect of BPWE Staphylococcus aureus FDA209P 0.5 was inversely correlated to an increase in serum insulin Staphylococcus aureus ATCC29213 0.25 levels, suggesting that BPWE acts to lower blood glucose via Staphylococcus aureus N315 (MRSA) 0.5 increased insulin production. However, BPWE had different insulin secretion kinetics to glimepiride [24]. One flaw in Enterococcus faecalis ATCC29212 2 current anti-diabetics is their decreasing efficacy over time. Enterococcus faecalis NCTC12201 (VRE) 1 The authors investigated the long term anti-diabetic effect Bacillus subtilis ATCC6633 0.5 of BPWE in db/db mice. BPWE reduced blood glucose, Candida albicans ATCC10231 0.25 increased blood insulin, improved glucose tolerance, and reduced the percentage of glycosylated hemoglobin (HbA1c). Both long-term and one-time experiments strongly support of action, it was hypothesized that cytopiloyne protects the anti-diabetic action of BPWE [24]. In sharp contrast to NOD mice from diabetes by a generalized suppression of glimepiride, BPWE protected against islet atrophy in mouse adaptive immunity. To evaluate this hypothesis, ovalbumin pancreas. The investigators further evaluated anti-diabetic (Ova) was used as a T-cell dependent antigen to prime properties of 3 B. pilosa varieties, B. pilosa L. var. radiate NOD mice, which had already received cytopiloyne or (BPR), B.pilosa L. var. pilosa (BPP), and B. pilosa L. var. minor PBS vehicle. Ova priming boosted similar anti-Ova titers (BPM) in db/db mice [8]. One single oral dose (10, 50 and in cytopiloyne-treated mice and PBS-treated mice, but a 250 mg/kg body weight) of BPR, BPP, or BPM crude extracts difference in immunoglobulin isotype was observed in the decreased postprandial blood glucose levels in db/db mice two groups. Thus, it was concluded that cytopiloyne is an foruptofourhours,andthereductionofglucoselevelsin immunomodulatory compound rather than an immunosup- the blood appeared to be dose-dependent. Comparing the pressive compound [50, 53]. three variants, BPR extract resulted in a higher reduction in T2D is a chronic metabolic disease with serious com- blood glucose levels when administered at the same dose as plications resulting from defects in either insulin secretion, 42 Evidence-Based Complementary and Alternative Medicine

Table 20: Antifungal activity of B. pilosa [92].

Strain, % Inhibition Part/extract Concentration (ppm) Cortiicum rolfsii Fusarium solani Fusarium oxysporum Essential 100 85.7 ± 0.9 68.2 ± 0 74.5 ± 1.7 oils ± ± ± Leaves 250 96.0 0.8 77.9 1.8 87.9 0.4 Aqueous 100 44.6 ± 1.7 60.5 ± 2.1 71.6 ± 0.7 Extracts 250 94.2 ± 0.3 68.9 ± 0.7 82.4 ± 1.9 Essential 100 60.4 ± 0.9 89.2 ± 0.4 86.9 ± 0.5 oils ± ± ± Flowers 250 89.4 1.2 98.0 0.3 94.9 0.6 Aqueous 100 33.1 ± 1.1 71.4 ± 0.7 57.3 ± 2.2 Extracts 250 66.1 ± 1.4 91.2 ± 0 90.0 ± 0.7

Table 21: Antifungal activity of B. pilosa root extracts [93]. production involving the calcium/DAG/PKC𝛼 cascade in b cells. LC (mg/mL) Strain 50 The studies detailed above point to the conclusion that Acetone Methanol Water cytopiloyne and related polyynes (compounds 69 and 49) extracts extracts extracts are anti-diabetics in animal models. The data uncover a new Aspergillus niger 0.14 0.06 0.07 biological action of polyynes. It should be noted that, like Aspergillus flavus 10.91 6.58 0 all anti-diabetic drugs, cytopiloyne failed to prevent or cure Penicillium notatum 0.05 0.05 0.05 diabetes completely but reduced diabetic complications [125]. Intriguingly, 34 polyynes have been found in B. pilosa so far. It remains to be seen whether all the polyynes present in this plant have anti-diabetic activities. the other two varieties. In terms of serum insulin levels, a dose of 50 mg/kg of each extract was used, and the BPR extract, 3.4. Antioxidant Activity. Free radicals can damage cellular together with the three polyynes, significantly increased the components via a series of chemical reactions [89]leading serum insulin level in db/db mice. Long-term experiments to development and progression of cardiovascular disease, (28-day treatment) were then conducted using diabetic mice cancer, neurodegenerative diseases and ageing [70]. Free with postprandial glucose levels from 370 to 420 mg/dL, and radicals, nitric oxide (NO), and superoxide anions can be glimepiride was used as positive control. The range of dosages produced in macrophages to kill microbes. However, an applied was from 10 mg/kg BW to 250 mg/kg BW. Results excessive generation of the free radicals under pathological showed that the positive control as well as the crude extracts conditions is associated with a wide range of illnesses. ofthethreevarietiesloweredthebloodglucoselevelsin Plants are known to be rich in antioxidant phytochemicals. db/db mice. However, only BPR extract, containing a higher Chiangandcolleaguesevaluatedthefreeradicalscaveng- percentage of cytopiloyne (53), reduced blood glucose levels ing activity of crude extract, fractions, and compounds of andaugmentedbloodinsulinlevelsmorethanBPPandBPM. B. pilosa using 1,1-diphenyl-2-picrylhydrazyl (DPPH) and The percentage of glycosylated hemoglobin A1c (HbA1c) was hypoxanthine/xanthine oxidase assays [70]. Using DPPH also measured and found to be 7.9% ± 0.5%inmiceaged10– and hypoxanthine/xanthine oxidase assays, they found that 12 weeks, and 6.6% ± 0.2%, 6.1% ± 0.3%and6.2% ± 0.3%in the B. pilosa crude extract and the ethyl acetate, butanol, thebloodofage-matchedmicefollowingtreatmentwithBPR and water fractions had free radical scavenging activity. crude extract (50 mg/kg), glimepiride (1 mg/kg), and com- Nine compounds, Heptyl- 2-O-𝛽-xylofuranosyl-(1 → 6)-𝛽- pound 53 (0.5 mg/kg), respectively [8]. Among the polyynes glucopyranoside (199), 3-O-Rabinobioside (124), Quercetin found in B. pilosa, cytopiloyne was the most effective against 3-O-rutinoside (130), Chlorogenic acid (167), 3,4-Di-O- T2D. Hence, cytopiloyne was used for further study on anti- caffeoylquinic acid (169), 3,5-Di-O-caffeoylquinic acid (170), diabetic action and mechanism [125]. The data confirmed 4,5-Di-O-caffeoylquinic acid171 ( ), Jacein (119), and Cen- that cytopiloyne reduced postprandial blood glucose lev- taurein (110) had DPPH radical scavenging activity [70]. els, increased blood insulin, improved glucose tolerance, The IC50 values of the B. pilosa crude extract/fractions and suppressed HbA1c level, and protected pancreatic islets in compounds are summarized in Tables 14 and 15,respectively. db/db mice. Nevertheless, cytopiloyne failed to decrease Measurement of free radical scavenging activities is one blood glucose in streptoztocin (STZ)-treated mice whose b way of assessing the antioxidant activities of B. pilosa and cells were already destroyed. Additionally, cytopiloyne dose- its fractions and compounds. It is interesting that the ethyl dependently increased insulin secretion and expression in b acetate and butanol fractions are more active than the water cellsaswellascalciuminflux,diacylglycerol,andactivation fraction and B. pilosa crude extract [70]. Of the secondary ofproteinkinaseC𝛼. Collectively, the mechanistic studies metabolites, only phenolic compounds 124, 130, 167, 169,and suggest that cytopiloyne treats T2D via regulation of insulin 171 showed significant DPPH-radical scavenging activities. Evidence-Based Complementary and Alternative Medicine 43

Further analysis of the structure-activity relationship of the cells and sustains immunity against pathogens. Defects in compounds suggested that substitution of the C3 hydroxyl IFN-𝛾 expression, regulation, and activation result in vulner- group with glycosides increased the activity approximately ability to diseases caused by bacteria and viruses [69]. An 2-fold (for example, in compounds 124 and 130) relative to elegant study, performed by Chang and colleagues using IFN- quercetin (-OH in its C3) [70]. Further modification of the 𝛾 promoter-driven luciferase reporter construct in Jurkat structures of the active compounds needs to be performed to T cells, showed that hot water crude extracts of B. pilosa test the effects of various substituents on activity. The reason increased IFN-𝛾 promoter activity two-fold [69]. Out of the why most of the antioxidant compounds contain phenol subfractions of this extract, the butanol fraction, but not the moieties in their structure could be that the reduction- water or ethyl acetate, fractions, increased IFN-𝛾 promoter oxidation (redox) properties of phenols allow them to act activity six-fold. Centaurein (110) and centaureidin (109) as reducing agents, singlet oxygen quenchers, and hydrogen were identified from the butanol fraction and were stated to donors. causeafour-foldincreaseinIFN-𝛾 promoter activity with A complementary study by Muchuweti and colleagues EC50 values of 75 𝜇g/mL and 0.9 𝜇g/mL, respectively. The determined phenolic content, antioxidant activity, and the mechanism of action of centaurein was determined using phenolic profile of B. pilosa methanol extract [89]. They transcription factors such as AP-1, NFAT, and NF𝜅Bwhich estimated that the phenolic content of the methanol extract arereportedtobindtoIFN-𝛾 promoter and regulate IFN-𝛾 of B. pilosa was 1102.8 ± 2.2 mg/g [89]. Vanillin, hydroxyben- transcription. Unlike with the activity of the positive control, zaldehyde, caffeic acid, coumaric acid, and ferulic acid were PHA, centaurein caused a four-fold increase in NFAT, a found in this extract. The B. pilosa extract also showed DPPH 3-fold increase in NF𝜅Bandhadlittle,ifany,effecton radical scavenging activity. Furthermore, the antioxidant AP-1 enhancer activities (23-fold, three-fold, and ten-fold activity of the flavonoids found in B. pilosa was correlated increases, respectively, were seen with PHA) [69]. The authors with its hepatoprotective effects through their inhibition concluded that centaurein modulates IFN-𝛾 expression by the of NF-𝜅B activation which may lessen the oxidative stress NFAT and NF𝜅B pathways. The article only determined the caused by the production of free radicals during liver injury mechanism of action of centaurein. Its aglycone centaureidin [60]. This activity might also be due to the anti-inflammatory may act through the same mechanism though this conclusion effects of the aqueous extracts of B. pilosa aerial parts on the needstobefurtherverified. inhibition of COX-2 and PGE2 production [88]. B. pilosa extract and its compounds are reported to inhibit + Essential oils from B. pilosa flowers and leaves are also differentiation of¨ naıve CD4 helperT(Th0)cellsintoTh1 reported to possess antioxidant activity. With the aim of cells [49]. Using the Th cell differentiation assay as a screening replacing chemically synthesized additives, Deba and col- platform, 3 polyynes, 2-𝛽-D-glucopyranosyloxy-1 - leagues [92] worked on the antioxidant, antibacterial, and hydroxytrideca-5,7,9,11-tetrayne (53), 3-𝛽-D-glucopyranosyl- antifungal activities of essential oils and water extracts of B. 1-hydroxy-6(E)- tetradecene-8,10,12-triyne (69), and 2-𝛽-D- pilosa’s leaves and flowers. Table 16 summarizes the results glucopyranosyloxy-1-hydroxy-5(E)- tridecene-7,9,11-triyne obtained from DPPH free radical scavenging assay. (49) were discovered from B. pilosa [49, 53]. The data shows It can be inferred from Table 16 that essential oils from the that cytopiloyne and other two polyynes suppressed the leaves possessed the highest activity. It is reported elsewhere differentiation of type 1 helper T (Th1) cells and production that monoterpenes present in essential oils such that of B. of Th1 cytokines and promoted that of type 2 helper T (Th2) pilosa have protective effects and antioxidant properties92 [ ]. cells and production of Th2 cytokines, thus explaining the Beta-carotene bleaching method was also performed. Leaves immunomodulatory and anti-inflammatory effects of B. essential oils and aqueous extracts of the leaves and flowers pilosa and its polyynes. showed higher activity than the flower essential oils. This is Chang and colleagues were the first to report the effect of due to the volatility of the flower essential oils. The activity the butanol extract of B. pilosa on the autoimmune diabetes exhibited by the aqueous extracts was accounted to the and airway inflammation in mice [48]. Imbalance in the levels presence of phenolic compounds that are reported to donate of Th1 and Th2 and of various cytokines leads to autoimmune ahydrogenatomtofreeradicalssuchthatthepropagationof diseases. T1D and other autoimmune diseases (rheumatoid thechainreactionduringlipidoxidationisterminated[92]. arthritis, Crohn’s disease, among others) are exacerbated by + Overall, essential oils and phenolics present in B. pilosa can an increase Th1 levels (specifically, CD4 Th1 cells) while be thought of as major antioxidant compounds. Th2 cells antagonize this effect19 [ ]. Moreover, Th2 cells mediate asthma in ovalbumin-induced hypersensitivity in BALB/c mice [48]. In their work [48], Chang and colleagues 3.5. Immunomodulatory Activity. B. pilosa is thought to be showed that 10 mg/kg butanol extracts with 1.5% (w/w) an immunomodulatory plant and is reported to be effective compound 69 and 1.1% (w/w) compound 49 (Table 11) in the treatment of immune disorders such as allergy [75], ameliorated the development of Th1-mediated diabetes in arthritis [75], and T1D [48, 50, 53]. NOD mice through inhibition of 𝛽 cell death and leukocyte As pointed out in the discussion of its anti-diabetic infiltration. At the same dosage, the butanol extracts also activities (Section 3.3), a combination of phytochemicals exacerbated ovalbumin-induced pulmonary inflammation method and T-cell activation assays was used to study in BALB/c mice with an increase in the infiltration of immunomodulatory properties of B. pilosa.IFN-𝛾 is a key eosinophils and mast cells into the airway of the mice [48]. cytokine released by T and NK cells that mediates immune Despite the different outcomes, both mouse models proved 44 Evidence-Based Complementary and Alternative Medicine the concept that control over the Th1/Th2 shift is associated identifying structure- and bioactivity-related phytochemicals with autoimmune diseases and that the B. pilosa butanol from medicinal plants. extract can shift the differentiation of Th0 cells to Th2cells [48, 49]. 3.6. Antimalarial Activity. The use of chemical drugs against An extended study presented by Chiang and colleagues pathogens has resulted in drug-resistant mutants. Examples showed that compound 53 modulates T-cell functions [50]. + of drug resistance can be found in the species of the Using CD4 T cells from BALB/c mice, they demonstrated Plasmodium that cause malaria. It is important to search for that cytopiloyne decreased levels of IFN-𝛾 producing cells new compounds to combat Plasmodium parasites [47]. A (Th1) by 12.2% (from 72% to 59.8%). Since Th1 and Th2 cell study of the anti-malarial activity of the leaf extracts of B. differentiation is antagonistic, it was expected that compound pilosa using a combination of phytochemistry and bioassays 53 increased the percentage of mouse IL-4 producing cells showed that compound 49 (Table 11)showedactivityagainst (Th2) by 7.2% (from 23.7% to 30.9%). Subsequent assess- amalariaparasite(P. fal c ip ar um NF54 strain) with an IC50 mentofeffectofcompound53 on the modulation of the value of 6.0 𝜇g/mL [77]. In addition, compound 49,isolated transcription of IL-4 and IFN-𝛾 showed that cytopiloyne, as from the aerial parts of B. pilosa, inhibited growth of the P. expected, decreased the splenocyte levels of IFN-𝛾 mRNA falciparum FCR-3 strain with an IC50 value of 0.35 𝜇g/mL. and increasing that of IL-4 in a dose-dependent manner. In This compound was tested for its in vivo effect in mice the range of 0.1 to 3 𝜇g/mL of compound 53,theeffectswere infected with P. b e rg he i NK-65 strain. Results showed that not attributed to its cytotoxicity. Consequently, using 3 𝜇g/mL compound 49 decreased the average parasitemia in the red cytopiloyne for 72 and 96 hours, the protein concentration of blood cells by 20.7 (from 32.8% of that of the control to IFN-𝛾 decreased to 18.6% and 44.4%, respectively. Under the 12.1%) after an intravenous injection of 0.8 mg/kg BW/day for same conditions, cytopiloyne increased IL-4 concentrations four days [47]. Further studies addressing the anti-malarial to 198.5% and 247.0% (for 72 and 96 hours, resp.). This mechanism underlying both polyynes and clinical studies are modulation of T-cell differentiation exhibited by compound needed. 53 was used to explain its anti-diabetic activity [50]. The anti-diabetic role of cytopiloyne was extensively discussed above (Section 3.3, anti-diabetic activity). The 3.7. Antibacterial Activity. Emergence of multiple antibiotic- molecular basis of the regulation of cytokine expression by resistantmicrobesisbecomingaglobalthreattopublichealth cytopiloyne has been described. Cytopiloyne directly elevated andachallengetodiseasetreatment.Forinstance,penicillin the expression level of IL-4 via GATA-3 upregulation in is commonly used to combat a food-borne intracellular Tcells[53]. However, reduction of IFN-𝛾 expression in T bacterium Listeria;however,penicillin-resistantbacteriahave cells seemed to come from the indirect opposing effect IL-4 been discovered recently [69]. Chang and coworkers iso- cytokinebecausetheexpressionlevelofT-betwasunaltered lated centaurein (110) and centaureidin (109)fromB. pilosa [53]. In this way, cytopiloyne skewed Th1 polarization into the extract [69, 70]. Centaurein enhances expression of IFN-𝛾, Th2 state, conferring protection against T1D in NOD mice. a key cytokine for macrophage activation and, consequently, Aside from polarization of Th cell differentiation, cytopiloyne enhances bactericidal activity in macrophages [69, 70]. In also activated the expression of Fas ligand in pancreatic agreement with observed in vitro effects, centaurein was b cells, this increase leading to the partial depletion of T reported to prevent and treat Listeria infection in C57BL/6J cells and reduction of immune response in local areas such mice [91]. Mechanistic studies confirmed that centaurein as the pancreas. Of note, cytopiloyne also inhibited T-cell exerted antilisterial action via IFN-𝛾 expression in wild-type proliferation and activation. By targeting T cells from three mice but not IFN-𝛾 knockout mice [91]. Further in vitro immunomodulatory actions, cytopiloyne protects against studies on centaurein showed that this compound increased T1D and probably other Th1-mediated autoimmune diseases IFN-𝛾 expression by 13% (from 17% to 20%), 20% (from 21% + + [19]. to 41%), and 11% (from 6% to 17%) in CD4 Tcells,CD8 T The phytochemical constituents of B. pilosa exert their cells, and NK cells, respectively. That is to say, there was functions on different immune cells to modulate immune an increase in IFN-𝛾 producing immune cells. As expected, response. It is possible that some of the compounds may centaurein also enhanced the expression level of T-bet, a key have agonistic or antagonistic effects on immune response. nuclear factor for IFN-𝛾 expression. Consistently, centaurein Immune function of B. pilosa may depend on its compo- augmentedtheserumIFN-𝛾 levels in C57BL/6J mice, and this sition and amount of compounds, which could explain the augmentation peaked 24 hours after compound injection. apparently conflicting report of B. pilosa butanol extract The quantity of mouse serum IFN-𝛾 was sufficient to activate aggravating allergy in mice [48] while cellulosine-treated macrophages in vitro and eradicated GFP-producing Listeria extract ameliorated allergy [48, 75]. IFN-𝛾 promoter reporter inside macrophages. However, the entry of Listeria into assays and T-cell differentiation assays were used to isolate macrophages was not affected by centaurein-treated mouse 2 flavonoids [69] and 3 polyynes [49, 50]asimmunomod- sera. Centaurein treatment at 20 𝜇g per mouse rescued 30% ulatory compounds from the butanol fraction of B. pilosa. ofthemiceinfectedwithalethaldoseofListeria (2 × 6 Interestingly, the flavonoids promote IFN-𝛾 expression in 10 CFU). It is noteworthy that in the presence of ampicillin NK and T cells. In marked contrast, the polyynes promote (5 𝜇g/mouse), centaurein (20 𝜇g/mouse) rescued 70% of the IL-4 expression and indirectly inhibit IFN-𝛾 expression in mice, suggesting an additive effect between ampicillin and differentiating T cells. Sensitivity appears to be the key to centaurein [91]. Despite lower abundance, centaureidin was Evidence-Based Complementary and Alternative Medicine 45

30 times more active than centaurein in terms of IFN-𝛾 appeared to have better fungicidal activity than water extracts production [91]. as summarized in Table 20. Aside from the indirect antibacterial action mentioned Another study by Ashafa and colleagues showed that above, extract and/or compounds of B. pilosa also showed acetone, methanol, and water extracts of the B. pilosa roots direct bacteriostatic and/or bactericidal action. One study showed antifungal activities against Aspergillus niger, A. reported that essential oils and leaf/flower extracts of B. flavus, and Penicillium notatum using the agar dilution pilosa could suppress the growth of gram positive and gram method.TheresultsaretabulatedinTable21 [93]. Negative negative bacteria as evidenced by zone of inhibition assays. controls showed 0% growth inhibition. The methanol extract In this study, antibacterial activity of the essential oils from of the B. pilosa rootsat10mg/mLwasalsoeffectiveagainst the leaves and flowers of B. pilosa was determined in an Candida albicans [93]. Of note, B. pilosa obtained from Papua attempt to identify natural products as food preservatives for New Guinea had no activity against A. niger and C. albicans prevention of microbial multiplication and food oxidation. [126], but the South African (Eastern Cape) ecotype exhibited The essential oils and extracts of B. pilosa leaves and flow- moderate activity against C. albicans [93]. This discrepancy ers showed moderate but different extents of antibacterial may depend on extraction solvents, extraction procedure, activity (Table 17). In general, essential oils had higher assay techniques, different plant parts, and abundance of antibacterial activity than crude extracts. One explanation for active compounds. this could be that monoterpenes in the essential oils destroy B. pilosa produces a variety of secondary metabolites cellular integrity and, subsequently, inhibit the respiration such as flavonoids, phenylacetylenes, alkaloids, sterols, ter- and ion transport processes. The presence of antibacterial penoids, and tannis [92, 93]. However, none of them have 𝛽-caryophyllene could be another explanation as reported been confirmed as active compounds against fungi. Further elsewhere [92]. Another study reported that the methanol investigation of active compounds from B. pilosa is necessary and acetone extract of B. pilosa roots displayed antibacterial to further understand the antifungal efficacy of this plant. activities against the bacteria listed in Table 18 [93]and methanol extracts from the roots seemed to be the most effective. 3.9. Hypotensive and Vasodilatory Activities. In early studies, Another study indicated that the polyyne, (R)-1,2- Dimoandcolleaguesusedthreeratmodels,normotensive dihydroxytrideca-3,5,7,9,11-pentayne (49), from this plant Wistar rats (NTR), salt-loading hypertensive rats (SLHR), also suppressed bacterial growth as shown by the minimum and spontaneous hypertensive rats (SHR) to investigate the inhibitory concentration required to inhibit 50% bacterial hypotensive effect of the methanol crude extract of B. pilosa growth (MIC50)inTable19. This compound was highly leaves [28, 127]. The extract lowered systolic blood pressure effective against several Gram positive and Gram negative in hypertensive rats (SLHR and SHR) to a greater degree bacteria including the drug-resistant bacteria Staphylococcus than NTR [28, 127]. In addition, a decrease in urinary aureus N315 (MRSA) and Enterococcus faecalis NCTC12201 sodiumionsandanincreaseinurinarypotassiumionswere (VRE) [47]. Strikingly, compound 49 had a similar MIC50 observed after treatment with the methanol extract of B. value to antibiotics (ampicillin, tetracycline, norfloxacin, and pilosa leaves although neither differences were statistically amphotericin B) in most of the bacteria tested. significant28 [ , 127]. Taking the data together, the study Antibacterial activity of B. pilosa extracts and compo- proposed that B. pilosa leaf extract reduced blood pressure nents, expressed as MIC50 and the mean zone of inhibition, via vasodilation [28, 127]. The same group continued to is tabulated in Tables 17, 18,and19. The zone of inhibition for test the antihypertensive effect of aqueous and methylene Ampicilline (positive control) ranges from 15.3 ± 0.3 mm to chloride extracts of B. pilosa leaves in a hypertensive rat 44.3 ± 0.2 mm [92]. model [28, 127]. To establish a fructose-induced hypertension model, male Wistar rats were given 10% fructose solution to drink ad libitum for three weeks. In addition to free access to 10% fructose, the rats were treated with the aqueous 3.8. Antifungal Activity. B. pilosa has traditionally been used (150 mg/kg) or methyl chloride (350 mg/kg) extracts of B. to treat microbial infection. Recently, different parts of B. pilosa for additional three weeks [28, 127]. Both extracts of pilosa have been tested for antifungal activities. Deba and B. pilosa leaves had a hypotensive effect on rats. However, colleagues first evaluated the antifungal effect of the hot neither extracts reversed the elevation of serum insulin in water extracts of the B. pilosa roots, stems, and leaves against fructose-fed rats. Therefore, B. pilosa lowered blood pressure Corticium rolfsii, Fusarium solani, and Fusarium oxysporum. irrespective of insulin [28, 127]. To better understand the They discovered that C. rolfsii was most suppressed by hypotensive mechanism, the authors investigated the effect treatment with B. pilosa as its growth was reduced at almost of a neutral extract of B. pilosa (NBP), a mixture of methanol all the tested doses, followed by F. oxysporum and F. solani and methylene chloride (1 : 1) extract after neutralization with [99]. However, the fungicidal activities of the stems, and NaOHandHCl,ontheheartandthebloodpressureof roots were greater than the leaves [99]. Moreover, the same NTR and SHR [110]. This study showed that an intravenous group assessed the antifungal activity of the essential oils injection of the NBP resulted in a biphasic reduction in and aqueous extracts from B. pilosa flowers and leaves [92]. systolic blood pressure. In addition, one intravenous dose of They showed that the extracts and oils had antifungal activity the extract at 10, 20, and 30 mg/kg BW decreased systolic against C. rolfsii, F. solani,andF. oxysporum.Essentialoils blood pressure in normal rats by 18.3%, 42.5%, and 30%, 46 Evidence-Based Complementary and Alternative Medicine respectively,andthesamedosesreducedthebloodpressure the effect of methanol, cyclohexane, and methyl chloride in hypertensive rats by 25.8%, 38.9%, and 28.6%, respectively. extracts of B. pilosa on gastric ulcers in Wistar rats fed Only the highest dose (30 mg/kg) affected the force of the with 1 mL HCl/ethanol gastric necrotizing solution (150 mM contraction of the heart. Atropine and propranolol were used HCl in 60% ethanol), and macroscopically visible lesions to interfere with the hypotensive action of the NBP. Atropine were scored [26]. Among the three extracts, methylene chlo- reduced the initial phase of the hypotensive response in NBP ride extracts exhibited the highest activity showing 46.4% and completely abolished the second phase of hypotensive inhibition of lesion formation at a dose of 500 mg/kg BW response in NBP. In contrast, propranolol increased the and complete inhibition at 750 mg/kg [26]. The efficacy of first hypotensive response but partially abolished the second theethylenechlorideextractwasfollowedbythatofthe hypotensive response provoked by the NBP [110]. This mech- methanol extracts which had inhibition ranging from 30.4% anistic study suggested that B. pilosa invokes the biphasic to 82.2% at concentrations of 500 mg/kg and 1000 mg/kg hypotensive responses via targeting cardiac pump efficiency BW, respectively [26]. The cyclohexane extracts showed the during the first phase and vasodilation at the second phase lowest activity against gastric ulcers in rats with 13.3%, [110]. 40%, and 79.7% inhibition at 500, 750, and 1000 mg/kg BW, A further study was performed to investigate the relaxing respectively [26]. To better understand the mode of action effect of a neutral extract of B. pilosa (NBP) on rat aorta con- of the methylene chloride extract of B. pilosa,ratswere tractedwithKCl(60mM)andnorepinephrine(0.1mM)[94]. pretreated with indomethacin, a COX-2 inhibitor involved in Cumulative addition of NBP relaxed the rat aorta previously prostaglandin synthesis. Pretreatment significantly reduced contracted by KCl in a dose-dependent manner. The EC50 the protection against HCl/ethanol-induced ulcers to 31.3% value of the NBP for vasorelaxation was 0.32 mg/mL. The data inhibition at 750 mg/kg BW, suggesting a link between the also showed that the NBP reduced the contraction of aorta antiulcerative activity of B. pilosa and prostaglandin syn- previously contracted by KCl irrespective of the presence of thesis. Unexpectedly, the methylene chloride extract of B. aortic endothelium [94]. pilosa showed little gastric mucosal protection against gastric + Pretreatment with glibenclamide, an ATP-dependent K lesions induced by 95% ethanol (1 mL) [26]. Absolute alcohol channel blocker, did not considerably affect the relaxant effect is known to cause mucosal/submucosal tissue destruction via of the NBP on KCl-induced contraction, suggesting that cellular necrosis and the release of tissue-derived mediators the vasodilatory effect of B. pilosa was not related to the (histamine and leukotriene C4). Thus, these data imply that B. + opening of this ATP-dependent K channel [94]. On the pilosa did not prevent the generation or the necrotic action of otherhand,inthepresenceofindomethacinorpyrilamine these mediators on the gastric microvasculature. In addition, maleate,therelaxantresponseinducedbytheplantextract pylorus ligation can increase gastric acid secretion without was significantly inhibited at the lower concentrations. The an alteration of mucosal histamine content. The methylene plant extract was able to reduce the aorta resting tone, inhibit chloride extract of B. pilosa did not possess antisecretory the KCl-induced contractions by 90% at 1.5 mg/mL and the activity. On the contrary, it was observed that increases in the CaCl2-induced contractions by 95% at 0.75 mg/mL. These dose of the extract led to elevated gastric juice acidity [26]. results demonstrate that B. pilosa can act as a vasodilator Results with both absolute ethanol and pylorus ligation rat probably via acting as a calcium antagonist [94]. models suggested the possibility that the ineffectiveness of B. However, no specific compound for the above activity pilosa against gastric ulcers was due to lack of antihistaminic has been identified from B. pilosa to date. A bioactivity- activity in the plant. In summary, overall the data suggest that guided identification approach may be adopted to identify B. pilosa protects against HCL/ethanol-mediated ulcers via theactivecompoundsinB. pilosa that possess hypotensive inhibition of prostaglandin biosynthesis. and vasodilatory effects and understand their mechanism of Previous phytochemical studies showed that a group action. of flavonoids, acyclichalcones, are present in B. pilosa [129, 130] and the chalcones were proposed to have anti- ulcerative activity [131]. Moreover, nine hydroxychalcones 10 3.10. Wound Healing Activity. B. pilosa has been traditionally were reported to possess gastric cytoprotective effects with used to treat tissue injury in Cameroon, Brazil, and Venezuela 2,4-dihydroxychalcone being the most active [132]. Since [26]. Hassan and colleagues investigated the wound healing methylene chloride extracts appear to be the most active B. potential of B. pilosa in Wistar rats [128]. Mirroring the pilosa extracts, next, the specific anti-ulcerative phytochemi- positive control neomycin sulfate, the ethanol extract of B. cals in the methylene chloride extracts of B. pilosa and their pilosa hadfasterwoundclosurethancontrolrats3,6,and9 modes of action needs to be probed. days after topical application. Histological examination also Despite the claims listed in Table 3, relatively few scientific revealed better collagenation, angiogenesis, and organization studies have been conducted in vitro and in vivo to address the of wound tissue seven days after application. Epithelialization traditional ethnomedical uses of B. pilosa. Information about and total healing time in B. pilosa-treated rats were compara- the use of B. pilosa as a botanical therapy recorded so far is ble to those of neomycin sulfate. Together, these data suggest far from complete. Studies conducted thus far only serve as that B. pilosa may be a viable alternative to neomycin lotion a starting point for further investigation of B. pilosa, and the for the treatment of wounds. ultimate efficacious use of the herb in clinical applications. In addition to studying the wound healing effect of B. pilosa on external ulcers, Tan and colleagues also examined Evidence-Based Complementary and Alternative Medicine 47

4. Toxicology Acknowledgments Despiteitsuseasaningredientinfoodforhumancon- The authors thank their laboratory members for construc- sumption, studies on systemic toxicity (e.g., acute, subacute, tive suggestions and the authors whose publications they chronic and subchronic toxicities) of B. pilosa in humans cited for their contributions. The authors also thank Ms and animals are still inadequate and insufficient. So far, Miranda Loney of Academia Sinica for English editing of this acute, and/or subchronic toxicities have been evaluated in paper. This work was supported by Grants 99-CDA-L11 and rats and mice. Oral acute and 28-day toxicities of water 101S0010056 from Academia Sinica, Taiwan. Arlene P. Bar- extract of B. pilosa leaveswereevaluatedinWistarrats tolome is a recipient of MECO-TECO Sandwich Scholarship [133]. An oral dose of water extract of B. pilosa leaves at Program between Taiwan and the Philippines. 10 g/kg BW showed no obvious mortality or changes in theappearanceinrats[134]. The same extract at 0.8 g/kg References BW/day, once a day, showed no obvious sub-chronic toxicity in rats over 28 days, as measured by survival rate, body [1]T.Shen,G.H.Li,X.N.Wang,andH.X.Lou,“Thegenus weight, and gross examination of organs [134]. These data Commiphora: a review of its traditional uses, phytochemistry are consistent with our data indicating that oral delivery and pharmacology,” Journal of Ethnopharmacology,vol.142,no. of the water extract of the B. pilosa whole plant at 1 g/kg 2, pp. 319–330. BW/day, once a day, is safe in rats over 28 days (unpublished [2] C. Long, P. Sauleau, B. David et al., “Bioactive flavonoids of data). Taken together, these studies suggest that ingestion of Tanacetum parthenium revisited,” Phytochemistry,vol.64,no. B. pilosa aqueous extract at up to at 1 g/kg BW/day, once 2, pp. 567–569, 2003. aday,ishighlysafeinrats.Inaddition,theacutetoxicity [3]P.O.KarisandO.Ryding,Asteraceae Cladistics and Classifica- of aqueous and ethanol extracts of B. pilosa in mice have tion,BremerK.Eds,pp.559–569,Timberpress,Portland,Ore, been reported [134]. Five- to six-week-old mice with weights USA, 1994. between 28 and 35 g received a peritoneal injection of both [4]O.N.Pozharitskaya,A.N.Shikov,M.N.Makarovaetal., extracts at the different doses. The LD50, the dose that causes “Anti-inflammatory activity of a HPLC-fingerprinted aqueous 50% lethality, of the aqueous and ethanol extracts in mice infusion of aerial part of Bidens tripartita L,” Phytomedicine,vol. was 12.30 g/kg BW and 6.15 g/kg BW, respectively [134]. A 17,no.6,pp.463–468,2010. complete toxicological study has not been completed for [5] F. Q. Oliveira, V. Andrade-Neto, A. U. Krettli, and M. humans. Furthermore, the drug interactions of B. pilosa with G. L. Brandao,˜ “New evidences of antimalarial activity of other drugs are unknown. Further safety verification and Bidens pilosa roots extract correlated with polyacetylene and clinical trials should be performed before B. pilosa can be flavonoids,” Journal of Ethnopharmacology,vol.93,no.1,pp.39– considered for medicinal use. 42, 2004. [6] Agriculture USDA, “Plants database,” 2013, in: Natural Resources Conservation Service, http://www.nrcs.usda.gov/wps/portal/nrcs/site/national/home . [7] Y. Zhou and X. Z. Yan, “Experimental study of qi deficiency 5. Conclusions syndrome and Codonopsis pillosulae and Astragalus injection ontheimmuneresponseinmice,”Zhong Xi Yi Jie He Za Zhi, B. pilosa is an erect, perennial plant with green leaves, white vol. 9, no. 5, pp. 286–262, 1989. or yellow flowers and tiny black seeds. As it is distributed [8] S. C. Chien, P. H. Young, Y. J. Hsu et al., “Anti-diabetic worldwide and is widely used as a folk remedy, B. pilosa properties of three common Bidens pilosa variants in Taiwan,” can be thought of as an extraordinary source of food and Phytochemistry, vol. 70, no. 10, pp. 1246–1254, 2009. medicine. However, a comprehensive up-to-date review of [9] M. J. Alcaraz and M. J. Jimenez, “Flavonoids as anti- research on B. pilosa hashithertobeenunavailable.Inthis inflammatory agents,” Fitoterapia, vol. 59, no. 1, pp. 25–38, 1988. article, scientific studies on B. pilosa have been summarized [10] FAO, Agriculture Food and Nutrition for Africa—A Resource and critically discussed from the perspectives of botany, Book for Teachers of Agriculture, Publishing Management ethnomedicine, phytochemistry, pharmacology, and toxicol- Group, FAO Information Division, Rome, Italy, 1997. ogy. B. pilosa is claimed to treat more than 40 disorders, [11] M. B. Rokaya, Z. Munzbergova, B. Timsina, and K. R. Bhattarai, and201compoundshavebeenidentifiedfromthisplant. “Rheum australe D. Don: a review of its botany, ethnobotany, The medicinal utility of B. pilosa and its modes of action phytochemistry and pharmacology,” Journal of Ethnopharma- in relation to its known phytochemicals were discussed cology,vol.141,no.3,pp.761–774,2012. herein. Polyynes, flavonoids, phenylpropanoids, fatty acids, [12]P.H.Young,Y.J.Hsu,andW.C.Yang,“Bidens pilosa L. and and phenolics are the primary bioactive compounds of B. its medicinal use,” in Recent Progress in Medicinal Plants Drug pilosa, and they have been reported to be effective in the Plant II,A.S.Awaad,V.K.Singh,andJ.N.Govil,Eds.,Standium treatment of tumors, inflammation/immune modulation, Press, Houston, Tex, USA, 2010. diabetes, viruses, microbes, protozoans, gastrointestinal dis- [13] C. Ge, “Cytologic study of Bidens bipinnata L,” Zhongguo Zhong eases, hypertension, and cardiovascular diseases. Caution Yao Za Zhi,vol.15,no.2,pp.72–125,1990. should be exercised in the therapeutic use of B. pilosa for [14]L.C.Chiang,J.S.Chang,C.C.Chen,L.T.Ng,andC.C. hypoglycemia, hypotension, bleeding, and allergy. Lin, “Anti-herpes simplex virus activity of Bidens pilosa and 48 Evidence-Based Complementary and Alternative Medicine

Houttuynia cordata,” American Journal of Chinese Medicine,vol. [32] C. K. Lee, “The low polar constituents from Bidens pilosa L. var. 31, no. 3, pp. 355–362, 2003. minor (blume) sherff,” Journal of the Chinese Chemical Society, [15]N.P.Rybalchenko,V.A.Prykhodko,S.S.Nagornaetal., vol. 47, no. 5, pp. 1131–1136, 2000. “In vitro antifungal activity of phenylheptatriyne from Bidens [33] M. C. Zulueta, M. Tada, and C. Y. Ragasa, “A diterpene from cernua L. against yeasts,” Fitoterapia,vol.81,no.5,pp.336–338, Bidens pilosa,” Phytochemistry,vol.38,no.6,pp.1449–1450, 2010. 1995. [16]K.Redl,W.Breu,B.Davis,andR.Bauer,“Anti-inflammatory [34]J.Wang,H.Yang,Z.W.Lin,andH.D.Sun,“Flavonoidsfrom active polyacetylenes from Bidens campylotheca,” Planta Med- Bidens pilosa var. radiata,” Phytochemistry,vol.46,no.7,pp. ica,vol.60,no.1,pp.58–62,1994. 1275–1278, 1997. [17] M. Ayyanar and S. Ignacimuthu, “Traditional knowledge of [35] A. Zhao, Q. Zhao, L. Peng et al., “Anew chalcone glycoside from Kani tribals in Kouthalai of Tirunelveli hills, Tamil Nadu, India,” Bidens pilosa,” Acta Botanica Yunnanica,vol.26,no.1,pp.121– Journal of Ethnopharmacology,vol.102,no.2,pp.246–255,2005. 126. [18] P. Geissberger and U. Sequin, “Constituents of Bidens pilosa L.: [36] P. Gao, Y. Wu, S. Cui, and Y. Zou, “Synthesis and absolute do the components found so far explain the use of this plant in configuration of the 7-phenylhepta-4, 6-diyne-1, 2-diol isolated traditional medicine?” Acta Tropica,vol.48,no.4,pp.251–261, from Bidens pilosa,” Synthesis, no. 13, pp. 2131–2135, 2011. 1991. [37] S. Wang, B. Yang, D. Zhu, D. He, and L. Wang, “Active [19] A. K. Abbas, K. M. Murphy, and A. Sher, “Functional diversity of components of Bidens pilosa L,” Zhongcaoyao,vol.36,pp.20– helper T lymphocytes,” Nature,vol.383,no.6603,pp.787–793, 21. 1996. [38] F. Bohlmann, T. Burkhardt, and C. Zdero, Naturally Occuring [20] J. H. Cano and G. Volpato, “Herbal mixtures in the traditional Acetylenes, Academic Press, New York, NY, USA, 1973. medicine of Eastern Cuba,” JournalofEthnopharmacology,vol. [39]T.M.Sarg,A.M.Ateya,N.M.Farrag,andF.A.Abbas, 90, no. 2-3, pp. 293–316, 2004. “Constituents and biological activity of Bidens pilosa L. grown [21] J. Namukobe, J. M. Kasenene, B. T. Kiremire et al., “Traditional in Egypt,” Acta Pharmaceutica Hungarica,vol.61,no.6,pp.317– plants used for medicinal purposes by local communities 323, 1991. around the Northern sector of Kibale National Park, Uganda,” [40] H. A. L. ValdesandH.P.Rego,“´ Bidens pilosa Linne,”´ Revista Journal of Ethnopharmacology, vol. 136, no. 1, pp. 236–245, 2011. Cubana de Plantas Medicinales,vol.6,pp.28–33. [22] C. Lans, “Comparison of plants used for skin and stomach [41] F. Bohlmann, H. Bornowski, and K. M. Kleine, “New polyynes problems in Trinidad and Tobago with Asian ethnomedicine,” from the tribe Heliantheae,” Chemische Berichte,vol.97,pp. Journal of Ethnobiology and Ethnomedicine,vol.3,article3,2007. 2135–2138. [23] S. Dharmananda, “APopular Remedy Ecapes Notice of Western [42] R. Wang, Q. X. Wu, and Y.P.Shi, “Polyacetylenes and flavonoids Practitioners,” 2013, http://www.itmonline.org/arts/bidens.htm from the aerial parts of Bidens pilosa,” Planta Medica,vol.76,no. . 9,pp.893–896,2010. [43] L. W. Wu, Y. M. Chiang, H. C. Chuang et al., “A novel [24] Y. J. Hsu, T. H. Lee, C. L. T. Chang, Y. T. Huang, and W. C. Yang, polyacetylene significantly inhibits angiogenesis and promotes “Anti-hyperglycemic effects and mechanism of Bidens pilosa apoptosis in human endothelial cells through activation of the water extract,” Journal of Ethnopharmacology,vol.122,no.2,pp. CDK inhibitors and caspase-7,” Planta Medica,vol.73,no.7,pp. 379–383, 2009. 655–661, 2007. [25] E. Noumi, F. Houngue, and D. Lontsi, “Traditional medicines [44] L. W. Wu, Y. M. Chiang, H. C. Chuang et al., “Polyacetylenes in primary health care: plants used for the treatment of function as anti-angiogenic agents,” Pharmaceutical Research, hypertension in Bafia, Cameroon,” Fitoterapia,vol.70,no.2,pp. vol. 21, no. 11, pp. 2112–2119, 2004. 134–139, 1999. [45]H.L.Yang,S.C.Chen,N.W.Changetal.,“Protectionfrom [26] P. V. Tan, T. Dimo, and E. Dongo, “Effects of methanol, oxidativedamageusingBidens pilosa extracts in normal human cyclohexane and methylene chlo ride extracts of Bidens pilosa erythrocytes,” Food and Chemical Toxicology,vol.44,no.9,pp. on various gastric ulcer models in rats,” Journal of Ethnophar- 1513–1521, 2006. macology,vol.73,no.3,pp.415–421,2000. [46] H. Q. Wang, S. J. Lu, H. Li, and Z. H. Yao, “EDTA-enhanced [27] R. L. C. Pereira, T. Ibrahim, L. Lucchetti, A. J. R. Da Silva, and V. phytoremediation of lead contaminated soil by Bidens maxi- L. G. De Moraes, “Immunosuppressive and anti-inflammatory mowicziana,” JournalofEnvironmentalSciences,vol.19,no.12, effects of methanolic extract and the polyacetylene isolated pp.1496–1499,2007. from Bidens pilosa L,” Immunopharmacology,vol.43,no.1,pp. [47] S. Tobinaga, M. K. Sharma, W.G. L. Aalbersberg et al., “Isolation 31–37, 1999. and identification of a potent antimalarial and antibacterial [28] T. Dimo, J. Azay, P. V. Tan et al., “Effects of the aqueous and polyacetylene from Bidens pilosa,” Planta Medica,vol.75,no.6, methylene chloride extracts of Bidens pilosa leaf on fructose- pp. 624–628, 2009. hypertensive rats,” Journal of Ethnopharmacology,vol.76,no.3, [48] C. L. T. Chang, H. K. Kuo, S. L. Chang et al., “The distinct pp.215–221,2001. effects of a butanol fraction of Bidens pilosa plant extract on [29] C. Wiart, Medicinal Plants of Southeast Asia, Pelanduk, Selabgor the development of Th1-mediated diabetes and Th2-mediated Darul Ehsan, Malaysia, 2000. airway inflammation in mice,” Journal of Biomedical Science, [30] F. L. Silva, D. C. H. Fischer, J. F. Tavares, M. S. Silva, P. F. vol.12,no.1,pp.79–89,2005. De Athayde-Filho, and J. M. Barbosa-Filho, “Compilation of [49] S. L. Chang, C. L. T. Chang, Y. M. Chiang et al., “Polyacetylenic secondary metabolites from Bidens pilosa L,” Molecules,vol.16, compounds and butanol fraction from Bidens pilosa can mod- no. 2, pp. 1070–1102, 2011. ulate the differentiation of helper T cells and prevent autoim- [31]A.H.Chen,S.R.Lin,andC.H.Hong,“Phytochemicalstudy mune diabetes in non-obese diabetic mice,” Planta Medica,vol. on Bidens pilosa L. var. minor,” Huaxue,pp.38–42,1975. 70,no.11,pp.1045–1051,2004. Evidence-Based Complementary and Alternative Medicine 49

[50]Y.M.Chiang,C.L.T.Chang,S.L.Chang,W.C.Yang,andL. [68]S.D.Sarker,B.Bartholomew,R.J.Nash,andN.Robinson,“5- F. Shyur, “Cytopiloyne, a novel polyacetylenic glucoside from O-methylhoslundin: an unusual flavonoid from Bidens pilosa Bidens pilosa, functions as a T helper cell modulator,” Journal of (Asteraceae),” Biochemical Systematics and Ecology,vol.28,no. Ethnopharmacology,vol.110,no.3,pp.532–538,2007. 6, pp. 591–593, 2000. [51] R.P.Ubillas,C.D.Mendez,S.D.Joladetal.,“Antihyperglycemic [69] S. L. Chang, Y. M. Chiang, C. L. T. Chang et al., “Flavonoids, acetylenic glucosides from Bidens pilosa,” Planta Medica,vol. centaurein and centaureidin, from Bidens pilosa,stimulateIFN- 66,no.1,pp.82–83,2000. 𝛾 expression,” Journal of Ethnopharmacology,vol.112,no.2,pp. [52] L. Alvarez, S. Marquina, M. L. Villarreal, D. Alonso, E. Aranda, 232–236, 2007. and G. Delgado, “Bioactive polyacetylenes from Bidens pilosa,” [70]Y.M.Chiang,D.Y.Chuang,S.Y.Wang,Y.H.Kuo,P.W. Planta Medica,vol.62,no.4,pp.355–357,1996. Tsai, and L. F.Shyur, “Metabolite profiling and chemopreventive [53] C. L. T. Chang, S. L. Chang, Y. M. Lee et al., “Cytopiloyne, a bioactivity of plant extracts from Bidens pilosa,” Journal of polyacetylenic glucoside, prevents type 1 diabetes in nonobese Ethnopharmacology,vol.95,no.2-3,pp.409–419,2004. diabetic mice,” Journal of Immunology,vol.178,no.11,pp.6984– [71] A. Kusano, Y. Seyama, E. Usami et al., “Studies on the antioxi- 6993, 2007. dant active constituents of the dried powder from Bidens pilosa [54]G.Kusano,A.Kusano,andY.Seyama,Novel Hypoglycemic L. var. radiata Sch,” Natural Medicines,vol.57,no.3,pp.100–104, and Anti-Inflammatory Polyacetylenic Compounds, Their Com- 2003. positions, Bidens Plant Extract Fractions, and Compositions [72] I. Doleckovˇ a,´ L. Rarov´ a,´ J. Gruz´ et al., “Anti-proliferative and Containing the Plant or Fraction,JPO,Tokyo,Japan,2004. anti-angiogeniceffectsofflavoneeupatorin,anactivecon- [55] M. G. L. Brandao,A.U.Krettli,L.S.R.Soares,C.G.C.˜ stituent of chloroform extract of Orthosiphon stamineus leaves,” Nery, and H. C. Marinuzzi, “Antimalarial activity of extracts Fitoterapia,vol.83,no.6,pp.1000–1007. and fractions from Bidens pilosa and other Bidens species [73]K.Bairwa,R.Kumar,R.J.Sharma,andR.K.Roy,“Anupdated (Asteraceae) correlated with the presence of acetylene and review on Bidens pilosa L,” Der Pharma Chemica,vol.2,no.3, flavonoid compounds,” Journal of Ethnopharmacology,vol.57, pp. 325–337. no. 2, pp. 131–138, 1997. [74] Q. Xia, Y. Liu, and Y. Li, “Determination of hyperoside in [56] A. U. Krettli, V. F. Andrade-Neto, M. D. G. L. Brandao,˜ and different parts and different species of Herba Bidens by RP- W. M. S. Ferrari, “The search for new anti-malarial drugs from HPLC,” West China Journal of Pharmaceutical Sciences,vol.50, plants used to treat fever and malaria or plants ramdomly pp. 48–53. selected: a review,” Memorias do Instituto Oswaldo Cruz,vol.96, [75] M. Horiuchi and Y. Seyama, “Improvement of the antiinflam- no. 8, pp. 1033–1042, 2001. matory and antiallergic activity of Bidens pilosa L. var. radiata [57] C. K. Lee, “The low polar constituents from Bidens pilosa L. var. SCHERFF treated with enzyme (Cellulosine),” Journal of Health minor (blume) sherff,” Journal of the Chinese Chemical Society, Science,vol.54,no.3,pp.294–301,2008. vol. 47, no. 5, pp. 1131–1136, 2000. [76] M. G. L. Brandao,C.G.C.Nery,M.A.S.Mam˜ ao,˜ and A. [58]C.K.Wat,R.K.Biswas,E.A.Graham,L.Bohm,G.H.N. U. Krettli, “Two methoxylated flavone glycosides from Bidens Towers, and E. R. Waygood, “Ultraviolet-mediated cytotoxic pilosa,” Phytochemistry, vol. 48, no. 2, pp. 397–400, 1998. activity of phenylheptatriyne from Bidens pilosa L,” Journal of [77]P.Kumari,K.Misra,B.S.Sisodiaetal.,“Apromisinganticancer Natural Products, vol. 42, no. 1, pp. 103–111, 1979. and antimalarial component from the leaves of Bidens pilosa,” [59] Y. Sashida, K. Ogawa, M. Kitada, H. Karikome, Y. Mimaki, Planta Medica,vol.75,no.1,pp.59–61,2009. and H. Shimomura, “New aurone glucosides and new phenyl- [78] G. Seelinger, I. Merfort, U. Wolfle,¨ and C. M. Schempp, “Anti- propanoid glucosides from Bidens pilosa,” Chemical and Phar- carcinogenic effects of the flavonoid luteolin,” Molecules,vol.13, maceutical Bulletin,vol.39,no.3,pp.709–711,1991. no.10,pp.2628–2651,2008. [60]L.P.Yuan,F.H.Chen,L.Lingetal.,“Protectiveeffectsoftotal [79] G. Seelinger, I. Merfort, and C. M. Schempp, “Anti-oxidant, flavonoids of Bidens pilosa L. (TFB) on animal liver injury and anti-inflammatory and anti-allergic activities of luteolin,” liver fibrosis,” Journal of Ethnopharmacology,vol.116,no.3,pp. Planta Medica,vol.74,no.14,pp.1667–1677,2008. 539–546, 2008. [80] C. C. Yit and N. P. Das, “Cytotoxic effect of butein on human [61] B. Hoffmann and J. Holzl,¨ “Acylated compounds from Bidens colon adenocarcinoma cell proliferation,” Cancer Letters,vol. pilosa,” Planta Medica,vol.55,pp.108–109. 82, no. 1, pp. 65–72, 1994. [62] B. Hoffmann and J. Holzl,¨ “A methylated chalcone glucoside [81] J. A. Beutler, E. Hamel, A. J. Vlietinck et al., “Structure-activity from Bidens pilosa,” Phytochemistry,vol.27,no.11,pp.3700– requirements for flavone cytotoxicity and binding to tubulin,” 3701, 1988. Journal of Medicinal Chemistry,vol.41,no.13,pp.2333–2338, [63] B. Hoffmann and J. Holzl,¨ “Chalcone glucosides from Bidens 1998. pilosa,” Phytochemistry,vol.28,no.1,pp.247–250,1989. [82] M. R. Kviecinski, K. B. Felipe, T. Schoenfelder et al., “Study of [64] B. Hoffmann and J. Holzl,¨ “New chalcones from Bidens pilosa,” the antitumor potential of Bidens pilosa (Asteraceae) used in Planta Medica,vol.54,no.1,pp.52–54. Brazilian folk medicine,” Journal of Ethnopharmacology,vol.117, [65] B. Hoffmann and J. Holzl,¨ “Weitere acylierte chalkone aus no. 1, pp. 69–75, 2008. Bidens pilosa,” Planta Medica, vol. 54, pp. 450–451, 1988. [83]P.Sundararajan,A.Dey,A.Smith,A.G.Doss,M.Rajappan, [66] P. Geissberger and U. Sequin, “Constituents of Bidens pilosa L.: and S. Natarajan, “Studies of anticancer and antipyretic activity 11 do the components found so far explain the use of this plant in of Bidens pilosa whole plant,” African Health Sciences,vol.6,no. traditional medicine?” Acta Tropica,vol.48,no.4,pp.251–261, 1, pp. 27–30, 2006. 1991. [84] Y.M.Chiang,C.P.Lo,Y.P.Chenetal.,“Ethylcaffeatesuppresses [67] V.V.Pham,V.V.T.KP,V.L.Hoang,andV.K.Phan,“Flavonoid NF-𝜅B activation and its downstream inflammatory mediators, compounds from the plant Bidens pilosa L., (Asteraceae),” Tap iNOS, COX-2, and PGE2 in vitro or in mouse skin,” British Chi Duoc Hoc,vol.50,pp.48–53. Journal of Pharmacology,vol.146,no.3,pp.352–363,2005. 50 Evidence-Based Complementary and Alternative Medicine

[85] J. S. Kim, H. J. Lee, M. H. Lee, J. Kim, C. Jin, and J. H. [99]F.Deba,T.D.Xuan,M.Yasuda,andS.Tawata,“Herbicidaland Ryu, “Luteolin inhibits LPS-stimulated inducible nitric oxide fungicidal activities and identification of potential phytotoxins synthase expression in BV-2 microglial cells,” Planta Medica, from Bidens pilosa L. var. radiata Scherff: research paper,” Weed vol.72,no.1,pp.65–68,2006. Biology and Management,vol.7,no.2,pp.77–83,2007. [86] P. A. Ruiz and D. Haller, “Functional diversity of flavonoids [100] J. K. Kumar and A. K. Sinha, “Anew disubstituted acetylacetone in the inhibition of the proinflammatory NF-𝜅B, IRF, and Akt from the leaves of Bidens pilosa Linn,” Natural Product Research, signaling pathways in murine intestinal epithelial cells,” Journal vol.17,no.1,pp.71–74,2003. of Nutrition,vol.136,no.3,pp.664–671,2006. [101] K. Ogawa and Y. Sashida, “Caffeoyl derivatives of a sugar [87] A. Xagorari, A. Papapetropoulos, A. Mauromatis, M. lactone and its hydroxy acid from the leaves of Bidens pilosa,” Economou, T. Fotsis, and C. Roussos, “Luteolin inhibits Phytochemistry,vol.31,no.10,pp.3657–3658,1992. an endotoxin-stimulated phosphorylation cascade and [102]Q.Xia,Y.Liu,andY.Li,“Determinationofgallicacidfrom proinflammatory cytokine production in macrophages,” different species and different medical parts of Herba Bidens by Journal of Pharmacology and Experimental Therapeutics,vol. RP-HPLC,” West China Journal of Pharmaceutical Sciences,vol. 296, no. 1, pp. 181–187, 2001. 24, no. 03, pp. 308–310. [88] N. Yoshida, T. Kanekura, Y. Higashi, and T. Kanzaki, “Bidens [103]W.J.Lee,L.F.Wu,W.K.Chen,C.J.Wang,andT.H. 𝛽 pilosa suppresses interleukin-1 -induced cyclooxygenase-2 Tseng, “Inhibitory effect of luteolin on hepatocyte growth fac- expression through the inhibition of mitogen activated protein tor/scatter factor-induced HepG2 cell invasion involving both kinases phosphorylation in normal human dermal fibroblasts,” MAPK/ERKs and PI3K-Akt pathways,” Chemico-Biological Journal of Dermatology,vol.33,no.10,pp.676–683,2006. Interactions,vol.160,no.2,pp.123–133,2006. [89] M. Muchuweti, C. Mupure, A. Ndhlala, T. Murenje, and M. A. [104]J.A.Beutler,J.H.Cardellina,C.M.Lin,E.Hamel,G.M. N. Benhura, “Screening of antioxidant and radical scavenging Cragg, and M. R. Boyd, “Centaureidin, a cytotoxic flavone from activity of Vigna ungiculata, Bidens pilosa and Cleome gynan- Polymnia fruticosa, inhibits tubulin polymerization,” Bioorganic dra,” American Journal of Food Technology,vol.2,no.3,pp.161– and Medicinal Chemistry Letters,vol.3,no.4,pp.581–584,1993. 168, 2007. [105]A.Verma,A.Su,A.M.Golin,B.O’Marrah,andJ.K.Amorosa, [90]L.P.Yuan,F.H.Chen,L.Lingetal.,“Protectiveeffectsoftotal “The lateral view: a screening method for knee trauma,” Aca- flavonoids of Bidens bipinnata L. against carbon tetrachloride- demic Radiology,vol.8,no.5,pp.392–397,2001. induced liver fibrosis in rats,” Journal of Pharmacy and Pharma- [106] J. Corren, M. Lemay, Y. Lin, L. Rozga, and R. K. Randolph, cology,vol.60,no.10,pp.1393–1402,2008. “Clinical and biochemical effects of a combination botanical [91] S. L. Chang, H. H. Yeh, Y. S. Lin, Y. M. Chiang, T. K. Wu, product (ClearGuard) for allergy: a pilot randomized double- and W. C. Yang, “The effect of centaurein on interferon- 𝛾 blind placebo-controlled trial,” Nutrition Journal,vol.7,no.1, expression and Listeria infection in mice,” Toxicology and article 20, 2008. Applied Pharmacology,vol.219,no.1,pp.54–61,2007. [107]M.S.Gachet,J.S.Lecaro,M.Kaiseretal.,“Assessmentofanti- [92] F. Deba, T. D. Xuan, M. Yasuda, and S. Tawata, “Chemical com- protozoal activity of plants traditionally used in Ecuador in the position and antioxidant, antibacterial and antifungal activities treatment of leishmaniasis,” Journal of Ethnopharmacology,vol. of the essential oils from Bidens pilosa Linn. var. Radiata,” Food 128,no.1,pp.184–197,2010. Control,vol.19,no.4,pp.346–352,2008. [108] W. C. Yang, M. Ghiotto, B. Barbarat, and D. Olive, “The role of [93] A. O. T. Ashafa and A. J. Afolayan, “Screening the root extracts Tec protein-tyrosine kinase in T cell signaling,” The Journal of from Biden pilosa L. var. radiata (Asteraceae) for antimicrobial Biological Chemistry,vol.274,no.2,pp.607–617,1999. potentials,” Journal of Medicinal Plant Research,vol.3,no.8,pp. 568–572, 2009. [109] S. Tewtrakul, H. Miyashiro, N. Nakamura et al., “HIV-1 inte- grase inhibitory substances from Coleus parvifolius,” Phytother- [94] T. B. Nguelefack, T. Dimo, E. P. Nguelefack Mbuyo, P. V. Tan, apy Research,vol.17,no.3,pp.232–239,2003. S. V. Rakotonirina, and A. Kamanyi, “Relaxant effects of the neutral extract of the leaves of Bidens pilosa linn on isolated rat [110]T.Dimo,T.B.Nguelefack,P.V.Tanetal.,“Possiblemechanisms vascular smooth muscle,” Phytotherapy Research,vol.19,no.3, of action of the neutral extract from Bidens pilosa L. leaves on pp.207–210,2005. the cardiovascular system of anaesthetized rats,” Phytotherapy Research, vol. 17, no. 10, pp. 1135–1139, 2003. [95] M. T. Grombone-Guaratini, K. L. Silva-Brandao,˜ V.N. Solferini, J. Semir, and J. R. Trigo, “Sesquiterpene and polyacetylene [111] T. S. C. Li, Chinese and Related North American Herbs,CRC profile of the Bidens pilosa complex (Asteraceae: Heliantheae) Press, New York, NY, USA, 2002. from Southeast of Brazil,” Biochemical Systematics and Ecology, [112] H. Wu, H. Chen, X. Hua, Z. Shi, L. Zhang, and J. Chen, vol.33,no.5,pp.479–486,2005. “Clinical therapeutic effect of drug-separated moxibustion on [96] L. L. Lin, C. Y. Wu, H. C. Hsiu, M. T. Wang, and H. Chuang, chronicdiarrheaanditsimmunologicmechanisms,”Journal of “Studies on diabetes mellitus. I. The hypoglycemic activity of Traditional Chinese Medicine,vol.17,no.4,pp.253–258,1997. phytosterinonalloxandiabeticrats,”Taiwan Yi Xue Hui Za Zhi, [113] S. W. Wright, R. R. Harris, J. S. Kerr et al., “Synthesis, chemical, vol. 66, no. 2, pp. 58–65, 1967. and biological properties of vinylogous hydroxamic acids: dual [97] M. A. N. Benhura and I. C. Chitsiku, “The extractable 𝛽- inhibitors of 5-lipoxygenase and IL-1 biosynthesis,” Journal of carotene content of Guku (Bidens pilosa)leavesaftercooking, Medicinal Chemistry,vol.35,no.22,pp.4061–4068,1992. drying and storage,” International Journal of Food Science and [114] W. R. Almiron´ and M. E. Brewer, “Classification of immature Technology,vol.32,no.6,pp.495–500,1997. stage habitats of Culicidae (Diptera) collected in Cordoba, [98] J. S. Chang, L. C. Chiang, C. C. Chen, L. T. Liu, K. C. Wang, Argentina,” Memorias do Instituto Oswaldo Cruz,vol.91,no.1, andC.C.Lin,“AtileukemicactivityofBidens pilosa l. var. pp.1–9,1996. minor (blume) sherff and Houttuynia cordata thunb,” American [115] N. L. Wang, J. Wang, X. S. Yao, and S. Kitanaka, “Two new Journal of Chinese Medicine,vol.29,no.2,pp.303–312,2001. monoterpene glycosides and a new (+)-jasmololone glucoside Evidence-Based Complementary and Alternative Medicine 51

from Bidens parviflora Willd,” Journal of Asian Natural Products [132] K. Yamamoto, H. Kakegawa, H. Ueda et al., “Gastric cytopro- Research,vol.9,no.5,pp.473–479,2007. tective anti-ulcerogenic actions of hydroxychalcones in rats,” [116] P. Champagnat, “Role of the terminal bud in the action Planta Medica,vol.58,no.5,pp.389–393,1992. exercised by the cotyledon of Bidens pilosus L. var. radiatus on [133] J. O. C. Ezeonwumelu, A. K. Julius, C. N. Muhoho et al., its axillary bud,” ComptesRendusdesSeances´ et Memoires´ de la “Biochemical and histological studies of aqueous extract of Societ´ edeBiologie´ , vol. 145, no. 17-18, pp. 1374–1376, 1951. Bidens pilosa leaves from Ugandan Rift valley in rats,” British [117] S. F. Nielsen, S. B. Christensen, G. Cruciani, A. Kharazmi, Journal of Pharmacology and Toxicology,vol.2,no.6,pp.302– and T. Liljefors, “Antileishmaniai chalcones: statistical design, 309. synthesis, and three-dimensional quantitative structure-activity [134] L. Frida, S. Rakotonirina, A. Rakotonirina, and J. P. Savineau, relationship analysis,” Journal of Medicinal Chemistry,vol.41, “In vivo and in vitro effects of Bidens pilosa L. (Asteraceae) no.24,pp.4819–4832,1998. leaf aqueous and ethanol extracts on primed-oestrogenized rat [118] Y. C. Hwang, J. J. H. Chu, P. L. Yang, W. Chen, and M. V. Yates, uterine muscle,” African Journal of Traditional, Complementary “Rapid identification of inhibitors that interfere with poliovirus and Alternative Medicines,vol.5,no.1,pp.79–91,2008. replication using a cell-based assay,” Antiviral Research,vol.77, no. 3, pp. 232–236, 2008. [119]V.F.Andrade-Neto,M.G.L.Brandao,˜ F. Q. Oliveira et al., “Antimalarial activity of Bidens pilosa L. (Asteraceae) ethanol extracts from wild plants collected in various localities or plants cultivated in humus soil,” Phytotherapy Research,vol.18,no.8, pp.634–639,2004. [120]R.J.MarlesandN.R.Farnsworth,“Anti-diabeticplantsand their active constituents,” Phytomedicine,vol.2,no.2,pp.137– 189. [121]M.Habeck,“Diabetestreatmentsgetsweethelpfromnature,” Nature Medicine,vol.9,no.10,p.1228,2003. [122] H. W. Lin, G. Y. Han, and S. X. Liao, “Studies on the active constituents of the Chinese traditional medicine Polygonatum odoratum (Mill.) Druce,” Acta Pharmaceutica Sinica,vol.29,no. 3, pp. 215–222, 1994. [123]C.L.T.Chang,Y.C.Chen,H.M.Chen,N.S.Yang,and W. C. Yang, “Natural cures for type 1 diabetes: a review of phytochemicals, biological actions, and clinical potential,” Current Medicinal Chemistry,vol.20,no.7,pp.899–907,2013. [124]L.Dey,A.S.Attele,andC.S.Yuan,“Alternativetherapiesfor type 2 diabetes,” Alternative Medicine Review,vol.7,no.1,pp. 45–58, 2002. [125] C.L.T.Chang,H.Y.Liu,T.F.Kuoetal.,“Anti-diabeticeffectand mode of action of cytopiloyne,” Evidence-Based Complementary and Alternative Medicine,vol.2013,ArticleID685642,13pages, 2013. [126]M.R.Khan,M.Kihara,andA.D.Omoloso,“Anti-microbial activity of Bidens pilosa, Bischofia javanica, Elmerillia papuana and Sigesbekia orientalis,” Fitoterapia,vol.72,no.6,pp.662–665, 2001. [127] T. Dimo, T. B. Nguelefack, P. Kamtchouing, E.´ Dongo, A. Rakotonirina, and S. V. Rakotonirina, “Hypotensive effects of methanol extract from Bidens pilosa Linn on hypertensive rats,” ComptesRendusdel’AcademiedesSciences.SerieIII,vol.322, no. 4, pp. 323–329, 1999. [128]A.K.Hassan,O.Deogratius,J.F.Nyafuono,O.Francis,andO. P. Engeu, “Wound healing potential of the ethanolic extracts of Bidens pilosa and Ocimum suave,” African Journal of Pharmacy and Pharmacology,vol.5,no.2,pp.132–136,2011. [129] B. Hoffmann and J. Holzl,¨ “Chalcone glucosides from Bidens pilosa,” Phytochemistry,vol.28,no.1,pp.247–250,1989. [130] B. Hoffmann and J. Holzl, “New chalcones from Bidens pilosa,” Planta Medica,vol.54,no.1,pp.52–54. [131] C. Kandaswami and E. Middleton, “Free radical scavenging and antioxidant activity of plant flavonoids,” Advances in Experi- mental Medicine and Biology,vol.366,pp.351–376,1994. 52 Evidence-Based Complementary and Alternative Medicine

Composition Comments

1. Please provide valid postal code to the address if possible.

2. We made the highlighted change in Table 4 for the sake of correctness. Please check.

3. Please check the correctness of the highlighted part in Table 5.

4. We considered the highlighted part in Table 11 as reference. Please check similar cases throughout.

5. Please check the format of Table 18.

6. We reformatted Table 20. Please check.

7. We made the highlighted change in Table 4 for the sake of consistency. Please check similar cases throughout the paper.

8. We made the highlighted change(s) for the sake of consistency. Please check simila highlighted cases throughout.

9. We made the highlighted change in Table 9 for the sake of clarity. Please check similar cases through out.

10. We made the highlighted change(s) for the sake of correctness. Please check.

11. Comment on ref. [66]: This reference is a repetition of [18]. Please check similar cases throughout. Author(s) Name(s)

                                        

Author 1 Author 3       !"     ( )* #   $  #   + Author 2       %" #   & '

             ,-*. ,  -     *  ." ,-*.                       /         "

0   ,-*.1       2    33 "" 3  3   %   0 4 5         4   ,-*. 4       " 0 4  4    ,-*.                " ,    1    ,-*.       %   0 4 5    "