Sei sulla pagina 1di 22

Current Pharmaceutical Analysis, 2009, 5, 47-68 47

Noteworthy Secondary Metabolites Naphthoquinones – their Occurrence,


Pharmacological Properties and Analysis

Petr Babula1, Vojtech Adam2,3, Ladislav Havel4 and Rene Kizek2,*

1
Department of Natural Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Palackeho
1-3, CZ-612 42 Brno, Czech Republic; 2Department of Chemistry and Biochemistry, 3Department of Animal Nutrition
and Forage Production, and 4Department of Plant Biology, Faculty of Agronomy, Mendel University of Agriculture and
Forestry, Zemedelska 1, CZ-613 00 Brno, Czech Republic

Abstract: Chemical investigation of many bacterial and fungal, as well as plant species has revealed the presence of in-
teresting compounds derived from naphthalene – 1,4-naphthoquinones and rarely also 1,2-naphthoquinones. They were
detected in many species of families Bignoniaceae, Droseraceae, Plumbaginace, Boraginaceae, Juglandaceae as well as
in species of small families, such as Dioncophyllaceae or Acanthaceae. Naphthoquinones have very interesting spectrum
of biological actions, including antibiotic, antiviral, anti-inflammatory, antipyretic, antiproliferative and cytotoxic effects.
Because of these properties the plants containing them are used in folk medicines, mainly by natives in Asia, where espe-
cially Chinese medicine uses aerial as well as subterranean parts of these plants for hundreds years, and South America.
The utilizing of naphthoquinones for medicinal purposes and their occurrence in nature is reviewed and discussed.
Moreover, we review analytical techniques using for their analysis.

Keywords: Naphthoquinones, Plants, Pharmacology, Liquid chromatography, Electrochemical detection, UV-VIS Spectrome-
try.

INTRODUCTION parasitic and cytotoxic activities emerge due to their ability


to act as potent inhibitors of electron transport, as uncouplers
Naphthoquinones are one of the groups of secondary me-
of oxidative phosphorylation, as intercalating agents in the
tabolites widespread in nature. The most important higher DNA double helix, as bioreductive alkylating agents of bio-
plant families containing naphthoquinones are Avicenniaceae
molecules, and as producers of reactive oxygen radicals by
[1], Bignoniaceae [2, 3], Boraginaceae [4], Droseraceae [5,
redox cycling under aerobic conditions. Naphthoquinones,
6], Ebenaceae [7, 8], Juglandaceae [9], Nepenthaceae [10]
especially juglone, are widely studied for allelopathic activ-
and Plumbagnaceae [11, 12], they have been determined as
ity. Plants with naphthoquinone content are world-wide used
secondary metabolism products of actinomycetes (Strepto-
in the traditional medicines of countries where they grow.
myces) [13] and fungi (Fusarium, Marasmius, Verticillium) Naphthoquinones are highly toxic. Moreover, 1,4-naphtho-
[14] lichens and algae [15]. In plants, they commonly occur
quinone has been evaluated as a corrosion inhibitor in 0.5 M
in the reduced and glycosidic forms. In some species (e.g.
NaCl solutions. This effect is a result of its adsorption on the
Diospyros, Ebenaceae) are naphthoquinones present as
metal surface and blocking the corrosion process [18]. High
monomers as well as dimers or trimers. They are biosynthe-
performance liquid chromatography with spectrometric de-
sized via a variety of pathways including acetate and
tection and electrochemical techniques are most suitable for
malonate pathway (plumbagin), shikimate/succinyl CoA their determination in biological samples.
combined pathway (lawsone) and shikimate/mevalonate
pathway (alkannin). Napthoquinones have many physiologi- CHARACTERIZATION OF NAPHTHOQUINONES
cal roles – ubiquinone, plastoquinone and K vitamins are
functional constituents of biochemical systems. Naphthoqui- Chemical structure of monomeric naphthoquinones is
nones are usually coloured, especially they are responsible based on bicyclic system – naphthalene skeleton substitute in
for yellow or brown colour and thus, they play important position C1 and C4 (1,4-naphthoquinones) or C1 and C2 (1,2-
roles as dyes in pigmentation. The interest of many investi- naphthoquinones). Dimeric and trimeric naphthoquinones
gators in these compounds is due to their broad-range of bio- are also known. Most of them are coloured compounds.
logical activities: antibacterial, fungicidal, antiparasitic and Their colours usually vary between yellow, orange and
insecticidal. In addition, they have inhibitory effect on insect brown.
larval development and sedative or toxic effect on aquatic Almost all naphthoquinones are soluble in alcohol, ace-
organisms and animals [16, 17]. Their antimicrobial, anti- tone, chloroform, benzene, DMSO and acetic acid, plum-
bagin and juglone are slightly soluble in hot water. The sim-
plest, and in the nature the most widespread naphthoqui-
*Address correspondence to this author at the Department of Chemistry and nones, are juglone, lawsone, plumbagin and lapachol (Table
Biochemistry, Mendel University of Agriculture and Forestry, Czech Re-
public; Tel: +420-5-4513-3350; Fax: +420-5-4521-2044; 1).
E-mail: kizek@sci.muni.cz

1573-4129/09 $55.00+.00 © 2009 Bentham Science Publishers Ltd.


48 Current Pharmaceutical Analysis, 2009, Vol. 5, No. 1 Babula et al.

Table 1. Structural Formulas, Systematic and Trivial Names and Physical Properties of the Certain 1,4-naphthoquinones

Substituent Systematic (Trivial) Name Molecular Weight Melting Point [°C]

- 1,4-naphthoquinone 158.15 119 – 122

5-hydroxy-1,4-naphthoquinone
R3 = -OH 174.14 155
(juglone)

2-hydroxy-1,4-naphthoquinone
R1 = -OH 174.15 192 – 195
(lawsone)

5-hydroxy-2-methy-1,4-
R1 = -CH3, R3 = -OH naphthoquinone 188.18 78 – 79

R6 O
(plumbagin)
R5 R1 2-methyl-1,4-naphthoquinone
R1 = -CH3 172.18 105 – 107
R4 R2
(menadion)
R3 O
5,8-dihydroxy-1,4-naphthoquinone
R3, R6 = -OH 190.16 237
(naphtazalin)

2,5,7-trihydroxy-1,4-naphthoquinone
R1, R3, R5 = -OH 206.17 164 – 168.5
(flaviolin)

R1 = - 5,8-dihydroxy-2-(1-hydroxy-4-methyl-
CHOHCH2CH=C(CH3) 2 pent-3-enyl)-1,4-naphthoquinone 288.30 Not available
R3, R6 – OH (shikonin)

2-hydroxy-3-(3-methyl-but-2-enyl)-1,4-
R1 = -OH
naphthoquinone 242.27 141 – 143
R2 = -CH2CH=C(CH3) 2
(lapachol)

OH O O OH O OH
OH
CH3O CH3 CH3O CH3 CH3O
CH3
N OH O
CH3
OH O O OH O OH
bostrycoidin solaniol fusarubin
OH O
OH O CH2OH
CH3O CH3 O
OH
O HO
O
OH O
O OH
novarubin biflaviolin

Fig. (1). Selected fungal naphthoquinone metabolites.

NAPTHOQUINONES IN NATURE ticillium [14, 19-22]. Naphthoquinone metabolites of fungi


differ significantly in their chemical composition (Fig. 1).
Actinomycetes and Fungi
The simplest naphthoquinone produces by a fungus is
The ability of naphthoquinone synthesis is widespread juglone (Verticillium dahliae).
among fungal organisms. The complete list of fungi produc- Fungal naphthoquinones may be produced in in vitro
ers includes about 63 species [14]. They are widespread in conditions. Composition of produced naphthoquinones can
genus Fusarium – 10 species. They can be also found in be affected by initial ratio of carbon and nitrogen sources
other important genera such as Aspergillus, Cladosporium, and pH of cultivation medium [23, 24]. Fungal naphthoqui-
Microsporium, Mollisia, Penicillium, Trichophyton and Ver-
Noteworthy Secondary Metabolites Naphthoquinones Current Pharmaceutical Analysis, 2009, Vol. 5, No. 1 49

O O OH O

5 malonyl-CoA S-CoA S-CoA


COOH COOH
O HO
O O O
-CO2

OH OH
OH O

HO OH
HO OH
O naphthalene-1,3,6,8-tetraol
flavioline
Fig. (2). Mevalonate pathway of flaviolin biosynthesis.

H3C CH3
OH O OH O O OH
H3C CH3

HO O OH
OH O OH O OH OH O OH
naphthazarin hybocarpone

Fig. (3). Naphthazarin and hybocarpone.

nones biosynthesis was investigated using 14C and 3H la- simple redish-brown pigment flaviolin (Fig. 2) originates in
belled acetate or methyl group of methionin. This data indi- the presence of rppA gene via malonate pathway and repre-
cate that synthesis of naphthoquinones proceeds via the for- sents a precursor for other naphthoquinones [13]. Production
mation of a precursor. This precursor is product of the ace- of mostly prenylated naphthoquinones, such as naphterpin,
tate/malonate pathway [23, 25]. The fungal naphthoquinones naphthgeranines, fumaquinone, furaquinocins, marinone,
have a broad range of biological activities. They are active neomarinone hygrocins, chloroquinocins, napyradiomycin
against bacteria, yeasts and fungi. Cytotoxic properties of and other naphthoquinone compounds, such as heterocyclic
fungal naphthoquinones were demonstrated on mouse leu- naphthoquinones with significant anti-inflammatory efect is
kaemia and HeLa cells. Antibiotic activity is manifest espe- also reported [31-46].
cially against Gram-positive bacteria [26]. Antibacterial ac-
tivity of fungal naphthoquinones depends on their chemical Lichens
structure – the most important requirement is water solubility
Lichens were determined as producers of wide spectrum
because of penetration to cells trough cell membranes. The
of secondary metabolites that demonstrate many biological
toxicity of the naphthoquinones decreases in the presence of
actions, such as antioxidant, antimicrobial or cytotoxic. Ali-
metal ions (Cu, Fe, Al) [27]. Antibacterial effect is based on phatic acids, pulvinic acid, depsides, depsidones, dibenzo-
inhibition of mitochondrial respiration (respiratory chain
furans, anthraquinones and also naphthoquinones were de-
interruption), suppress of RNA biosynthesis, and biosynthe-
scribed as compounds responsible for these activities [47-
sis of proteins, lipids and transport of glucose. Fungal naph-
51]. The best-known genera that produce them are Cetraria
thoquinones are capable of redox conversions, whereas their
and Cladonia [52, 53]. Naphthazarin and its derivates were
antibiotic, cytotoxic and phytotoxic effect are due to interac-
isolated from Cetraria islandica (Fig. 3). This naphthoqui-
tions with the oxidative systems of cells [28]. They are able none demonstrate in in vitro experiments strong cytotoxic
to accept the reducing equivalents from the redox enzymes
effect to human epidermal carcinoma cells. Dimer of this
and transfer them to oxygen. Two basic systems providing
naphthoquinone, hybocarpone, was isolated from Lecanora
resistance of fungi to their own secondary metabolites –
hybocarpa [54].
naphthoquinones – is also known. The first one bases in
change of the sensitivity of the target or its absence in the Bignononiaceae (incl. Avicenniaceae)
cells of producers and second one in change the naphthoqui-
none toxicity by metabolic modification, such as phosphory- Family Bignoniaceae includes about 120 genera and 650
lation or acetylation [29]. Many naphthoquinones exhibit the species of tropical trees, shrubs or lianes, mainly abundant in
properties of mycotoxins (in Penicillium, Aspergillus, Mi- Brazil. The presence of naphthoquinone production was re-
crosporium and Trichophyton) [30]. corded in genera Catalpa [55], Heterophragma [56, 57],
Kigelia [58-61], Macfadyena [62], Mansona [2], Markhamia
Actinomycetes (genus Streptomyces) are the producers of [63, 64], Newbouldia [65-70], Oroxylum [71, 72], Rader-
substitute isoprene-derived naphthoquinones. Structurally
machera [73], Stereospermum [74], Tabebuia [3, 75, 76] and
50 Current Pharmaceutical Analysis, 2009, Vol. 5, No. 1 Babula et al.

O O O

O O OH
O OH O O

-lapachone 9-hydroxy--lapachone lapachol

Fig. (4). Some naphthoquinones of Bignoniaceae family.

COOH O
COOH COOH COOH COOH
+
HOOC
HO OH
OH O O O

shikimic acid -ketoglutaric acid o-sukcinylbenzoic acid droserone, juglone

Fig. (5). Some naphthoquinones of family Droseraceae.

O O O OH O
CH3 H3C CH3 CH3

OH OH
OH O OH O OH O OH O

plumbagin 7-methyljuglone droserone hydroxydroserone

O O O
O H3C CH3
OH
CH3
O
O OH OH O O OH
diomuscione muscipulone biramentaceone

Fig. (6). Droserone and juglone biosynthesis via shikimate pathway.

Zeyheria [77]. Majority naphthoquinone in this family is mechanism in four genera (Aldrovanda, Dionaea, Drosera
lapachol and its derivates (Fig. 4). Kigelia pinata bark is and Drosophyllum). The majority naphthoquinones detected
used in Nigerian medicine for antimicrobial, tripanocidal and in almost all species of this family are plumbagin and 7-
anticarcinogenic (mainly melanoma) activities [78-81]. Bark methyljuglone. Plumbagin is majority naphthoquinone of
and wood of Tabebuia species, namely T. avellanedae, T. Aldrovanda, Dionaea [93, 94], Drosophyllum [95] and some
pentaphylla and T. ochracea, are widely used in traditional Drosera species – D. anglica, D. auriculata, D. binata, D.
medicines of South America countries against cancer and for capensis, D. cistiflora, D. indica, D. intermedia, D. longifo-
anti-inflammatory properties [82-89]. The root bark of lia, D. peltata, D. ramentacea, D. rotundifolia and D. whi-
Stereospermum kunthianum is used by some tribes in takeri [96, 97]. The other Drosera species contain alternative
Uganda to treat fever. Naphthoquinones – sterekunthals and monomeric naphthoquinones, such as droserone, 7-methyl-
pyranokunthones – with potent antiplasmodial effect were juglone and their glycosides and dimeric naphthoquinones
found in liphophilic extract of root bark [90]. Derivates of (Fig. 5) [98]. They are also produced in in vitro cultures [99-
lapachol with cancer chemoprotective activity were recorded 106]. Two compounds biosynthetically connected with
in different parts of Avicennia alba and A. rumphiana [1, 91, plumbagine, diomuscione and muscipulone, were identified
92]. in Dionaea muscipula [107]. Droserone or juglone are naph-
thoquinones biosynthetised via shikimate pathway. Molecule
Droseraceae of shikimic acid condenses with molecule of -ketoglutaric
acid and forms naphthoquinones of droserone or juglone
Family Droseraceae include about 105 species of car-
type (Fig. 6). The second biosynthetic pathway includes syn-
nivorous perennial or annual herbs with active trapping
thesis of shikimic acid trough aromatic amino-acid L-
Noteworthy Secondary Metabolites Naphthoquinones Current Pharmaceutical Analysis, 2009, Vol. 5, No. 1 51

COOH COOH COOH


shikimate kinase
COOH
chorismate synthase
+
P O CH2 CH2
HO OH HO OP O
OH OH OH COOH

shikimate shikimate 3-phosphate phosphoenolpyruvate chorismate


chorismate mutase

COOH HOOC COOH


COOH prephenate aminotransferase

O
NH2
HO

glutamate OH
arogenate a-ketoglutarate
prephenate
4-hydroxyphenylpyruvate
arogenat dehydratase
NADPH+H
arogenat-
dehydrogenase
phenylalanine
NADP H2O, CO2
Phenylalanine and Tyrosine,
pathway in yeast and E. coli
alkaloids

COOH
transamination COOH
NH2
HO O
L-tyrosine
O2 phenylpyruvate

CO2
OH OH OH O
H3C CH3
CH3 CH3 H3C CH3
COOH + C5

OH OH OH O
1,4-naphthoquinone
homogentisate toluquinone (droserone, 7-methyljuglone)

Fig. (7). 7-methyljuglone and plumbagin synthesis through L-tyrosine.

tyrosine. Plumbagin and 7-methyljuglone are two important sidase to juglone. Naphthoquinones have been detected in
products of this pathway (Fig. 7). The European sundew, Juglans nigra [112] and J. regia [9, 113]. Extracts, as well as
Drosera rotundifolia, has been utilized in folk medicines for miscellaneous parts of walnuts, have long traditions of use in
long time and has been included in some pharmacopoeias. In folk medicines for acne, allergies, gastrointestinal disorders
the Czech Republic it has been using from 13th century and intestinal parasitosis treatment. External use is reserved
against cold, cough, bronchitis and asthma. Anti- for fungal, bacterial and viral infections (herpes) therapy.
inflammatory effect was found out too [108]. Nature medi- Most recently published analyses demonstrate the presence
cine in USA recommends extract from D. rotundifolia as of juglone in other members of Juglandaceae family – genus
geriatric. In Italy, it is an ingredient of a liqueur. Drosera Pterocarya (P.fraxinifolia) [114].
burmanni is widely used in Chinese traditional medicine as
well as rubefacient in Indic folk medicine [109]. The crushed Plumbaginaceae
leaves of Drosera indica and D. peltata are used as rubefa-
Cosmopolitan-spread family Plumbaginaceae includes
cient and vesicant [109]. about 775 species of herbs, shrubs or lianas in 24 genera.
Naphthoquinones, above all plumbagin and its derivates, are
Juglandaceae
reported in genera Ceratostigma, Limonium, Plumbago and
North temperate and subtropical family Juglandaceae ex- Plumbagella. Plumbago zeylanica is one of the most exam-
tending to India, Indochina, Malaysia and Andean South ined plants. Naphthoquinones, bisnaphthoquinones and tris-
America contains about 50 species in 8 genera. The most naphthoquinones (plumbagin, chitranone, maritinone, ellipti-
important genus is Juglans (valuable source of edible nuts) none and isoshinanolon) [115-118] and plumbagic acid with
with majority naphthoquinone juglone. Juglone is present in two glucosides [116] have been isolated from this semi-
free as well as glucosidic form (1,5-dihydroxy-4-naftalenyl- climbing subshrub (Fig. 8).
-D-glucopyranoside) [9, 110, 111]. This glucosidic form is
The whole plant and the roots are used in folk medicine
decomposing by hydrojuglone--D-glucopyranosid--gluco-
in Thailand for the treatment of carbuncle, ulcers, injuries,
52 Current Pharmaceutical Analysis, 2009, Vol. 5, No. 1 Babula et al.

OH O
O O
CH3 H3C
OH O OH O
CH3
O
OH O O OH
O
CH3 CH3
CH3
O O

OH O
chitranone elliptinone
maritinone
Fig. (8). Naphthoquinones of Plumbago zeylanica.
OH O OH O O O OH
CH3

OH O
H3C H3C
O O OH O O
H3C
H3C

O O diospyrin O O isozeylanone
CH3
HO OH OH O O OH
O OH

8´-hydroxyisodiospyrin
OH O
O H3C CH3
CH3 H3C
OMe OMe O O
OH O
diosindigo A CH3 3,3´- biplumbagin
O
OH O
CH3
OH O
O
chitranone 2,3-epoxyplumbagin

Fig. (9). Napthoquinones of different Diospyros species: 8´-hydroxyisodiospyrin, diospyrin (D. assimilis, D. montana, D. paniculata), dios-
indigo A (D. sylvatica), isozeylanone, 3,3´-biplumbagin, chitranone and 2,3-epoxyplumbagin (D. maritima).

elimination internal parasites and rheumatic pains [119]. Fruits of Diospyros maritima contain very rare brominate
Moreover in traditional medicine of India have a use for di- derivate of plumbagin – 3-bromoplumbagin [153]. Other
arrhoea, digestion disorders and skin diseases treatment as compounds biosynthetically connected with 1,4-naphtho-
well as abortion induction [120] and in Nigerian (Africa) for quinones are naphthalene derivates and naphthaldehydes
parasitic diseases, scabies and ulcers [121]. Other Plumbago (Fig. 9). These naphthoquinones exhibit very interesting
species (P. auriculata, P. europaea, P. rosea, and P. scan- pharmacological properties – extracts of many species this
dens) as well as their in vitro cultures (callus, suspension, genus are widely used in African, Chinese and Indian folk-
immobilised cells and hairy root cultures) produce plum- lore medicine for whooping cough, leprosy, scabies, skin
bagin and other naphthoquinones too [11, 12, 122-133]. diseases eye infections, menstrual troubles and parasitosis
Plumbagin, its glycosides and derivates, shinanolon and epi- [154]. 7-methyljuglon and other naphthoquinone derivates
isoshinanolon, have been detected in extracts of Cerato- were determined in parts of Euclea divinorum [155] and
stigma minus and Ceratostigma willmottianum in addition to Euclea natalensis subsp. natalensis [156]. Body parts of dif-
phenolic compounds [134-136]. ferent Euclea species are used for various purposes, such as
toothbrushes, hypnotic, for toothache and headache by peo-
Ebenaceae ple of South Africa. All naphthoquinones of genus Euclea
have potent antituberculosis activity, especially 7-methyl-
Family Ebenaceae, containing about 500 species in two
juglone [156].
genera, is mainly extended in subtropical and tropical areas,
especially Indomalayan. Some species are important sources
Lythraceae
of timber (ebony) and fruits. The majority naphthoquinone of
genus Diospyros is plumbagin [7, 8, 137-150]. Some species Lawsonia inermis, a shrub cultivated in North Africa,
contains glycosides of plumbagin – diospyrosids A – D Egypt, India and Ceylon, has been using from ancient world
[151]. Dimers (D. greeniwai) were determined too [152]. to obtain henna. Henna (powder of dried leaves) consist of
Noteworthy Secondary Metabolites Naphthoquinones Current Pharmaceutical Analysis, 2009, Vol. 5, No. 1 53

OH OH OH O OH

C10
+

COOH COOH OH O

p-hydroxybenzoic acid alkannin

Fig. (10). Combined shikimate/mevalonate pathway of alkannin biosynthesis.

colouring matter – lawsone – and various phenolic com-


CH3
pounds, coumarins, xanthones, quinoids, flavonoids, fats,
OH O alkannan:
resin and henna-tanin [157, 158]. In vitro cultivated plants as
R CH3
well as hairy root cultures produce lawsone too [159]. H OH
CH3
Extracts from stem-bark are traditionally used in India for alkannin:
the treatment of liver and spleen diseases, jaundice and skin H OH CH3
diseases. Isoplumbagin was determined as anti-inflammatory OH O CH3
agent comparable to phenylbutazone [157]. Hepatoprotective shikonin
activity of stem-bark compounds was determined too [160]. CH3
Application of henna can induce severe haemolytic anaemia;
lawsone was determined as causative agent [161]. Fig. (11). Main napthoquinone derivates of Alkanna tinctoria roots.

Boraginaceae alkannan and shikonin (Fig. 11) [179, 180]. Most of these
This family includes annual to perennial herbs, shrubs, pigments are produced in plant cell suspension cultures [162,
small trees or lianas with 2000 species in 120 genera. The 181]. Other members of family Boraginaceae, such as Mac-
most important plants containing naphthoquinones belongs rotomia cephalotes, produce similar red pigment.
to genera Alkanna [162], Cordia [163, 164], Lithospermum Derivates of shikonin were discovered in roots of Turkish
[165], Arnebia and Onosma [166, 167]. The most important species Onosma argentatum, as well as Onosma armeniacum
naphthoquinones detected in species of these genera are [182]. Antifungal properties of these derivates were con-
shikonin, acetylshikonin, deoxyshikonin, isobutylshikonin firmed. In addition to that larvicidal meroterpenoid naphtho-
and isovalerylshikonin. The compounds of these family quinones were isolated from roots of Cordia linnae [183];
members are biosynthesized via combined shiki- roots of other species – Cordia corymbosa and C. curas-
mate/mevalonate pathway (Fig. 10). The reported naphtho- savica – contain sesquiterpenic (meroterpenoid) napthoqui-
quinone constituent of genus Lithospermum is shikonin. nones – cordiaquinones – with activity against some fungal
Roots of L. erythrorhizon are widely used for shikonin isola- pathogens like Cladosporium or Candida [163, 184]. Roots
tion and are produced for cosmetic and pharmaceutical in- of Arnebia densiflora, widespread plant in Anatolia, contain
dustries in Japan [4, 165]. In vitro plant cell suspension cul- alkannin derivates [185]. The latest report is the utilization of
tures stimulated by specific oligogalacturonides, ethylene roots of Rindera graeca (in vivo and in vitro as transgenic
[168, 169], ultrasound [170] and low-dose gamma irradiation roots) as source of shikonin and its derivates [175].
[171]. Transformed organ cultures are used for shikonin pro-
duction too [172-175]. Other species widely utilised in Chi- There is one important, but unanswered question, do
nese folk medicine are Arnebia guttata and A. euchroma – similar plant families closely related to Boraginaceae (Ru-
these two species are listed in Chinese pharmacopoeia as biaceae with important genera Galium and Rubia) contain
official sourcing of drug “zicao” [167, 176]. In the Far East, naphthoquinones or similar compounds [186, 187]?
preparations from the purple roots have long been used for
treatment of inflammations, burns, wounds and ulcers, but in Nepenthaceae
in vitro laboratory tests skinonin (alkannine too) have no Family Nepenthaceae with 70 species at 2 genera in-
significant anti-inflammatory activity. Other species, such as cludes carnivorous plants with specialized leaves as passive
Lithospermum arvense, are used as oral contraceptive in traps - pitchers. Naphthoquinones are characteristic com-
Central Europe and suppress the oestrus cycle. Similar North pounds of these plants. Plumbagin, isoshinanolone, epishi-
American species, Lithospermum ruderale, has similar hor- nanolone, shinanolone, quercetin and kaempferol were iso-
monal activity and Lithospermum canescens contains lated from the leaves of Nepenthes gracilis [188], from roots
shikonin derivates [177, 178]. of N. rafflesiana droserone, hydroxydroserone, plumbagin
Dried roots of Alkanna tinctoria, herb cultivated in and the nepenthones A-C [189, 190]. Roots of N. thorelli
Southern Europe, Hungary and Turkey, are used for colour- contain plumbagin, droserone, 2-methylnaphthazarin and
ing oil and tars. The tincture of these pigments is used for isoshinanolon [10]. By means of application of labelled L-
microscopical detection of oils and fats. These coloured alanine to the pitchers the incorporation of L-alanine to
compounds are naphthoquinone derivates too – alkannin, plumbagin was explained (Fig. 12) [191]. The closely related
54 Current Pharmaceutical Analysis, 2009, Vol. 5, No. 1 Babula et al.

CoASH+NAD+
-ketoglutarate
glutamate
CO2+NADH
H3C COOH H3C COOH H3C SCoA

alanine-aminotransferase pyruvate-dehydrogenase
NH2 O O

L-alanine pyruvate acetyl CoA

6x

OH O OH OH O O O

oxidation reduction,aldol condensation


SCoA
decarboxylation
CH3 CH3 O CH3
O O
O
plumbagin

Fig. (12). Incorporation of L-alanine to plumbagin structure.

CH3
O O O
R

R R OH
O O O

impatienol - R = -OH balsaminol - R = -OH


impatienolate - R = -ONa balsaminolate - R = -ONa

Fig. (13). Naphthoquinones isolated from aerial parts of Impatiens balsamina.

families are Ancistrocladaceae and Dioncophyllaceae, both Ancistrocladus with derivates of plumbagin [202], Annona-
with naphthoquinone contents. ceae (roots of Goniothalamus cheliensis) [203], Dioncophyl-
laceae with only genus Triphyophyllum peltatum (droseron,
Balsaminaceae plumbagin) [204], Eriocaulaceae (Paepalanthus –
semixanthomegnin and its methoxy derivates) [205, 206],
This family contains approximately 600 species in only 4
Euphorbiaceae – Pera benesis (plumbagin and its derivates)
genera. Some species of genus Impatiens, such as I. balsa-
used as leihsmanicidal agent in Bolivia [207], Fabaceae
mina, one of the most known species, are widely used in
Chinese medicine for rheumatism, bruises, pain and swelling (naphthoquinone juglone was isolated from heartwood of
tree Caesalpinia sappan) [208], Gencianaceae (antifungal
treatment and in Japan topically as potent agent against sev-
properties of Swertia calycina extracts – 2-methoxy-1,4-
eral types of dermatitis including urticaria. The main active
naphthoquinone) [209], Iridaceae (Aristea, Eleutherine bul-
compounds were determined derivates of 1,4-naphthoqui-
bosa and E. americana – eleutherinon, eleutherin,
none, e. g. lawsone, 2-methoxy-1,4-naphthoquinone and
isoeleutherin and their derivates) [210-212], Liliaceae (naph-
other naphthoquinones, such as impatienol and balsaminol
and their derivates; plumbagin was determined later (Fig. 13) thol-naphthoquinone derivates such as imbricatonol isolated
from Stypandra imbricata and Dianella revoluta) [213, 214],
[94, 192-194], and flavonoid compounds in addition.
Malvaceae (genus Hibiscus with very rare derivates of 1,2-
Other Plant Families naphthoquinones called hibiscones A-D) [215], Leguminosae
– Caesalpinioideae (heartwood of Caesalpinia sappan with
Other plant families containing naphthoquinones are juglone content) [208], Pedaliaceae (Sesamum indicum, in
Acanthaceae with the genus Rhinacathus nasutus containing vitro production by roots transformed by Agrobacterium
esteric naphthoquinones marked as rhinacathins (A-D) in rhizogenes) [216], Proteaceae (Conospermum incurvum, C.
roots [195-198] using in Thai traditional medicine for cancer sphacelatum and C. stocheadis) [217-220], Pyrolaceae (5,8-
treatment [199-201], Ancistrocladaceae with only genus dihydro-2,7-dimethyl-1,4-naphthoquinone from the roots of
Noteworthy Secondary Metabolites Naphthoquinones Current Pharmaceutical Analysis, 2009, Vol. 5, No. 1 55

COOH pyruvate + CO2 OH


2-oxoglutarate COOH COOH
OH - H2O
COOH
COOH COOH
O CH2
O O
izochorismate 6-hydroxy-2-sukcinyl- 2-sukcinylbenzoate
cyklohexa-2,4-dienkarboxylate
ATP + HS-CoA

HS-CoA
O OH AMP + PPi
H COOH COOH

CO-S-CoA
CH3
O OH O

n = 2 - phylloquinone (vitamin K1) 1,4-dihydroxy-2-naphtoate 4-(2-karboxyphenyl)-4-


n = 1 - 13 - menaquinone (vitamin K2) oxobutanoyl-CoA

Fig. (14). Biosynthesis of phylloquinone and menaquinone via isochorismate.

NAD(P)H NAD(P)+
OH O
H Any
H N
N
H
3. CH3
2-RSH RS-SR
OH CO2H
O
vitamin K - quinol protrombin
1.
H precursor
O2, CO2

CH3
O
vitamin K

O O
H Any
2. H N
N
O H
CO2H
CH3
2-RSH RS-SR
O CO2H
vitamin K - 2,3-epoxide protrombin
Fig. (15). The role of vitamin K in coagulation.

Pyrola japonica) [221], Scrophulariaceae (Calceolaria at the C3 position by side chain (Fig. 14). They are synthe-
andina – 2-(1,1-dimethylprop-2-enyl)3-hydroxy-1,4-naphtho- sized by plants (phylloquinone, vitamin K1 with phytyl side
quinone and the corresponding acetate, 2-acetoxy-3-(1,1- chain with 20 carbons) and by bacteria (menaquinones with
dimethylprop-2-enyl)-1,4-naphthoquinone [222, 223]; Ca- repeating unsaturated prenyl units (MK-n, where n is number
praria biflora – biflorin [224], Paulownia tomentosa with of prenyl units) and compounds where one or more double
the low content of plumbagin [94]), Verbenaceae (Lippia bonds of side chain is saturated) [227]. Basic menadion (2-
microphylla – prenylated naphthoquinone dimer, mixture of methyl-1,4-naphthoquinone) is thought to be a provitamin
6-methoxy- and 7-methoxy-naphtho[2,3-b]-furan-4,9- and can be converted by vertebrates to MK-4. Vitamin K is
quinones and tecomaquinone from the roots) [225] and Zy- important cofactor for a specific carboxylation reaction that
gophyllaceae (larreantin from the root of Larrea tridentata) selectively transforms glutamate residues to -carboxygluta-
[226]. mate residues. Four procoagulants depend on vitamin K –
(prothrombin – factor II and factors VII, IX and X). All of
VITAMINS OF K GROUP – A STRUCTURAL ANA- them are serine proteases synthesized in the liver (Fig. 15).
LOGUES OF 1, 4-NAPHTHOQUINONE Vitamin K is metabolized in the liver and excreted in the
urine and bill as a water soluble conjugates, mainly with
Vitamin K is a title for group of fat-soluble compounds
glucuronic acid [228].
that have 2-methyl-1,4-naphthoquinone skeleton substituted
56 Current Pharmaceutical Analysis, 2009, Vol. 5, No. 1 Babula et al.

CTP
1.
karbamoyl-phosphate dihydroorotate orotate

dTTP

2.
O

intercalation
mitochondrion ROS DNA

O
3.

cytochrome-c
anti-apoptic genes BAX p53
Bcl-2, Bcl-xL

APAF-1 replication

APOPTOSIS caspase-3 caspase-9 IAP1,2

Fig. (16). Different mechanisms of 1,4-naphthoquinone derivates cytotoxic effect – inhibition of orotate synthesis (1), inhibition of
thymidine incorporation to DNA (2), inhibition of DNA replication by topoisomerase I and/or topoisomerase II cleavage (3), reactive oxygen
species (ROS) generation and mitochondrion damage with down-regulation of antiapoptic genes and releasing of apoptosis inducing factor.

Naphthoquinones and Interactions Between Plants mitochondrial cytochrome c and apoptosis inducing factor
(AIF). Activation of caspase-dependent (especially caspase-
Naphthoquinones, especially juglone, are widely studied
3) and caspase–independent pathways by plumbagin, but
for allelopathic activity. Massey in 1925 hypothesized that
also shikonin has been also demonstrated [241, 243, 244].
inhibitory effect of black walnut (Juglans nigra L.) on
Plumbagin and shikonin inhibits activation of NF-kappa B
growth of some associated species caused compound exuded
pathway by tumour necrosis factor (TNF), carcinogens and
by roots. This compound was determined in 1928 by Davis other inflammatory stimuli (hydrogen peroxide, cigarette
as juglone. Juglone, as well as other naphthoquinones, is able
smoke condensate etc.) [245, 246]. It was confirmed that
to release to environment through disintegration of necrotic
inhibition of tumour necrosis factor alpha by shikonin is
plant tissues, may persists in the soil and can be attributed to
based on selective blocade of Pre-mRNA splicing [247].
phytotoxicity. Growth inhibition and interaction by naphtho-
Plumbagin down-regulates the expression of NF-kappa B-
quinones has been reported for numerous plant species [229-
regulated anti-apoptotic (IAP1, IAP2, Bcl-2, Bcl-xL, cFLIP,
231]. Mechanisms of plant growth reduction by juglone ba- Bfl-1/A1, and survivin), proliferative (cyclin D1 and COX-
sis in decrease of H +-ATP-ase activity in corn and soybean
2), and angiogenic (matrix metalloproteinase-9 and vascular
root microsomal fraction [232], inhibition of p-hydroxy-
endothelial growth factor) gene products [248]. This may
phenylpyruvate dioxygenase (the crucial enzyme of plas-
explain its cell growth modulatory, anticarcinogenic, and
toquinone synthesis), inhibition of electron transport in mito-
radiosensitizing effects previously described in Ref. No.
chondria and chloroplasts [233], decrement photosynthesis in
[248, 249]. A following essays indicate that plumbagin
leaf tissues and transpiration and stomatal conductance inhi- blocks of cell cycle in association with increased levels of
bition [234] and reduction of water reuptake by the roots
p21 protein and reduced amounts of cyclinB1, Cdc2, and
[232]. Induction of reactive oxygen species (ROS) may play
Cdc25C. Treatment of tumour cells increases the activation
a critical role in the protection of plants against pathogens
of apoptosis signal-regulating kinase 1 and extracellular sig-
[235]. Plumbagin tested in in vitro plant cell models induces
nal regulate kinase 1/2 and c-Jun N-terminal kinase and also
polyploidy, micronuclei formation and chromosomal disor-
enhances the levels of inactivated phosphorylated Cdc2 and
ders (root tips of Allium cepa) [236]. Cdc25C [250, 251]. Synergic antitumour effect of plumbagin
may be also connected with inhibion of efflux of cytostatic
Pharmacological Properties of Napthoquinones
drug mitoxanthrone by multidrug resistance-linked ATP
Naphthoquinones, such as plumbagin, lapachol or bis- binding cassette drug transporter (ABCG2) [252]. Cytotoxic
naphthoquinoid diospyrin, have an interesting potential as a properties of naphthoquinones can be enhanced by inhibition
cytostatic agents. Naphthoquinone-induced cell death has of thymidin incorporation to DNA of cancer cells [253], in-
been determined in many cell including cancer cell lines - hibition of dihydroorotate dehydrogenase [254, 255], oxida-
HEPA-3B, COLO-25, A-459, ME-180, HaCaT and HeLa tive DNA damage [256], intercalation to DNA and mammal-
[237-242]. The cytotoxic effect of these compounds is based ian topoisomerase I or II DNA-cleavage (Fig. 16) [257-261].
on generation of reactive oxygen species and apoptosis in- Juglone in some work that has been recently published dem-
duction. Plumbagin-induced apoptosis involved release of onstrated ability to interfere with processes of mitosis. They
Noteworthy Secondary Metabolites Naphthoquinones Current Pharmaceutical Analysis, 2009, Vol. 5, No. 1 57

can inactivate cysteine-rich proteins (parvulins) that are es- The highest yield of naphthoquinones was obtained using
sential in mitosis progression [262]. Inhibition role in mitosis methanol and the lowest using n-hexane. Chloroform was
has also plumbagin; this naphthoquinone disrupts microtu- employed as suitable solvent for naphthoquinones extrac-
bule network, in more details inhibits tubuline polymeriza- tions too [7, 104, 114, 321]. The yield of naphthoquinones
tion [263]. Various novel synthetic naphthoquinone derivates depends on temperature – for example yield of juglone sig-
with interesting anticarcinogenic properties have been pre- nificantly decrease with increasing temperature [7, 104,
pared [264-280]. 321]. Very important problem may be the ability of some
naphthoquinones, such as alkannin or shikonin, to subject to
Antimicrobial activities of naturally occurring naphtho-
some transformations during extraction processes, such as
quinones have been investigated. Plumbagin, juglone and
photochemical or thermal degradation and especially polym-
lawsone demonstrated this effect on Streptococcus,
Prevotella, Peptostreptococcus, Mycobacterium, Clostrid- erization; these processes can importantly decrease the re-
covery of these naphthoquinones [322].
ium, Helicobacter and Escherichia [143, 281, 282], lapachol
on Kleibsiella, Proteus, Helicobacter and Streptococcus The solvent is often evaporated to obtain dry residue con-
[283, 284]. Other naphthoquinones have also antibiotic activ- taining naphthoquinones, which is consequently dissolved in
ity [285]. Shikonin and its derivates is potent antifungal other solvent and analysed [114]. In addition, very simple
compound against Candida and Saccharomyces [286]. Syn- methods of plumbagin isolation were developed – cold mac-
thetic derivates can be potentially antibacterial and antifun- eration of dry material by acetone in connection with precipi-
gal compounds [287-293]. Lapachol and furanonaphthoqui- tation by water, redissolution in chloroform and separation
nones has inhibitory effect on Epstein-Barr virus activation using column chromatography. In addition, plubagin ability
[1, 294, 295], some intricately substituted naphthoquinones to sublime at 90°C can be used in its isolation from plant
have inhibitory activity against HIV [218, 296], inhibit HIV- material [323].
1 integrase [297] and RNAse-H activity associated with
HIV-1 reverse transcriptase [298]. Analytical Techniques for Detection of Napthoquinones
Monomer and dimeric, natural and synthetic naphthoqui- High performance liquid chromatography with UV/VIS
nones showed in in vitro and in in vivo tests activity against detection (HPLC-UV/VIS), especially normal-phase HPLC-
Leishmania [299-304], Pneumocystis [3], Trypanosoma UV/VIS or eventually liquid chromatography-tandem mass
[299, 301, 305], Trichomonas [301] and Plasmodium [10]. spectroscopy is one of the most commonly used techniques
Other described pharmacological activities of naphthoqui- for naphthoquinone determination and characterization in
nones are anti-inflammatory [165, 306], cardiotonic and biological samples [324, 325]. Optimization of detecting
ionotropic [307, 308], antikoagulation [309, 310], hypolipi- conditions of naphthoquinone derivates depends on kind of
demic (the mechanisms is probably based on inhibition of compound. The most used analytical columns are C18 col-
cholesterol acyltransferase) [311, 312], spasmolytic [313], umns, but some authors favour C8 column because of resolu-
antifertility and antigonatropic [120, 314-317]. tion advance [324, 326]. Some naphthoquinones, particularly
isohexenyl-naphthoquinone derivatives (alkannin, acetyl-
NAPHTHOQUINONES ANALYSIS alkannin, deoxyalkannin etc.), can not be separated by nor-
mal-phase or reverse-phase because of the fact that the pair
Extraction of Naphthoquinones
of isomers coeluted as a mixture or as single component, but
Number of authors has extracted naphthoquinones by only by ion-pair method with tetramethylammonium chlo-
various organic solvents using different extraction condi- ride and nonylamine as the ion-pair reagent. These naphtho-
tions, such as temperature. The most frequent used solvent quinones could not be eluted from the column by the normal-
for naphthoquinones extraction is methanol [167, 197]. Some phase and reverse-phase (various compositions of methanol-
works indicate that methanol is the most suitable solvent to water and acetonitrile-water) [327]. Optical isomers shikonin
obtain the highest extraction yields, but hexane provides high and alkannin can be separated by chiral phase high perform-
recovery with highest degree of naphthoquinones purity ance liquid chromatography, high performance liquid chro-
[318]. Other, less frequently used solvents are dichlor- matography in connection with atmospheric pressure chemi-
methane [183], chloroform [114, 156], petrol [137] or buta- cal ionization quadrupole mass spectrometry [328], high
nol [134], eventually ethanol [319]. performance liquid chromatography in connection with pho-
todiode array/mass spectrometry method [329] or high-speed
Conventional methods, as maceration or hot extraction, counter-current chromatography [182, 328, 330]; these tech-
are widely used to obtain plant extracts. Some articles com- niques may be used also in purification of alkannin and
pared different available techniques. Extraction in Soxhlet shikonin [328]. They have usually UV absorption at 214, 275
apparatus for 5 hours demonstrates the highest efficiency to and 520 nm [185]. There was also described technique based
plumbagin isolation from dried roots of Plumbago scandens on capillary electrophoresis with high freequency conductiv-
in the confrontation with static maceration or dynamic mac- ity detection for shikonin determination in biological sam-
eration [123]. Naphthoquinones juglone, plumbagin and syn- ples [176]. HPLC is the effective method for detection of
thetic 1,4-naphthoquinone demonstrate good solubility in lapachol, -lapachone and other isomers too [331], but utiliz-
supercritical CO2 [320]. Sonification can be an extraordinary ing the chiral columns for quality resolution is necessary
technique for naphthoquinone extraction from plant material [332]; some advantages brings using of counter-current
because some naphthoquinones are heat-labile. The sonifica- chromatography in lapachol and its derivates determination
tion also allows use of low volumes of the extraction sol- [333]. Normal-phase liquid chromatography is a simple and
vents. rapid method for the determination for pharmacological im-
58 Current Pharmaceutical Analysis, 2009, Vol. 5, No. 1 Babula et al.

Fig. (17). HPLC-UV/VIS and electrochemical chromatograms standards (left) and Nepenthes real samples (right). HPLC conditions were as
follows: mobile phase: 0.1 mol.l-1 acetic acid:methanol in ratio of 35:65 (%,v/v) except; flow rate: 0.75 ml.min -1; temperature: 42°C. Chroma-
tograms were registered at 260 nm and at applied potential 900 mV.

portant plumbagin and other naphthoquinones, such as aprotic solvents [348]. Cyclic voltammetry was used in one
juglone and lawsone. As the mobile phase mixtures n- study to determine the toxicity of different alkannin derivates
hexane, chloroform and 2-propanol [334, 335] or methanol by monitoring of their ability to induce reactive oxygen spe-
and acetic acid are used [94, 336]. Typical HPLC-UV/VIS cies [349]. Some naphthoquinones, such as plumbagin,
chromatogram of naphthoquinones is shown in Fig. (17). juglone or lawsone, may be analysed very sensitively by
The compounds of interest are well separated and analysed differential pulse voltammetry coupled with hanging mer-
within 30 minutes. Moreover, detection limits of the tech- cury drop and carbon paste electrodes and by injection
nique enable us to employ it for analysing of a real sample analysis coupled with carbon printed electrode. Electro-
(Table 2). This fact is well demonstrated on chromatogram chemical methods are suitable for the construction of the
of extract from Nepenthes plant, where we can easily identi- miniaturized sensors and biosensors [350-352]. Besides this
fied and quantified plumbagin. Rapid techniques of plum- electrochemical detector coupled with liquid chromatogra-
bagin determination in plant and soil samples by thin layer phy (HPLC-ED) is an attractive alternative method for elec-
chromatography were developed [337] or high performance troactive species detection, because of its inherent advan-
thin layer chromatography [338]. tages of simplicity, ease of miniaturization, high sensitivity
and relatively low cost. HPLC-ED has been many times suc-
The electrochemical techniques have been utilizing
mainly to study of naphthoquinones chemical interactions cessfully utilized for determination of plant secondary me-
tabolites as flavonoids [353, 354]. The electrochemical de-
(particularly naphthoquinone-amino acid or protein interac-
tector is also suitable for determination of naphthoquinones
tion) [339-345], but not for determination in biological sam-
not only as standards but also in real samples (Table 2 and
ples or plant material. Electrochemistry, such as cyclic volt-
Fig. 17). Therefore, we employed this technique to quantify
ammetry, is also good tool to study menadione (vitamin K3)
plumbagin in extracts from Nepenthes plants (Fig. 18). Con-
and its chemical interactions [346, 347]. Cyclic voltammetry,
chronoamperometry and chronopotentiometry at the surface tent of plumbagin varied in different plant tissues. The high-
est content was determined in apex and the lowest ones in
of Au, Pt and glassy carbon electrodes are good tools to
petiole. There have been also used for analysis of naphtho-
study of some naphthoquinones (such as plumbagin) in
quinones micellar electrokinetic chromatography [355],
Noteworthy Secondary Metabolites Naphthoquinones Current Pharmaceutical Analysis, 2009, Vol. 5, No. 1 59

Table 2. HPLC with UV/VIS and Electrochemical Detection (ED) Characteristics of Analysed Naphthoquinones (n = 5)

LOD LOQ R.S.D.


Naphthoquinone tR (min) a Equationb,c R2
(ng.ml-1) d (ng.ml-1) d (%)e

y = 0.5185x + 0.1097* 1.0000* 10* 33* 2.9*


Lawsone 3.05
y = 1.4701x + 2.8418** 0.9965** 1** 3** 3.9**

y = 0.7451x + 0.0882* 0.9999* 15* 50* 2.3*


1,4-naphthoquinone 3.78
y = 0.0334x - 22.669** 0.9901** 2** 7** 4.2**

y = 0.8126x - 0.5371* 0.9971* 10* 33* 2.0*


Juglone 4.27
y = 0.5235x + 3.3759** 0.9933** 1** 3** 4.1**

y = 0.5141x - 0.3141* 0.9996* 40* 133* 1.1*


Plumbagin 7.49
y = 0.4196x + 0.7479** 0.9974** 5** 17** 3.6**

2,2-(3-hydroxy)-1,4- y = 0.5141x - 0.3141* 0.9996* 90* 300* 4.6*


9.51
naphthoquinone y = 0.006950x + 0.3329** 0.9965** 15** 50** 6.9**
a
… Retention time.
b
… The concentration range was from 50 to 5,000 μg.ml-1.
c
… The equation was derived from the dependence of the peak area on the naphthoquinone concentration.
d
… The detection limits (LOD, 3 S/N) and quantification limits (LOQ, 10 S/N) were calculated according to Long [359], whereas N was expressed as
standard deviation of noise determined in the signal domain.
e
… Relative standard deviation (R.S.D.).
*
… HPLC-UV/VIS.
**
… HPLC-ED.

Fig. (18). Content of plumbagin in tissues of Nepenthes plants. The tissues were processed according to [336]. Particularly, the tissues were
extracting for 24 hours in methanol, then centrifuged and filtered prior to HPLC-ED analysis. Plumbagin content 140 M corresponds to
100 %.

spectrophotometric methods coupled with mathematical plumbagin [357]. There is one work utilizing plumbagin for
processing of data [356] and enzyme-linked immunosorbent glassy carbon electrode modification for determination of
assay using highly-specific monocloal antibodies against electrocatalytic oxygen reduction [358].
60 Current Pharmaceutical Analysis, 2009, Vol. 5, No. 1 Babula et al.

ACKNOWLEDGEMENTS bioactivities of mollisin and two new, structurally related fungal


naphthoquinone metabolites. Chem. Biodiver., 2005, 2, 1109-15.
Financial support from the grant IGA MZ 1A/8666-3, [23] Kurobane, I.; Vining, L.C.; McInnes, A.G.; Gerber, N.N. Metabo-
1M06030 and FRVS A No. 280371 are greatly acknowl- lites of Fusarium solani related to dihydrofusarubin. J. Antibiot.,
1980, 33, 1376-79.
edged. [24] Medentsev, A.G.; Akimenko, V.K. Effect of culture conditions on
biosynthesis of naphthoquinone metabolites by Fusarium decem-
REFERENCES cellulare fungi. Microbiology, 1992, 61, 572-76.
[25] Kurobane, I.; Vining, L.C.; McIlnnes, A.G.; Walter, J.A. Use of C-
[1] Itoigawa, M.; Ito, C.; Tan, H.T. W.; Okuda, M.; Tokuda, H.; Ni-
13 in biosynthetic studies-The labeling pattern in dihydrofusarubin
shino, N.; Furukawa, H. Cancer chemopreventive activity of naph- enriched from C-13- and C-13,H-2- acetate in cultures of Fusarium
thoquinones and theit analogs from Avicennia plants. Cancer Lett.,
Solani. Can. J. Chem., 1980, 58, 1380-85.
2001, 135-39. [26] Werner, R.G.; Appel, K.R.; Merk, W.M.A. Gunacin, a new qui-
[2] Itokawa, H.; Matsumoto, K.; Morita, H.; Takeya, K. Cytotoxic
none antibiotic from Ustilago sp. J. Antibiot., 1979, 32, 1104-11.
naphthoquinones from Mansona alliacea. Phytochemistry, 1992, [27] Kern, H. The naphthazarins of Fusarium. Annals Phytopathol.,
31, 1061-62.
1978, 10, 327-45.
[3] De Moura, K.C.G.; Emery, F.S.; Neves-Pinto, C.; DoCarmo, M.; [28] Medentsev, A.G.; Akimenko, V.K. Mechanism of phytotoxic ac-
Pinto, F.R.; Dantas, A.P.; Salomao, K.; DeCastro, S.L.; Pinto, A.V.
tion of naphthoquinone pigments of the fungus Fusarium decem-
Tripanocidal activity of isolated Naphthoquinones from Tabebuia cellulare. Phytochemistry, 1992, 31, 77-79.
and some heterocyclic derivates: A rewiew from an interdiscipli-
[29] Vining, L.C. Antibiotic tolerance in producer organisms. Adv. Appl.
nary Study. J. Braz. Chem. Soc., 2001, 12, 325-38. Micobiol., 1979, 25, 147-68.
[4] Brigham, L. A.; Michaels, P. J.; Flores, H. E. Cell-specific produc-
[30] Robbers, J.E.; Hong, S.; Tiuite, J.; Carlton, W.W. Production of
tion and antimicrobial activity of naphthoquinones in roots of xanthomegnin and viomellein by species of Aspergillus correlated
Lithospermum erythrorhizon. Plant Physiol., 1999, 119, 417-28.
with mycotoxicosis produced in mice. Appl. Environ. Microbiol.,
[5] Zenk, M. H.; Furbringer, M.; Steglich, W. Occurence and distribu- 1978, 36, 819-23.
tion of 7-methyljuglon and plumbagin in the Droseraceae. Phyto-
[31] Shin-Ya, K.; Imai, S.; Furihata, K.; Hayakawa, Y.; Kato, Y.; Van-
chemistry, 1969, 8, 2199-220. duyne, G.D.; Clardy, J.; Seto, H. Isolation and structural elucida-
[6] Culham, A.; Gornall, R. J. The taxonomic significance of naphtho-
tion of an antioxidative agent, naphterpin. J. Antibiot., 1990, 43,
quinones in the Droseraceae. Biochem. Syst. Ecol., 1994, 22, 507- 444-47.
15.
[32] Wessels, P.; Gohrt, A.; Zeeck, A.; Drautz, H.; Zahner, H. Meta-
[7] Zhong, S.M.; Waterman, P.G.; Jeffreys, J.A.D. Naphthoquinones bolic products of microorganisms, 260. Naphthgeranines, new
and triterpenes from african Diospyros species. Phytochemistry,
naphthoquinone antibiotics from Streptomyces ssp. J. Antibiot.,
1984, 23, 1067-72. 1991, 44, 1013-18.
[8] Zakaria, M.B.; Jeffreys, J.A.D.; Waterman, P.G.; Zhong, S.M.
[33] Charan, R.D.; Schlingmann, G.; Bernan, V.; Feng, X.; Carter, G.T.
Naphthoquinones and triterpenes from some asian Diospyros spe- Fumaquinone, a new prenylated naphthoquinone from Streptomy-
cies. Phytochemistry, 1984, 23, 1481-84.
ces fumanus. J. Antibiot., 2005, 58, 271-74.
[9] Binder, R.G.; Benson, M.E.; Flath, R.A. Eight 1,4- [34] Funayama, S.; Ishibashi, M.; Anraku, Y.; Komiyama, K.; Omura,
naphthoquinones from juglans. Phytochemistry, 1989, 28, 2799-
S. Structures of novel antibiotics, furaquinocins A and B. Tetrahe-
801. dron Lett., 1989, 30, 7427-30.
[10] Likhitwitayaawud, K.; Kaewamatawong, R.; Ruandgungsi, N.;
[35] Ishibashi, M.; Funayama, S.; Anraku, Y.; Komiyama, K.; Omura,
Krungkrai, J. Antimalar naphthoquinones from nepenthes thorelii. S. Novel antibiotics, furaquinocins C, D, E, F, G and H. J. Anti-
Planta Med., 1998, 64, 237-41.
biot., 1991, 44, 390-95.
[11] Dinda, B.; Das, S.K.; Hajra, A.K. Naphthoquinones from the roots [36] Pathirana, C.; Jensen, P.R.; Fenical, W. Marinone and debro-
of Plumbago rosea Linn. Indian J. Chem. Sect. B, 1995, 34, 525-28.
momarinone: Antibiotic sesquiterpenoid naphthoquinones of a new
[12] Dinda, B.; Das, S.K.; Hajra, A.K. Chemical constituents of Plum- structure class from a marine bacterium. Tetrahedron Lett., 1992,
bago indica roots. Indian J. Chem. Sect. B, 1998, 37, 672-75.
33, 7663-66.
[13] Moore, S.B.; Hopke, N.J. Discovery of new bacterial polyketide [37] Kalaitzis, J.A.; Hamano, Y.; Nilsen, G.; Moore, B.S. Biosynthesis
biosynthetic pathway. Chem. Biochem., 2001, 2, 35-38.
and structural revision of neomarinone. Org. Lett., 2003, 5, 4449-
[14] Medentsev, A.G.; Akimenko, V.K. Naphthoquinone metabolites of 52.
the fungi. Phytochemistry, 1998, 47, 935-59.
[38] Hardt, I.H.; Jensen, P.R.; Fenical, W. Neomarinone, and new cyto-
[15] Perry, N.B.; Blunt, J.W.; Munro, M.H.G. A cytotoxic and antifun- toxic marinone derivatives, produced by a marine filamentous bac-
gal 1,4-napthoquinone and related compounds from a New-Zealand
terium (actinomycetales). Tetrahedron Lett., 2000, 41, 2073-76.
brown alga, Landsburgia quercifolia. J. Nat. Prod., 1991, 54, 978- [39] Cai, P.; Kong, F.M.; Ruppen, M.E.; Glasier, G.; Carter, G.T. Hy-
85.
grocins A and B, naphthoquinone macrolides from Streptomyces
[16] Wright, D.A.; Mitchelmore, C.L.; Dawson, R.; Cutler, H.G. The hygroscopicus. J. Nat. Prod., 2005, 68, 1736-42.
influence of water quality on the toxicity and degradation of
[40] Fukuda, D.S.; Mynderse, J.S.; Baker, P.J.; Berry, D.M.; Boeck,
juglone (5-hydroxy 1,4-naphthoquinone). Environ. Technol., 2007, L.D.; Yao, R.C.; Mertz, F.P.; Nakasukasa, W.M.; Mab, J.; Ott, J.;
28, 1091-101.
Counter, F.T.; Esminger, P.W.; Allen, N.E.; Alborn, W.E. A new
[17] Wright, D.A.; Dawson, R.; Cutler, S.J.; Cutler, H.G.; Orano- antibiotic complex produced by Streptomyces aculeolatus. Discov-
Dawson, C.E.; Graneli, E. Naphthoquinones as broad spectrum
ery, taxonomy, fermentation, isolation, characterization and anti-
biocides for treatment of ship's ballast water: Toxicity to phyto- bacterial evaluation. J. Antibiot., 1990, 43, 623-33.
plankton and bacteria. Water Res., 2007, 41, 1294-302.
[41] He, H.Y.; Yang, H.Y.; Luckman, S.W.; Roll, D.M.; Carter, G.T.
[18] Sherif, E.M.; Park, S.-M. Effect of 1,4-naphthoquinone on alumin- Chloroquinocin, a novel chlorinated naphthoquinone antibiotic
ium corrosion in 0.50 M sodium chloride solutions. Electrochim.
from Streptomyces sp., LL-A9227. J. Antibiot., 2002, 55, 1072-75.
Acta, 2006, 51, 1313-21. [42] Bieber, B.; Nuske, J.; Ritzau, M.; Grafe, U. Alnumycin a new
[19] Ciegler, A.; Lee, L.S.; Dunn, J.J. Product of naphthoquinone My-
naphthoquinone antibiotic produced by an endophytic Streptomy-
cotoxins and taxonomy of Penicillium viridicatum. Appl. Environ. ces sp. J. Antibiot., 1998, 51, 381-82.
Microbiol., 1981, 42, 446-49.
[43] Kamijo, M.; Suzuki, T.; Kawai, K.; Fujii, T.; Murase, H. Ytter-
[20] Medentsev, A.G.; Maslov, A.N.; Akimenko, V.K. Naphthoquinone bium-decreasing Streptomyces sp. and its naphthoquinone-pigment
metabolites of fungi of the genus Fusarium and Verticillium -
production in the presence of rare-earth elements. J. Biosci. Bio-
mechanism of the phytotoxic action. Biochem.-Moscow, 1990, 55, eng., 1999, 87, 340-43.
1319-23.
[44] Yamada, Y.; Hou, C.F.; Sasaki, J.; Tahara, Y.; Yoshioka, H. A new
[21] Baker, R.A.; Tatum, J.H.; Nemen, S. Antimicrobial activity of natural quinone, 2-methyl-3-II,III,VIII,IX-octahydro-multiprenyl-
napthoquinones from fusaria. Mycopathologia, 1990, 111, 9-15.
9-1,4-naphthoquinone isolated from Streptomyces albus. Agric.
[22] Johne, S.; Watzke, R.; Meusel, W.; Mollmann, U.; Hartl, A.; Da- Biol. Chem., 1982, 46, 1105-06.
hse, H.M.; Matthes, B.; Seifert, K. Biotechnological production and
Noteworthy Secondary Metabolites Naphthoquinones Current Pharmaceutical Analysis, 2009, Vol. 5, No. 1 61

[45] Fleck, W.F.; Strauss, D.G.; Prauser, H. Napthoquinone antibiotics furan from the stem barks of Newbouldia laevis. Phytochemistry,
from Streptomyces lateritius. 1. Fermentation, isolation and charac- 2003, 64, 583-87.
teristics of the granatomycins-A, granatomycins-C and granatomy- [68] Gafner, S.; Wolfender, J.L.; Nianga, M.; Stoeckli-Evans, H.;
cins-D. Zeits. Allgem. Mikrob., 1980, 20, 543-51. Hostettmann, K. Antifungal and antibacterial naphthoquinones
[46] Fosse, C.; LeTexier, L.; Roy, S.; Delaforge, M.; Gregoire, S.; from Newbouldia laevis roots. Phytochemistry, 1996, 42, 1315-20.
Neuwels, M.; Azerad, R. Parameters and mechanistic studies on the [69] Eyong, K.O.; Krohn, K.; Hussain, H.; Folefoc, G.N.; Nkengfack,
oxidative ring cleavage of synthetic heterocyclic naphthoquinones A.E.; Schulz, B.; Hu, Q.X. Newbouldiaquinone and newbouldia-
by Streptomyces strains. Appl. Microbiol. Biotechnol., 2004, 65, mide: A new naphthoquinone-anthraquinone coupled pigment and
446-56. a new ceramide from Newbouldia laevis. Chem. Pharm. Bull.,
[47] Misic, M.; Rankovic, B.; Sukdolak, S. Antimicrobial activity of 2005, 53, 616-19.
fumarprotocetraric acid, lecanoric acid, protocetraric acid and stic- [70] Eyong, K.O.; Folefoc, G.N.; Kuete, V.; Beng, V.P.; Krohn, K.;
tic acid isolated from different species of lichen. Planta Med., Hussain, H.; Nkengfack, A.E.; Saeftel, M.; Sarite, S.R.; Hoerauf,
2008, 74, 996-96. A. Newbouldiaquinone A: A naphthoquinone-anthraquinone ether
[48] Manojlovic, N.; Vasiljevic, P.; Bogdanovic-Dusanovic, G.; Mano- coupled pigment, as a potential antimicrobial and antimalarial
jlovic, I. Antioxidant activity of some lichen growing in Serbia. agent from Newbouldia laevis. Phytochemistry, 2006, 67, 605-09.
Planta Med., 2008, 74, 996-97. [71] Ali, R.M.; Houghton, P.J.; Raman, A.; Hoult, J.R.S. Antimicrobial
[49] Bessadottir, M.; Bjornsdottir, A.; Omarsdottir, S.; Ogmundsdottir, and antiinflammatory activities of extracts and constituents of
H. M. Effects of protolichesterinic acid, a lichen compound, on the Oroxylum indicum (L.) Vent. Phytomedicine, 1998, 5, 375-81.
ERK1/ERK2 signalling pathway in cancer cells and normal endo- [72] Kizu, H.; Habe, S.; Ishida, M.T.T. Studies on the nepalese crude
thelial cells. Planta Med., 2008, 74, 1022-22. drugs. 17. On the naphthalene related compounds drom the root
[50] Seo, J.M.; Jin, Y.R.; Ryu, C.K.; Kim, T.J.; Han, X.H.; Hong, J.T.; bark of Oroxylum indicum Vent. Yakugaku Zasshi-J. Pharm. Soc.
Yoo, H.S.; Lee, C.K.; Yun, Y.P. JM91, a newly synthesized in- Jpn., 1994, 144, 492-513.
doledione derivative, inhibits rat aortic vascular smooth muscle [73] Shetgiri, N.P.; Kokitkar, S.V.; Sawant, S.N. Radermachera xylo-
cells proliferation and cell cycle progression through inhibition of carpa: The highly efficient source of lapachol and synthesis of its
ERK1/2 and Akt activations. Biochem. Pharmacol., 2008, 75, derivates. Acta Pol. Pharm., 2001, 58, 133-36.
1331-40. [74] Onegi, B.; Kraft, C.; Kohler, I.; Freund, M.; Jenett-Siems, K.;
[51] Rankovic, B.; Misic, M.; Sukdolak, S. The antimicrobial activity of Siems, K.; Beyer, G.; Melzig, M. F.; Bienzle, U.; Eich, E. Herbal
substances derived from the lichens Physcia aipolia, Umbilicaria remedies traditionally used against malaria part 6 - Antiplasmodial
polyphylla, Parmelia caperata and Hypogymnia physodes. World J. activity of napthoquinones and one anthraquinone from Stereo-
Microbiol. Biotechnol., 2008, 24, 1239-42. spermum kunthianum. Phytochemistry, 2002, 60, 39-44.
[52] Krivoshchekova, O.E.; Maximov, O.B.; Stepanenko, L.S.; Mish- [75] Zani, C.L.; Oliveira, A.B.d.; Oliviera, G.d. Furanonaphthoquinones
chenko, N.P. Quinones of the lichen Cetraria cucullata. Phyto- from Tabebuia ochracea. Phytochemistry, 1991, 30, 2379-81.
chemistry, 1982, 21, 193-96. [76] Rogathi, B.K.; Gupta, B.R.; Roy, D.; Kjhanna, R.N. Quinones from
[53] Stepanenko, L.S.; Krivoshchekova, O.E.; Dmitrenok, P.S.; Maxi- Tecoma pentaphylla: constitution of tecomaquinones I a II. Ind. J.
mov, O.B. Quinones of Cetraria islandica. Phytochemistry, 1997, Chem., 1993, 22, 886-89.
46, 565-68. [77] Jacome, R.L.R.P.; de Oliveira, A.B.; Raslan, D.S.; Muller, A.;
[54] Ernst-Russell, M.A.; Elix, J.A.; Chai, C.L.L.; Willis, A.C.; Wagner, H. Analysis of naphtoquimones in Zeyheria montana
Hamada, N.; Nash, T.H. Hybocarpone, a novel cytotoxic naphthaz- crude extracts (bolsa-de-pastor). Quim. Nova, 1999, 22, 175-77.
arin derivative from mycobiont cultures of the lichen Lecanora hy- [78] Akunyili, D.N.; Houghton, P.J.; Raman, A. Antimicrobial activities
bocarpa. Tetrahedron Lett., 1999, 40, 6321-24. of the stembark of Kigelia pinnata. J. Ethnopharmacol., 1991, 35,
[55] Kuk, J.H.; Ma, S.J.; Moon, J.H.; Kim, K.Y.; Choi, S.H.; Park, K.H. 173-77.
Antibacterial and antifungal activities of a naphthoquinone deriva- [79] Houghton, P.J.; Photiou, A.; Uddin, S.; Shah, P.; Browning, M.;
tive isolated from the fruits of Catalpa ovata G. D-ON. J. Micro- KJackson, S.J.; Retsas, S. Activity of extracts of Kigelia pinnata
biol. Biotechnol., 2002, 12, 858-63. against melanoma and renal carcinoma cell lines. Planta Med.,
[56] Jassbi, A.R.; Singh, P.; Jain, S.; Tahara, S. Novel naphthoquinones 1994, 60, 430-33.
from Heterophragma adenophyllum. Helv. Chim. Acta, 2004, 87, [80] Houghton, P.J. The sausage tree (Kigelia pinnata): ethnobotany and
820-24. recent scientific work. South Africa J. Bot., 2002, 68, 14-20.
[57] Singh, P.; Natani, K.; Jain, S.; Arya, K.; Dandia, A. Microwave- [81] Weiss, C.R.; Moideen, S.V.K.; Croft, S.L.; Houghton, P.J. Activity
assisted rapid cyclization of lapachol, a main constituent of Hetero- of extracts and isolated naphthoquinones from Kigelia pinnata
phragma adenophyllum. Nat. Prod. Res., 2006, 20, 207-12. against Plasmodium falciparum. J. Nat. Prod., 2000, 63, 1306-039.
[58] Akunyili, D.N.; Houghton, P.J. Meroterpenoids and naphthoqui- [82] Schmeda-Hirschmann, G.; Papastergiou, F. Naphthoquinone de-
nones from Kigelia pinnata. Phytochemistry, 1993, 32, 1015-18. rivatives and lignans from the Paraguayan crude drug "tayi pyta"
[59] Inouye, K.; Chen, C.C.A naphtoquinone and lignan from the wood (Tabebuia heptaphylla, Bignoniaceae). Z.Naturforsch.(C), 2003,
of Kigelia pinata. Phytochemistry, 1981, 20, 2271-76. 58, 495-501.
[60] Moideen, S.V.K.; Houghton, P.J.; Rock, P.; Croft, S.L.; Aboagye- [83] Viana, L.M.; Freitas, M.R.; Rodrigues, S.V.; Baumann, W. Extrac-
Nyame, F. Activity of extracts and naphthoquinones from Kigelia tion of lapachol from Tabebuia avellanedae wood with supercritical
pinnata against Trypanosorna brucei brucei and Trypanosoma CO2: An alternative to Soxhlet extraction? Bras. J. Chem. Eng.,
brucei rhodesiense. Planta Med., 1999, 65, 536-40. 2003, 20, 317-25.
[61] Jain, P.S.; Belsare, D.R.; Pal, S.C.; Mandal, S.C. Phytochemical [84] de Oliveira, A.B.; Raslan, D.S.; de Oliveira, G.G.; Maia, J.G.S.
analysis of Kigelia pinnata. Asian J. Chem., 2006, 18, 3224-26. Lignans and naphthoquinones from Tabebuia incana. Phytochemis-
[62] Duarte, D.S.; Dolabela, M.F.; Salas, C.E.; Raslan, D.S.; Oliveiras, try, 1993, 34, 1409-12.
A.B.; Nenninger, A.; Wiedermann, B.; Wagner, H.; Lombardi, J.; [85] Diaz, F.; Medina, J.D. Furanonaphthoquinones from Tabebuia
Lopes, M.T.P. Chemical charakterization and biological activity of ochracea ssp. neochrysantha. J. Nat. Prod., 1996, 59, 423-24.
Macfadyena unguis-cati (Bignoniaceae). J. Pharm. Pharmacol., [86] Fujimoto, Y.; Eguchi, T.; Murasaki, C.; Ohashi, Y.; Kakinuma, K.;
2000, 52, 347-52. Takagaki, H.; Abe, M.; Inazawa, K.; Yamazaki, K.; Ikekawa, N.;
[63] Singh, P.; Singh, A. Quinonoid constituents of the bark of Mark- Yoshikawa, O.; Ikekawa, T. Studies on the Structure and Stereo-
hamia stipulata Wall. Pharm., 1980, 35, 701-02. chemistry of Cytotoxic Furanonaphthoquinones from Tabebuia-
[64] Kanchanapoom, T.; Kasai, R.; Yamasaki, K. Phenolic glycosides Impetiginosa - 5-Hydroxy-2-(1-Hydroxyethyl)Naphtho [2,3-B]Fur
from Markhamia stipulata. Phytochemistry, 2002, 59, 557-63. an-4,9-Diones and 8-Hydroxy-2-(1-Hydroxyethyl) Naphtho[2,3-B]
[65] Houghton, P.J.; Pandey, R.; Hawkes, J.E. Naphthoquinones an an Furan-4,9-Diones. J. Chem. Soc.-Perkin Trans., 1991, 1, 2323-
alkaloid from roots of Newbouldia laevis. Phytochemistry, 1994, 2327.
35, 1602-03. [87] Warashina, T.; Nagatani, Y.; MNoro, T. Constituents of the bark of
[66] Houghton, P.J. Napthoquinones and an alkaloid from roots of New- Tabebuia impetiginosa. Chem. Pharm. Bull., 2006, 54, 14-20.
bouldia laevis. Phytochemistry, 1994, 36, 1563-63. [88] Warashina, T.; Nagatani, Y.; Noro, T. Constituents from the bark
[67] Gormann, R.; Kaloga, M.; Li, X.C.; Ferreira, D.; Bergenthal, D.; of Tabebuia impetiginosa. Phytochemistry, 2004, 65, 2003-11.
Kolodziej, H. Furanonaphthoquinones, atraric acid and a benzo-
62 Current Pharmaceutical Analysis, 2009, Vol. 5, No. 1 Babula et al.

[89] Warashina, T.; Nagatani, Y.; Noro, T. Further constituents from the [112] Coder, K.D. Seasonal changes of juglone potential in leaves of
bark of Tabebuia impetiginosa. Phytochemistry, 2005, 66, 589-98. black walnut (Juglans nigra L.). J. Chem. Ecol., 1983, 9, 1203-12.
[90] Onegia, B.; Kraftb, C.; Kohlerb, I.; Freundb, M.; Jenett-Siemsb, K.; [113] Colaric, M.; Veberic, R.; Solar, A.; Hudina, M.; Stampar, F. Pheno-
Siemsc, K.; Beyerd, G.; Melzigd, M.F.; Bienzlee, U.; Eichb, E. An- lic acids, syringaldehyde, and juglone in fruits of different cultivars
tiplasmodial activity of naphthoquinones and one anthraquinone of Juglans regia L. J. Agric. Food Chem., 2005, 53, 6390-96.
from Stereospermum kunthianum. Phytochemistry, 2002, 60, 39- [114] Hadjmohammadi, M.R.; Kamel, K. Determination of juglone (5-
44. hydroxy 1,4-naphthoquinone) in Pterocarya fraxinifolia by RP-
[91] Ito, C.; Katsuno, S.; Kondo, Y.; Tan, H.T.W.; Furukawa, H. HPLC. Iran J. Chem. Chem. Eng. Int. Engl. Ed., 2006, 25, 73-76.
Chemical constituents of Avicennia alba. Isolation and structural [115] Sankaram, A.V.B.; Srinivasarao, A.; Sidhu, G.S. Chitranone - a
elucidation of new naphthoquinones and their analogues. Chem. new binaphthoquinone from Plumbago zeylanica. Phytochemistry,
Pharm. Bull., 2000, 48, 339-43. 1976, 15, 237-38.
[92] Jones, W.P.; Lobo-Echeverri, T.; Mi, Q.W.; Chai, H.; Lee, D.; [116] Gunaherath, G.M.; Kamal, B.L., G.A.A.; Sultanbawa, M.; Uvais,
Soejarto, D.D.; Cordell, G.A.; Pezzuto, J.M.; Swanson, S.M.; S.; Balasubramaniam, S. 1,2(3)-tetrahydro-3,3´-biplumbagin: a
Kinghorn, A.D. Antitumour activity of 3-chlorodeoxylapachol, a naphthalenone and other constituents from Plumbago zeylanica.
naphthoquinone from Avicennia germinans collected from an ex- Phytochemistry, 1983, 22, 1245-48.
perimental lot in southern Florida. J. Pharm. Pharmacol., 2005, 57, [117] Dinda, B.; Saha, S. A new binaphthoquinone from Plumbago zey-
1101-08. lanica Linn. Ind. J. Chem., 1989, 28B, 984-86.
[93] Kreher, B.; Neszmelyi, A.; Wagner, H. Naphthoquinones from [118] Kamal, G.M.; HGunaherath, B.; Gunatilaka, A.A.L.; Thomson,
Dionaea muscipula. Phytochemistry, 1990, 29, 605-06. R.H. Structure of plumbazeylone - a novel trimer of plumbagin
[94] Babula, P.; Mikelova, R.; Adam, V.; Potesil, D.; Zehnalek, J.; from Plumbago zeylanica. Tetrahedron Lett., 1984, 25, 4801-04.
Kizek, R.; Klejdus, B.; Havel, L.; Sladky, Z. Chromatographic [119] Li, H.L. Plumbaginaceae. In: Editorial Committee of the Flora of
analysis of naphthoquinones in plants. Chem. Listy, 2006, 100, 271- Thaiwan Eds.; Flora of Thaiwan, 2nd ed., Vol. IV. Editorial Com-
76. mittee of the Flora of Thaiwan, 1998, 79-82.
[95] Budzianowski, J.; Budzianowska, A.; Kromer, K. Naphtalene glu- [120] Premakumari, P.; Rathinam, K.; Santhakumari, G. Antifertility
coside and other phenolics from the shoot and callus cultures of activity of plumbagin. Ind. J. Med. Res., 1977, 65, 829-38.
Drosophyllum lusitanicum. Phytochemistry, 2002, 61, 421-25. [121] dePaiva, S.R.; Figueiredo, M.R.; Aragao, T.V.; Kaplan, M.A.C.
[96] Crouch, I. J.; Finnie, J. F.; vanStaden, J. Studies on the isolation of Antimicrobial activity in vitro of plumbagin isolated from Plum-
plumbagin from in vitro and in vivo grown Drosera species. Plant. bago species. Floresta e Ambiente, 2003, 98, 959-61.
Cell. Tiss. Org., 1990, 21, 79-82. [122] Dinda, B.; Chel, G. 6-hydroxyplumbagin, a naphthoquinone from
[97] Kamarainen, T.; Uusitalo, J.; Jalonen, J.; Laine, K.; Hohtola, A. Plumbago indica. Phytochemistry, 1992, 31, 3652-53.
Regional and habitat differences in 7-methyljuglone content of [123] dePaiva, S.R.; Lima, L.A.; Figueiredo, M.R.; Kaplan, M.A.C.
Finnish Drosera rotundifolia. Phytochemistry, 2003, 63, 309-14. Plumbagin quantification in roots of Plumbago scandens L. ob-
[98] Repák, M.; Galambosi, B.; Takkunen, N. The production of 7- tained by different extraction techniques. Annals Brasil. Acad. Sci.,
methyljuglone, quercetin and kaempfrol by D. anglica and D. ro- 2004, 76, 499-504.
tundifolia. Biologia, 2000, 55, 429-33. [124] dePaiva, S.R.; Fontoura, L.D.; Figueiredo, M.R.; Mazzei, J.L.;
[99] Budzianovski, J. Naphthohydroquinone glukosides of Drosera Kaplan, M.A.C. Chromatographic profile of two Plumbaginaceae
rotundifolia and Drosera intermedia from in vitro cultures. Phyto- species: Plumbago scandens L. and Plumbago auriculata LAM.
chemistry, 1996, 42, 1145-47. Quim. Nova, 2002, 25, 717-21.
[100] Budzianovski, J. 2-methylnaphtazarin-5-O-glukoside from the [125] Herble, M.R.; Narayanaswamy, S.; Chadba, M.S. Tissue differen-
methanol extracts of in vitro cultures of Drosera species. Phyto- tiation and plumbagin synthesis in variant cell strains of Plumbago
chemistry, 1997, 44, 75-78. zeylanica L. in vitro. Plant Sci. Lett., 1974, 2, 405-09.
[101] Budzianovski, J. Naphthoquinone glukosides of Drosera gigantea [126] Komaraiah, P.; Naga Amrutha, R.; Kavi Kishor, P.B.;
from in vitro cultures. Planta Med., 2000, 66, 667-69. Ramakrishna, S.V. Elicitor enhanced production of plumbagin in
[102] Marczak, L.; Kawiak, A.; Lojkowska, E.; Stobiecki, M. Secondary suspension cultures of Plumbago rosea L. Enzyme Microb. Bio-
metabolites in in vitro cultured plants of the genus Drosera. Phyto-
tech., 2002, 31, 634-39.
chem. Anal., 2005, 16, 143-49. [127] Komaraiah, P.; Ramakrishna, S.V.; Reddanna, P.; Kavi Kishor,
[103] Hook, I.L.I. Naphthoquinone contents of in vitro cultured plants
P.B. Enhanced production of plumbagin in immobilized cells of
and cell suspensions of Dionaea muscipula and Drosera species.
Plumbago rosea by elicitation and in situ adsorption. J. Biotech-
Plant. Cell. Tiss. Org., 2001, 67, 281-85.
nol., 2003, 101, 181-87.
[104] Pakulski, G.; Budzianowski, J. Ellagic acid derivates and naphtho-
[128] Komaraiah, P.; Kishor, P.B.K.; Ramakrishna, S.V. Production of
quinones of Dionaea muscipula from in vitro cultures. Phytochem-
plumbagin from cell cultures of Plumbago rosea L. Biotechnol.
istry, 1996, 41, 775-78.
Lett., 2001, 23, 1269-72.
[105] Nahalka, J.; Nahalkova, J.; Gemeiner, P.; Blanarik, P. Elicitation of
[129] dePaiva, S.R.; Figueiredo, M.R.; Kaplan, M.A.C. Isolation of sec-
plumbagin by chitin and its release into the medium in Drosophyl-
ondary metabolites from roots of Plumbago auriculata Lam. by
lum lusitanicum Link. suspension cultures. Biotechnol. Lett., 1998,
countercurrent chromatography. Phytochem. Anal., 2005, 16, 278-
20, 841-45.
[106] Jayaram, K.; Prasad, M.N.V. Rapidly in vitro multiplied Drosera 81.
as reliable source for plumbagin bioprospection. Curr. Sci., 2005, [130] Navaei, M.N.; Mirza, M.; Dini, M. Chemical composition of the
89, 447-48. essential oil of Plumbago europaea L. roots from Iran. Flavour
[107] Miyoishi, E.; Shizuri, Y.; Yamamura, S. Isolation and structures of Frag. J., 2005, 20, 213-14.
diomuscinone and diomuscipulone from Dionaea muscipula. Phy- [131] Mallavadhani, U.V.; Sahu, G.; Muralidhar, J. Screening of Plum-
tochemistry, 1984, 23, 2385-87. bago species for the bio-active marker plumbagin. Pharm. Biol.,
[108] Krenn, L.; Beyer, G.; Pertz, H.H.; Karall, E.; Kremser, M.; Galam- 2002, 40, 508-11.
bosi, B.; Melzig, M.E. In vitro antispasmodic and Antiinflamma- [132] Verma, P.C.; Singh, D.; Rahman, L.U.; Gupta, M.M.; Banerjee, S.
tory effects of Drosera rotundifolia. Drug Res., 2004, 54, 402-05. In vitro studies in Plumbago zeylanica: rapid micropropagation and
[109] Jayaram, K.; Prasad, M.N.V. Drosera indica L. and D. burmanii establishment of higher plumbaginyielding hairy root cultures. J.
Vahl., medicinally important insectivorous plants in Andhra Plant Physiol., 2002, 159, 547-52.
Pradesh - regional threats and conservation. Curr. Sci., 2006, 91, [133] Alnuri, M.A.; Hannoun, M.A.; Zatar, N.A. Plumbagin, a naturally-
943-46. occuring naphthoquinone-its isolation, spectrophotometric deter-
[110] Hirakawa, K.; Ogiue, E.; Motoyoshiya, J.; Yajima, M. Naphtho- mination in roots, stems and leaves in Plumbago europaea. Spectr.
quinones from Juglandaceae. Phytochemistry, 1996, 25, 1494-95. Lett., 1994, 27, 409-19.
[111] Duroux, L.; Delmotte, F.M.; Lancelin, J.M.; Keravis, G.; Jay- [134] Yue, J.; Lin, Z.; Wang, D.; Feng, Y.; Sun, H. Plumbasides A-C
Allemand, C. Insight into naphthoquinone metabolism: beta- three naphthoquinone derivates from Ceratostigma minus. Phyto-
glucosidase-catalysed hydrolysis of hydrojuglone beta-D- chemistry, 1994, 35, 1023-26.
glucopyranoside. Biochem. J., 1998, 333, 275-83.
Noteworthy Secondary Metabolites Naphthoquinones Current Pharmaceutical Analysis, 2009, Vol. 5, No. 1 63

[135] Yue, J.; Xu, J.; Zhao, Y.; Sun, H.D.; Lin, Z. Chemical components [157] Ali, B.H.; Bashir, A.K.; Tanira, M.O.M. Anti-inflammatory, Anti-
from Ceratostigma willmottianum. J. Nat. Prod., 1997, 60, 1031- pyretic and Analgesic effects of Lawsonia inermis L. (Henna) in
33. rats. Pharmacology, 1995, 51, 356-63.
[136] Yue, J.M.; Zhao, Y.; Zhao, Q.S.; Wu, H.M.; Xu, J.F.; Lin, Z.W.; [158] Takeda, Y.; Fatope, M. O. New phenolic glucosides from Lawsonia
Sun, H.D. A novel compound from Ceratostigma minus. Chin. inermis. J. Nat. Prod., 1988, 51, 725-29.
Chem. Lett., 1998, 9, 647-49. [159] Bakkali, A.T.; Jaziri, M.; Foriers, A.; Vander-Heyden, Y.; Van-
[137] Gafner, R.; Chapuls, J.C.; Msonthi, J.D.; Hostettmann, K. Cyto- haelen, N.; Homes, J. Lawsone accumulation in normal and trans-
toxic napthtoquinones, molluscidals saponins and flavonols from formed cultures of henna, Lawsonia inermis. Plant Cell Tissue
Diospyros zombensis. Phytochemistry, 1987, 26, 2501-04. Org., 1997, 51, 83-87.
[138] Gu, J.Q.; Graf, T.N.; Lee, D.H.; Chai, H.B.; Mi, Q.W.; Kardono, [160] Ahmed, S.; Rahman, A.; Alam, A.; Saleem, M.; Athar, M.; Sultana,
L.B.S.; Setyowati, F.M.; Ismail, R.; Riswan, S.; Farnsworth, N.R.; S. Evaluation of the efficacy of Lawsonia alba in the alleviation of
Cordell, G.A.; Pezzuto, J.M.; Swanson, S.M.; Kroll, D.J.; Falkin- carbon tetrachloride-induced oxidative stress. J. Ethnopharmacol.,
ham, J.O.; Wall, M.E.; Wani, M.C.; Kinghorn, A.D.; Oberlies, 2000, 69, 157.
N.H. Cytotoxic and antimicrobial constituents of the bark of [161] McMillan, D.C.; Sarvate, S.D.; Oatis, J.E.; Jollow, D. Role of
Diospyros maritima collected in two geographical locations in In- Oxidant stress in lawsone-induced hemolytic anemia. Toxicol. Sci.,
donesia. J. Nat. Prod., 2004, 67, 1156-1161. 2004, 82, 647-55.
[162] Mita, G.; Gerardi, C.; Miceli, A.; Bollini, R.; Deleo, P. Pigment
[139] Ganapaty, S.; Thomas, P.S.; Karagianis, G.; Waterman, P.G.; Brun,
production from In vitro cultures of Ankanna tinctoria Tausch.
R. Antiprotozoal and cytotoxic naphthalene derivates from Diospy-
Plant Cell Reports, 1994, 13, 406-10.
ros assimilis. Phytochemistry, 2006, 67, 1950-56.
[163] Bieber, L.W.; Brebs, H.C.; Schafer, W. Further meroterpenoid
[140] Ganapaty, S.; Thomas, P.S.; Karagianis, G. Mono and dimeric naphthoquinones from Cordia corymbosa. Phytochemistry, 1994,
naphthalene derivatives from the roots of Diospyros assimilis. Nat. 35, 1027-28.
Prod. Res., 2006, 20, 783-87. [164] de Menezes, J. E. S. A.; Lemos, T. L. G.; Pessoa, O. D. L.; Braz,
[141] Tangmouo, J.G.; Meli, A.L.; Komguem, J.; Kuete, V.; Ngounou, R.; Montenegro, R. C.; Wilke, D. V.; Costa-Lotufo, L. V.; Pessoa,
F.N.; Lontsi, D.; Beng, V.P.; Choudhary, M.I.; Sondengam, B.L. C.; de Moraes, M. O.; Silveira, E. R. A cytotoxic meroterpenoid
Crassiflorone, a new naphthoquinone from Diospyros crassiflora benzoquinone from roots of Cordia globosa. Planta Med., 2005,
(Hien). Tetrahedron Lett., 2006, 47, 3067-70. 71, 54-58.
[142] Prajoubklang, A.; Sirithunyalug, B.; Charoenchai, P.; Suvannakad, [165] Staniforth, V.; Wang, S.Y.; Shyur, L.F.; Yang, N.S. Shikonins,
R.; Sriubolmas, N.; Piyamongkol, S.; Kongsaeree, P.; Kittakoop, P. phytocompounds from Lithospermum erythrorhizon, inhibit the
Bioactive deoxypreussomerins and dimeric naphthoquinones from transcriptional activation of human tumor necrosis factor alpha
Diospyros ehretioides fruits: Deoxypreussomerins may not be plant promoter in vivo. J. Biol. Chem., 2004, 279, 5877-85.
metabolites but may be from fungal epiphytes or endophytes. [166] Ozgen, U.; Coskun, M.; Kazaz, C.; Secen, H. Naphthoquinones
Chem. Biodiver., 2005, 2, 1358-367. from the roots of Onosma argentatum Hub.-Mor. (Boraginaceae).
[143] Tangmouo, J.G.; Lontsi, D.; Ngounou, F.N.; Kuete, V.; Meli, A.L.; Turk. J. Chem., 2004, 28, 451-54.
Manfouo, R.N.; Kamdem, H.W.; Tane, P.; Beng, V.P.; Sondengam, [167] Hu, Y.; Jiang, Z.H.; Leung, K.S.Y.; Zhao, Z.Z. Simultaneous de-
B.L.; Connoly, J.D. Diospyrone, a new coumarinylbinaphthoqui- termination of naphthoquinone derivatives in Boraginaceous herbs
none from Diospyros canaliculata (Ebenaceae): structure and an- by high-performance liquid chromatography. Anal. Chim. Acta,
timicrobial activity. Bull. Chem. Soc. Ethiop., 2005, 19, 81-88. 2006, 577, 26-31.
[144] Ganapaty, S.; Thomas, P.S.; Mallika, B.N.; Balaji, S.; Karagianis, [168] Touno, K.; Tamaoka, J.; Ohashi, Y.; Shimomura, K. Ethylene
G.; Waterman, P.G. Dimeric napthoquinones from Diospyros dis- induced shikonin biosynthesis in shoor culture of Lithospermum
color. Biochem. Syst. Ecol., 2005, 33, 313-15. erythrhorhizon. Plant Physiol. Chem., 2005, 43, 101-05.
[145] Dinda, B.; Das, S.K.; Bhattacharya, A.; De, U.C.; Harigaya, Y. [169] Touno, K.; Harada, K.; Yoshimatsu, K.; Yazaki, K.; Shimomura,
Chemical constituens of Diospyros nigra. J. Indian Chem. Soc., K. Shikonin derivative formation on the stem of cultured shoots in
2002, 79, 553-54. Lithospermum erythrorhizon. Plant Cell Reports, 2000, 19, 1121-
[146] Khan, R.M.; Timi, D. Constituents of the root and stem barks, 26.
leaves and fruits of Diospyros hallierii. Fitoterapia, 1999, 70, 320- [170] Lin, L.D.; Wu, J.Y. Enhancement of shikonin production in single-
21. and two-phase suspension cultures of Lithospermum erythrorhizon
[147] Khan, R.M.; Timi, D. Constituents of the root and stem barks of cells using low-energy ultrasound. Biotechnol. Bioeng., 2002, 78,
Diospyros villosiuscula. Fitoterapia, 1999, 70, 209-11. 81-88.
[171] Chung, B.Y.; Lee, Y.B.; Baek, M.H.; Kim, J.H.; Wi, S.G.; Kim,
[148] Evans, P.H.; Bowers, W.S.; Litaudon, M.; Sevenet, T. Plumbagin
S.J. Effect of low-dose gamma-irradiation on production of
from Diospyros olen. Molecules, 1999, 4, m93-u10.
shikonin derivates in callus cultures of Lithospermum
[149] Khan, R.M.; Rwekika, E. 6,8´-bisdiosquinone from Diospyros erythrorhizon S. Radiat. Phys. Chem., 2006, 75, 1018-23.
mafiensis. Phytochemistry, 1999, 50, 143-46. [172] Brigham, L.A.; Michaels, P.J.; Flores, H.E. Developmental and
[150] Mallavadhani, U.V.; Panda, A.K.; Rao, Y.R. Pharmacology and induced production of shikonin in Lithospermum erythrorhizon
chemotaxonomy of Diospyros. Phytochemistry, 1998, 49, 901-51. roots. Plant Physiol., 1997, 114, 1133.
[151] Cai, L.; Wei, G.X.; van der Bijl, P.; Wu, C.D. Namibian Chewing [173] Yazaki, K.; Tanaka, S.; Matsuoka, H.; Sato, F. Stable transforma-
Stick, Diospyros lycioides, contains antibacterial compounds tion of Lithospermum erythrorhizon by Agrobacterium rhizogenes
against oral patogens. J. Agric. Food Chem., 2000, 48, 909-14. and shikonin production of the transformants. Plant Cell Reports,
[152] Khan, R. M.; Rwekika, E. A binaphthoquinone of Diospyros 1998, 18, 214-19.
greeniwayi. Phytochemistry, 1998, 49, 2501-04. [174] Haghbeen, K.; Mozaffarian, V.; Ghaffari, F.; Pourazeezi, E.; Saraji,
[153] Matsutake, H.; Kazuhito, O.; Seichi, Y. Bioactive naphthoquinone M.; Joupari, M.D. Lithospermum officinale callus produces ahikal-
derivates from Diospiros maritima Blume. Chem. Pharm. Bull., nin. Biologia, 2006, 61, 463-67.
1998, 46, 1089-103. [175] Syklowska-Baranek, K.; Pietrosiuk, A.; Furmanowa, M.; Szypula,
[154] Ito, Y.; Hayashi, Y.; Kato, A. Antifungal compounds drom trees of W.; Jeziorek, M. Production of shikonin derivatives in transgenic
the genus Diospyrus with complete assignment of nuclear- roots of Lithospermum canescens (Michx.) Lehm. cultivated in
magnetic-resonance data. Mokuzai Gakkaishi, 1995, 41, 694-98. mist bioreactor. Planta Med., 2008, 74, 1161-61.
[155] Mebe, P.; Cordell, G.A.; Pezzuto, J.M. Pentacyclic triterpenes and [176] Li, Q.W.; Chen, Z.G.; Zhou, X.; Pan, A.H.; Wang, L.S. Determina-
naphthoquinones from Euclea divinorum. Phytochemistry, 1998, tion of Shikonin in Arnebia euchroma (Royle) Johnst by capillary
47, 311-14. electrophoresis. Chin. J. Anal. Chem., 2006, 34, 991-94.
[156] Joubert, A.; van der Kooy, F.; Meyer, J.J.M.; Lall, N. HPLC in the [177] Pietrosiuk, A.; Wiedenfeld, H. Shikonin derivates from Lithosper-
comparative study of the content of naphthoquinones (quinonoid mum canescens. Pharm. Biol., 2005, 43, 189-91.
constituents) in Euclea species of South Africa. Chromatographia, [178] Miller, A.K.; Smith, C.L.; Jones, A.M.; VanVickle, N.L.; Kelley,
2006, 64, 399-403. R.B. Investigation of the naphthazarins found in Lithospermum
canescens and Lithodora diffusa. Abstr. Papers Am. Chem. Soc.,
2004, 227, 623.
64 Current Pharmaceutical Analysis, 2009, Vol. 5, No. 1 Babula et al.

[179] Papageorgiou, V.P.; Digenis, G.A. Isolation of 2 new alkannin yagul, S.; Siripong, P. Synthesis of novel rhinacanthins and related
esters from Alkanna tinctoria. Planta Med., 1980, 39, 81-84. anticancer naphthoquinone esters. J. Med. Chem., 2004, 47, 4427-
[180] Assimopoulu, A.N.; Boskou, D.; Papageorgiou, V.P. Antioxidant 38.
activities of alkannin, shikonin and Alkanna tinctoria root extracts [201] Siripong, P.; Yahuafai, J.; Shimizu, K.; Ichikawa, K.; Yonezava,
in oil substrates. Food Chem., 2004, 87, 433-38. S.; Asai, T.; Kanokmedakul, K.; Ruchirawat, S.; Oku, N. Induction
[181] Urbanek, H.; Bergier, K.; Saniewski, M.; Patykowski, J. Effect of of apoptosis in tumor cells by three naphthoquinone esters isolated
jasmonates and exogenous polysaccharides on production of alkan- from Thai medicinal plant: Rhinacanthus nasutus Kurz. Biol.
nin pigments in suspension cultures of Alkanna tinctoria. Plant Cell Pharm. Bull., 2006, 29, 2070-76.
Reports, 1996, 15, 637-41. [202] Anh, N.H.; Ripperger, H.; Porzel, A.; Sung, T.S.; Adam, G.
[182] Ozgen, U.; Ozturk, M.; Atila, A.; Sevindik, H.G.; Kadioglu, Y.; Tetralones from Ascistrocladus cochinchinensis. Phytochemistry,
Coskun, M. Quantitative HPLC analysis of deoxyshikonin, acetyl 1997, 44, 549-52.
shikonin and 3-hydroxy-isovaleryl shikonin in Onosma arme- [203] Wang, S.; Zhang, P.C.; Chen, R.Y.; Yu, D.Q. Studies on the
niacum root. Planta Med., 2007, 73, 883-83. chemical constituents of the roots of Goniothalamus cheliensis Hu.
[183] Ioset, J.R.; Marston, A.; Gupta, M.P.; Hostettmann, K. Antifungal Acta Chim. Sin., 2001, 59, 1823-26.
and larvicidal meroterpenoid naphthoquinones and a naphthoxirene [204] Bringman, G.; Rischer, H.; Wohlfahrt, M.; Schlauer, J.; Ake Assi,
from the roots of Cordia linnaei. Phytochemistry, 1998, 47, 729-34. L. Droserone from cell cultures of Triphyoplyllum peltatum (Dion-
[184] Bieber, L. W.; Messana, I.; Lins, S. C. N.; Filho, A. A. D.; Chiap- cophyllaceae) and its biosyntetic origin. Phytochemistry, 2000, 53,
peta, A. A.; Demello, J. F. Meroterpenoid naphthoquinones from 339-44.
Cordia corymbosa. Phytochemistry, 1990, 29, 1955-59. [205] Kitagawa, R.R.; Raddi, M.S.G.; dos Santos, L.C.; Vilegas, W. A
[185] Bozan, B.; Baser, K.H.C.; Kara, B. Quantitative determination of new cytotoxic naphthoquinone from Paepalanthus latipes. Chem.
naphthaquinones of Arnebia densiflora (Nordm.) Ledeb. by an im- Pharm. Bull., 2004, 52, 1487-88.
proved high-performance liquid chromatographic method. J. [206] Varanda, E.A.; Varella, S.D.; Rampazo, R.A.; Kitagawa, R.R.;
Chromatogr. A, 1997, 782, 133-36. Raddi, M.S.G.; Vilegas, W.; dos Santos, L.C. Mutagenic and cyto-
[186] Hayashi, T.; Smith, F.T.; Lee, K.H. Antitumor agents. 89. psycho- toxic effect of planifolin: A naphthopyranone dimer isolated from
rubrin, a new cytotoxic naphthoquinone from Psychotria rubra and Paepalanthus planifolius. Toxicol. Vitro, 2006, 20, 664-68.
its structure-activity-relationships. J. Med. Chem., 1987, 30, 2005- [207] Fournet, A.; Angelo, A.; Munoz, V.; Roblot, F.; Hocquemiller, R.;
08. VCave, A. Biological and chemical studies of Pera benensis, a Bo-
[187] Itokawa, H.; Ibraheim, Z.Z.; Qiao, Y.F.; Takeya, K. Anthraqui- livian plant used in folk medicine as a treatment of cutaneous
nones, naphthohydroquinones and naphthohydroquinone dimers leishmaniasis. J. Ethnopharmacol., 1992, 32, 159-64.
from Rubia cordifolia and their cytotoxic activity. Chem. Pharm. [208] Lee, C.H.; Lee, H.S. Color alteration and acaricidal activity of
Bull., 1993, 41, 1869-72. juglone isolated from Caesalpinia sappan heartwoods against Der-
[188] Aung, H.H.; Chia, L.S.; Goh, N.K.; Chia, T.F.; Ahmed, A.A.; Pare, matophagoides spp. J. Microbiol. Biotechnol., 2006, 16, 1591-96.
P.W.; Mabry, T.J. Phenolic constituents from the leaves of the car- [209] Hostettmann, K. Strategy for the Biological and Evaluation of
nivorous plant Nepenthes gracilis. Fitoterapia, 2002, 73, 445-47. Plant Extracts, práce institutu Farmakognosie a Fytochemie. Uni-
[189] Cannon, J.R.; Lojanopiwana, V.; Raston, C.L.; Sinchai, W.; White, versita Lausanne: Lausanne, vcarsko, 1999.
A.H. The quinones of Nepenthes rafflesiana. The crystal structure [210] Williams, C.A.; Harborne, J.B.; Goldblatt, P. Correlations between
of 2,5-dihydroxy-3-methoxy-7-methylnaphto-1,4-quinone (ne- phenolic patterns and tribal classification in the family Iridaceae.
penthone E) and a synthesis of 2,5-dihyroxy-3-methoxy-7- Phytochemistry, 1986, 25, 2135-54.
methylnaphto-1,4-quinone (nepenthone C). J. Chem., 1980, 33, [211] Alves, T.M.A.; Kloos, H.; Zani, C.L. Eleutherinone, a novel fungi-
1073-93. toxic naphthoquinone from Eleutherine bulbosa (Iridaceae). Mem.
[190] Rizzacasa, M.A.; Sargent, V.M. The structure and synthesis of Inst. Oswaldo Cruz, 2003, 98, 709-12.
Nepenthone-A, a naphthoquinone from Nepenthes rafflesiana. J. [212] Hara, H.; Maruyama, N.; Yamashita, S.; Hayashi, Y.; Lee, K.H.;
Chem. Soc. Perkin. Trans., 1987, 1, 2017-22. Bastow, K.F.; Chairul, M.R.; Imakura, Y. Elecanacin, a novel new
[191] Rischer, H.; Hamm, A.; Bringman, G. Nepenthes insignis uses a naphthoquinone from the bulb of Eleutherine americana. Chem.
C2-portion of the carbon skelet of L-alanine acquired via its car- Pharm. Bull., 1997, 45, 1714-16.
nivorous organs, to build up the alleochemical plumbagin. Phyto- [213] Colegate, S.M.; Dorling, P.R.; Huxtable, C.R. Stypandrone - a
chemistry, 2002, 59, 603-09. toxic naphthalene-1,4,-quinone from Stypandra imbricata and Di-
[192] Glennie, C.W.; Bohm, B.C. The isolation of 2-hydroxy-1,4- anella revoluta. Phytochemistry, 1987, 26, 979-81.
naphthoquinone from Impatiens balsamina L. (Balsaminaceae). [214] Byrne, L.T.; Colegate, S.M.; Dorling, P.R.; Huxtable, C.R. Imbri-
Can. J. Biochem., 1965, 43, 293-95. catonol, a Naphthol Naphthoquinone Dimer Isolated From Stypan-
[193] Panichayupakarant, P. Naphthoquinone Formation in Impatiens dra imbricata and Dianella revoluta. Aust. J. Chem., 1987, 40,
balsamina Cell Cultures. Pharm. Biol., 2001, 39, 1-4. 1315-20.
[194] Panichayupakarant, P.; Noguchi, H.; De-Eknamkul, W.; Sankawa, [215] Ali, S.; Sing, P.; Thomson, R.H. Naturally occured Quinones. Part
U. Naphthoquinones ans coumarins from Impatiens balsamina root 28. Sesquiterpenoid quinones and related compounds from Hibis-
cultures. Phytochemistry, 1995, 40, 1144. cus tiliaceus. J. Chem. Soc. Perkin. Trans., 1980, 1, 257-59.
[195] Wu, T.S.; Hsu, H.C.; Wu, P.L.; Leu, Y.L.; Chan, Y.Y.; Chern, [216] Ogasawara, T.; Chiba, K.; Tada, M. Production in high-yield of a
C.Y.; Yeh, M.Y.; Tien, H.J. Naphthoquinone esters from the root naphthoquinone by a hairy root culture of Sesamum indicum. Phy-
of Rhinacanthus nasutus. Chem. Pharm. Bull., 1998, 46, 413-18. tochemistry, 1993, 33, 1095-98.
[196] Sendl, A.; Chen, J.L.; Jolad, S.D.; Stoddart, C.; Rozhon, E.; Ker- [217] Dai, J.R.; Decosterd, L.A.; Gustafson, K.R.; Cardellina, J.H.; Gray,
nan, M.; Nanakorn, W.; Balick, M. Two new naphthoquinones with G.N.; Boyd, M.R. Novel napthoquinones from Conospermum in-
antiviral activity from Rhinacanthus nasutus. J. Nat. Prod., 1996, curvum. J. Nat. Prod., 1994, 57, 1511-16.
59, 808-11. [218] Decosterd, L.A.; Parsons, I.C.; Gustafson, K.R.; Cardellina, J.H.;
[197] Wu, T.S.; Tien, H.J.; Yeh, M.Y.; Lee, K.H. Isolation and cytotoxic- McMahon, J.B.; Cragg, G.M.; Murata, Y.; Pannell, L.K.; Steiner,
ity of Rhinacanthin-A and Rhinacanthin-B, 2 naphthoquinones J.R.; Clardy, J.; Boyd, M.R. HIV inhibitory natural products. 11.
from Rhinacanthus nasutus. Phytochemistry, 1998, 27, 3787-88. Structure, absolute stereochemistry and synthesis of conocucurvone
[198] Wu, T.S.; Hsu, H.C.; Wu, P.L.; Teng, C.M.; Wu, Y.C. Rhinacan- A, potent, novel HIV-inhibitory napthoquinone trimer rom a Cono-
thin-Q, a naphthoquinone from Rhinacanthus nasutus and its bio- spermum sp. J. Am. Chem. Soc., 1993, 115, 6673-79.
logical activity. Phytochemistry, 1998, 49, 2001-03. [219] Butler, M.S.; Katavic, P.L.; Forster, P.I.; Guymer, G.P. Twoo new
[199] Gotoh, A.; Sakaeda, T.; Kimura, T.; Shirakawa, T.; Wada, Y.; naphthoquinones from the roots of Conospermum sphacelatum.
Wada, A.; Kimachi, T.; Takemoto, Y.; Iida, A.; Iwakawa, S.; Hirai, Aust. J. Chem., 1999, 52, 813-15.
M.; Tomita, H.; Okamura, N.; Nakamura, T.; Okumura, K. Anti- [220] Aguinaldo, A.M.; Armstrong, J.A.; Cannon, J.R.; Colegate, S.M.;
proliferative activity of Rhinacanthus nasutus (L.) kurz extracts Comber, M.F.; Marchant, N.G.; Sargent, M.V. A new naphthoqui-
and the active moiety, rhinacanthin C. Biol. Pharm. Bull., 2004, 27, none from Conospermum stoechadis. Aust. J. Chem., 1996, 49,
1070-74. 197-98.
[200] Kongkathip, N.; Luangkamin, S.; Kongkathip, B.; Sangma, C.;
Grigg, R.; Kongsaeree, P.; Prabpai, S.; Pradidphol, N.; Pitaviri-
Noteworthy Secondary Metabolites Naphthoquinones Current Pharmaceutical Analysis, 2009, Vol. 5, No. 1 65

[221] Lee, S.; An, R.; Min, B.; Na, M.; Lee, C.; Kang, S.; Maeng, H.; [242] Hou, Y.; Guo, T.; Wu, C.F.; He, X.A.; Zhao, M.H. Effect of
Bae, K. A new naphthoquinone from Pyrola japonica. Archiev. shikonin on human breast cancer cells proliferation and apoptosis
Pharm. Res., 2001, 24, 522-23. in vitro. Yakugaku Zasshi-J. Pharm. Soc. Jpn., 2006, 126, 1383-86.
[222] Khambay, B.P.S.; Batty, D.; Cahill, M.; Denholm, I.; Mead-Briggs, [243] Yeh, C.C.; Kuo, H.M.; Li, T.M.; Lin, J.P.; Yu, F.S.; Lu, H.F.;
M.; Vinall, S.; Niemeyer, H.M.; Simmonds, M.S.J. Isolation, char- Chung, J.G.; Yang, J.S. Shikonin-induced apoptosis involves
acterization, and biological activity of naphthoquinones from Cal- caspase-3 activity in a human bladder cancer cell line (T24). In
ceolaria andina L. J. Agric. Food Chem., 1999, 47, 770-75. Vivo, 2007, 21, 1011-19.
[223] Garbarino, J.A.; Chamy, M.C.; Piovano, M. Chemistry of the Cal- [244] Thangapazham, R.L.; Singh, A.K.; Seth, P.; Misra, N.; Shafali;
ceolaria genus. Structural and biological aspects. Molecules, 2000, Mathad, V.T.; Raj, K.; Maheshwari, R.K. Shikonin analogue (SA)
5, 302-03. 93/637 induces apoptosis by activation of caspase-3 in U937 cells.
[224] Vasconcellos, M.C.; Montenegro, R.C.; Militao, G.C.G.; Fonseca, Front. Biosci., 2008, 13, 561-68.
A.M.; Pessoa, O.D.L.; Lemos, T.L.G.; Pessoa, C.; Moraes, M.O.; [245] Gomathinayagam, R.; Sowmyalakshmi, S.; Mardhatillah, F.;
Costa-Lotufo, L.V. Bioactivity of biflorin, a typical o- Kumar, R.; Akbarsha, M.A.; Damodaran, C. Anticancer mecha-
naphthoquinone isolated from Capraria biflora L. J. Biosci., 2005, nism of plumbagin, a natural compound, on non-small cell lung
60, 394-98. cancer cells. Anticancer Res., 2008, 28, 785-92.
[225] Santos, H.S.; Costa, S.M.O.; Pessoa, O.D.L.; Moraes, M.O.; Pes- [246] Min, R.; Tong, J.; Yang, W.J.; Duan, W.H.; Zhou, X.J.; He, J.C.;
soa, C.; Fortier, S.; Silveira, E.R.; Lemos, T.L.G. Cytotoxic naph- Zhou, J.; Chen, W.T.; Zhang, C.P. Growth inhibition and induction
thoquinones from roots of Lippia microphylla. J. Biosci., 2003, 58, of apoptosis in human oral squamous cell carcinoma Tca-8113 cell
517-20. lines by Shikonin was partly through the inactivation of NF-kappa
[226] Luo, Z.; Meksuriyen, D.; Erdelmeier, C.A.J.; Fong, H.H.S.; Cor- B pathway. Phytother. Res., 2008, 22, 407-15.
dell, G.A. Larreantin, a novel, cytotoxic naphthoquinone from Lar- [247] Chiu, S.C.; Yang, N.S. Inhibition of tumor necrosis factor-alpha
rea tridentata. J. Org. Chem., 1988, 53, 2183-85. through selective blockade of Pre-mRNA splicing by shikonin.
[227] Damon, M.; Zhang, N.Z.; Haytowitz, D.B.; Booth, S.L. Phylloqui- Mol. Pharmacol., 2007, 71, 1640-45.
none (vitamin K1) content of vegetables. J. Food Compos. Anal., [248] Thasni, K.A.; Rakesh, S.; Rojini, G.; Ratheeshkumar, T.; Srinivas,
2005, 18, 751-58. G.; Priya, S. Estrogen-dependent cell signaling and apoptosis in
[228] Harrington, D.J.; Soper, R.; Edwards, C.; Savidge, G.F.; Hodges, BRCA1-blocked BG1 ovarian cancer cells in response to plum-
S.J.; Shearer, M.J. Determination of the urinary aglycone metabo- bagin and other chemotherapeutic agents. Ann. Oncol., 2008, 19,
lites of vitamin K by HPLC with redox-mode electrochemical de- 696-705.
tection. J. Lipid Res., 2005, 46, 1053-60. [249] Sandur, S.K.; Ichikawa, H.; Sethi, G.; Ahn, K.S.; Aggarwal, B.B.
[229] Jose, S.; Gillespie, A.R. Allelopathy in black walnut (Juglans nigra Plumbagin (5-hydroxy-2-methyl-1,4-naphthoquinone) suppresses
L.) alley cropping. I. Spatio-temporal variation in soil juglone in a NF-kappa B activation and NF-kappa B-regulated gene products
black walnut-corn (Zea mays L.) alley cropping system in the through modulation of p65 and I kappa B alpha kinase activation,
midwestern USA. Plant Soil, 1998, 203, 191-97. leading to potentiation of apoptosis induced by cytokine and che-
[230] Wilis, R.J. Juglans spp, juglone and allelopathy. Allelopthy J., motherapeutic agents. J. Biol. Chem., 2006, 281, 17023-33.
2000, 7, 1-55. [250] Hsu, Y.L.; Cho, C.Y.; Kuo, P.L.; Huang, Y.T.; Lin, C.C. Plum-
[231] Picon-Cochard, C.; Nsourou-Obame, A.; Collet, C.; Guehl, J.M.; bagin (5-hydroxy-2-methyl-1,4-naphthoquinone) induces apoptosis
Eerhi, A. Competition for water between walnut seedlings (Juglans and cell cycle arrest in A549 cells through p53 accumulation via c-
regia) and rye grass (Lolium perenne) assessed by carbon isotope Jun NH2-terminal kinase-mediated phosphorylation at serine 15 in
discrimination and delta O-18 enrichment. Tree Physiol., 2001, 21, vitro and in vivo. J. Pharmacol. Exp. Ther., 2006, 318, 484-94.
183-91. [251] Wang, C.C.C.; Chiang, Y.M.; Sung, S.C.; Hsu, Y.L.; Chang, J.K.;
[232] Hejl, A.M.; Koster, K.L. Juglone disrupts root plasma membrane Kuo, P.L. Plumbagin induces cell cycle arrest and apoptosis
H+-ATPase Activity and impairs water uptake, root respiration, through reactive oxygen species/c-Jun N-terminal kinase pathways
and growth in soybean (Glycine max) and corn (Zea mays). J. in human melanoma A375.S2 cells. Cancer Lett., 2008, 259, 82-98.
Chem. Ecol., 2004, 30, 453-71. [252] Shukla, S.; Wu, C.P.; Nandigama, K.; Ambudkar, S.V. The naph-
[233] Jose, S.; Gillispie, A.R. Allelopathy in black walnut (Juglans nigra thoquinones, vitamin K3 and its structural analogue plumbagin, are
L.) alley cropping. II. Effect of juglone on hydroponically grown substrates of the multidrug resistance-linked ATP binding cassette
corn (Zea mays L.) and soybean (Glycine max L.) growt and physi- drug transporter ABCG2. Mol. Cancer Ther., 2007, 6, 3279-86.
ology. Plant Soil Environ., 1998, 203, 199-205. [253] Jones, M.E. Pyrimidine nucleotide biosynthesis in animals: genes,
[234] Hejl, A.M.; Koster, K.L. Effect of juglone on growth, photosynthe- enzymes and regulation of UMP biosynthesis. Annu. Rev. Bio-
sis, and respiration. J. Chem. Ecol., 1993, 19, 559-68. chem., 1980, 49, 253-79.
[235] Mahoney, N.; Molyneux, R.J.; Campbell, B.C. Regulation of afla- [254] Angermuller, S.; Loffler, M. Location of dihydroorotate oxidase in
toxin production by naphthoquinones of walnut (Juglans regia). J. myocardium and kidney cortex of the rat. An electron microscopic
Agric. Food Chem., 2000, 48, 4418-21. study using the cerium technique. Histochemistry, 1995, 103, 287-
[236] Krishnaswamy, M.; Purushothaman, K.K. Plumbagin: a study of 92.
anticancer, antibacterial and antifungal properties. Indian J. Exp. [255] Knecht, W.; Henseling, J.; Loffler, M. Kinetics of inhibition of
Biol., 1998, 18, 876-77. human and rat dihydroorotate dehydrogenase by atovaquone, law-
[237] Inbaraj, J.J.; Chignell, C.F. Cytotoxic action of juglone and plum- sone derivates, brequinar sodium and polyporic acid. Chem.-Biol.
bagin: A mechanistic study using HaCaT keratinocytes. Chem. Res. Interact., 2000, 124, 61-76.
Toxicol., 2004, 17, 55-62. [256] Lin, P.H.; Pan, W.C.; Kang, Y.W.; Chen, Y.L.; Lin, C.H.; Lee,
[238] Higa, M.; Ogihara, K.; Yogi, S. Bioactive naphthoquinone de- M.C.; Chou, Y.H.; Nakamura, J. Effects of naphthalene quinonoids
rivates from Diospyros maritima Blume. Chem. Pharm. Bull., on the induction of oxidative DNA damage and cytotoxicity in calf
1998, 46, 1189-93. thymus DNA and in human cultured cells. Chem. Res. Toxicol.,
[239] Chakrabarty, S.; Roy, M.; Hazra, B.; Bfilattacharya, R.K. Induction 2005, 18, 1262-70.
of apoptosis in human cancer cell lines by diospyrin, a plant- [257] Fujii, N.; Yamashita, Y.; Arima, Y.; Nagashima, M.; Nakano, H.
derived bisnaphthoquinonoid, and its synthetic derivatives. Cancer Induction of topoisomerase II-mediated DNA cleavage by the plant
Lett., 2002, 188, 85-93. naphthoquinones plumbagin and shikonin. Antimicrob. Agents
[240] Takashi, T.; Atsushi, D.; Noboru, T.; Minoru, U. CytotoxicAn- Chemother., 1992, 36, 2589-94.
tifeedant from Dionaea muscipula Ellis: A Defensive mechanism [258] Ting, C. Y.; Hsu, C. T.; Hsu, H. T.; Su, J. S.; Chen, T. Y.; Tarn, W.
of Carnivorous Plants against predators. Bull. Chem. Soc. Jpn., Y.; Kuo, Y. H.; Whang-Peng, J.; Liu, L. F.; Hwang, J. L. Iso-
2004, 77, 537-41. diospyrin as a novel human DNA topoisomerase I inhibitor. Bio-
[241] Srinivas, P.; Gopinath, G.; Banerji, A.; Dinakar, A.; Srinivas, G. chem. Pharmacol., 2003, 66, 1981-91.
Plumbagin induces reactive oxygen species, which mediate apopto- [259] Krishnan, P.; Bastow, K. F. Novel mechanisms of DNA topoi-
sis in human cervical cancer cells. Mol. Carcinog., 2004, 40, 201- somerase II inhibition by pyranonaphthoquinone derivatives -
11. Eleutherin, alpha lapachone, and beta lapachone. Biochem. Phar-
macol., 2000, 60, 1367-79.
66 Current Pharmaceutical Analysis, 2009, Vol. 5, No. 1 Babula et al.

[260] Krishnan, P.; Bastow, K.F. Novel mechanism of cellular DNA [279] Bakare, O.; Copeland, R.L.; Kanaan, Y.; Zalkow, L.H. Cytotoxic
topoisomerase II inhibition by the pyranonaphthoquinone deriva- activities of imido-substituted 2-chloro-1,4-naphthoquinone deriva-
tives alpha-lapachone and beta-lapachone. Cancer Chemother. tives on three human prostate cancer cell lines. Abstr. Papers Am.
Pharmacol., 2001, 47, 187-98. Chem. Soc., 2005, 229, U194-U95.
[261] Esteves-Souza, A.; Figueiredo, D.V.; Esteves, A.; Camara, C.A.; [280] Kim, B.H.; Yoo, J.; Park, S.H.; Jung, J.K.; Cho, H.; Chung, Y.S.
Vargas, M.D.; Pinto, A.C.; Echevarria, A. Cytotoxic and DNA- Synthesis and evaluation of antitumor activity of novel 1,4-
topoisomerase effects of lapachol amine derivatives and interac- naphthoquinone derivatives (IV). Archiev. Pharm. Res., 2006, 29,
tions with DNA. Brazilian J. Med. Biol. Res., 2007, 40, 1399-402. 123-30.
[262] Fila, C.; Metz, C.; van der Sluijs, P. Juglone inactivates cysteine- [281] Mossa, J.S.; El-Feraly, F.S.; Muhammad, I. Antimycobacterial
rich proteins required for progression through mitosis. J. Biol. constituents from Juniperus procera, Ferula communis and Plum-
Chem., 2008, 283, 21714-24. bago zeylanica and their in vitro synergistic activity with isonico-
[263] Acharya, B.R.; Bhattacharyya, B.; Chakrabarti, G. The natural tinic acid hydrazide. Phytoter. Res., 2004, 18, 934-37.
naphthoquinone plumbagin exhibits antiproliferative activity and [282] Didry, N.; Dubreuil, L.; Pinkas, M. Activity of anthraquinonic and
disrupts the microtubule network through tubulin binding. Bio- naphthoquinonic compounds on oral bacteria. Pharmazie, 1994, 49,
chemistry, 2008, 47, 7838-45. 681-83.
[264] Seguraaguilar, J.; Jonsson, K.; Tidefelt, U.; Paul, C. The cytotoxic [283] Park, B.S.; Kim, J.R.; Lee, S.E.; Kim, A.S.; Takeoka, G.R.; Ahn,
effect of 5-OH-1,4-naphthoquinone and 5,8-diOH-1,4- Y.J.; Kim, J.H. Selective growth-inhibiting effects of compounds
naphthoquinone on doxorubicin-rezistant humal leukemia cells identified in Tabebuia impetiginosa inner bark on human intestinal
(HL-50). Leuk. Res., 1992, 16, 631-37. bacteria. J. Agric. Food Chem., 2005, 53, 1152-57.
[265] Song, G.Y.; Kim, Y.; You, Y.J.; Cho, H.; Kim, S.H.; Sok, D.E.; [284] Park, B.S.; Lee, H.K.; Lee, S.E.; Piao, X.L.; Takeoka, G.R.; Wong,
Ahn, B.Z. Naphthazarin derivatives (VI): Synthesis, inhibitory ef- R.Y.; Ahn, Y.J.; Kim, J.H. Antibacterial activity of Tabebuia impe-
fect on DNA topoisomerase-I and antiproliferative activity of 2-or tiginosa Martius ex DC (Taheebo) against Helicobacter pylori. J.
6-(1-oxyiminoalkyl)-5,8-dimethoxy-1,4-naphthoquinones. Arch. Ethnopharmacol., 2006, 105, 2006.
Pharm., 2000, 333, 87-92. [285] Adeniyi, B.A.; Fong, H.H. S.; Pezzuto, J.M.; Luyengi, L.; Odelola,
[266] Bakare, O.; Ashendel, C.L.; Peng, H.R.; Zalkow, L.H.; Burgess, H. A. Antibacterial activity of diospyrin, isodiospyrin and bisiso-
E.M. Synthesis and MEK1 inhibitory activities of imido-substituted diospyrin from the root of Diospyros piscatoria (Gurke) (Ebena-
2-chloro-1,4-naphthoquinones. Bioorg. Med. Chem., 2003, 11, ceae). Phytoter. Res., 2000, 14, 112-17.
3165-70. [286] Sasaki, K.; Abe, H.; Yoshizaki, F. In vitro antifungal activity of
[267] Kumbhar, A.; Padhye, S.; Ross, D. Cytotoxic properties of iron- naphthoquinone derivatives. Biol. Pharm. Bull., 2002, 25, 669-70.
hydroxynaphthoquinone complexes in rat hepatocytes. Biometals, [287] Bogdanov, P.; Albesa, I.; Sperandeo, N.R.; Luna, C.; deBertorello,
1996, 9, 235-40. M.M. Antibacterial effect of 2-hydroxy-N-(3,4-dimethyl-5-
[268] Renou, S.G.; Asis, S.E.; Abasolo, M.I.; Bekerman, D.G.; Bruno, isoxazolyl)-1,4-naphthoquinone-4-imine on Staphylococcus aureus.
A.M. Monoarylhydrazones of alpha-lapachone: synthesis, chemical Experientia, 1996, 52, 600-04.
properties and antineoplastic activity. Pharmazie, 2003, 58, 690-95. [288] Oliveira, C.G. T.; Miranda, F.F.; Ferreira, V.F.; Freitas, C.C.; Ra-
[269] Kyong-Up, B.; Yong, S.G.; Yong, K.; Dai-Eun, S.; Byung-Zun, A. bello, R.F.; Carballido, J.M.; Correa, L.C.D. Synthesis and Antimi-
2-substituted naphthazarins; Synthesis and antitumor activity. Arch. crobial Evaluation of 3-Hydrazino-Naphthoquinones as Analogs of
Pharm., 1997, 330, 377-82. Lapachol. J. Braz. Chem. Soc., 2001, 12, 229-345.
[270] Chang, H.X.; Chou, T.C.; Savaraj, N.; Liu, L.F.; Yu, C.; Cheng, [289] Bogdanov, P.M.; Ortiz, C.S.; Eraso, A.J.; Albesa, I. Relationship
C.C. Design of antineoplastic agents based on the "2- between chemical structure and antibacterial activity in methylated
phenylnaphthalene-type" structural pattern. 4. Synthesis and bio- isoxazolylnaphthoquinones. Med. Chem. Res., 2001, 10, 577-86.
logical activity of 2-chloro-3-(substituted phenoxy)-1,4- [290] de Oliveira, C.G.T.; Ferreira, V.F.; Freitas, C.; Carballido, J.M.
naphthoquinones and related 5,8-dihydroxy-l,4-naphthoquinones. J. Synthesis and antimicrobial evaluation of oxazole-1,4-naphthqui-
Med. Chem., 1999, 42, 405-08. nones. Heterocycl. Commun., 2002, 8, 199-204.
[271] You, Y.J.; Ahn, B.Z. 6-(1-alkenoyloxyalkyl)-5,8-dimethoxy-1,4- [291] Tandon, V.K.; Yadav, D.B.; Singh, R.V.; Vaish, M.; Chaturvedi,
naphthoquinone derivatives: Synthesis and evaluation of antitumor A. K.; Shukla, P.K. Synthesis and biological evaluation of novel
activity. Archiev. Pharm. Res., 1998, 21, 738-43. 1,4-naphthoquinone derivatives as antibacterial and antiviral
[272] You, Y.J.; Zheng, X.G.; Yong, R.; Ahn, B.Z. Naphthazarin deriva- agents. Bioorg. Med. Chem. Lett., 2005, 15, 3463-66.
tives: Synthesis, cytotoxic mechanism and evaluation of antitumor [292] Tandon, V. K.; Yadav, D. B.; Singh, R. V.; Chaturvedi, A. K.;
activity. Archiev. Pharm. Res., 1998, 21, 595-98. Shukla, P. K. Synthesis and biological evaluation of novel (L)-
[273] Ravelo, A.G.; Estevez-Braun, A.; Chavez-Orellana, H.; Perez- alpha-amino acid methyl ester, heteroalkyl, and aryl substituted
Sacau, E.; Mesa-Siverio, D. Recent studies on natural products as 1,4-naphthoquinone derivatives as antifungal and antibacterial
anticancer agents. Curr. Top. Med. Chem., 2004, 4, 241-65. agents. Bioorg. Med. Chem. Lett., 2005, 15, 5324-28.
[274] Hirai, K.I.; Koyama, J.; Pan, J.H.; Simamura, E.; Shimada, H.; [293] Tandon, V.K.; Maurya, H.K.; Yadav, D.B.; Tripathi, A.; Kumar,
Yamori, T.; Sato, S.; Tagahara, K.; Tsuruo, T. Furanonaphthoqui- M.; Shukla, P. K. Naphtho[2,3-b][1,4]-thiazine-5,10-diones and 3-
none analogs possessing preferential antitumor activity compared substituted-1,4-dioxo-1,4-dihydronaphthalen-2-yl-thioalkanoate de-
to normal cells. Cancer Detect. Prev., 1999, 23, 539-50. rivatives: Synthesis and biological evaluation as potential antibac-
[275] Tanada, Y.; Mori, K. Synthetic microbial chemistry, XXXVII. terial and antifungal agents. Bioorg. Med. Chem. Lett., 2006, 16,
Synthesis and absolute configuration of Nocardione A and B, 5883-87.
furano-o-naphthoquinone-type metabolites of nocardia sp with anti- [294] Kapadia, G.J.; Balasubramanian, V.; Tokuda, H.; Konoshima, T.;
fungal, cytotoxic, and enzyme inhibitory activities. Eur. J. Org. Takasaki, M.; Koyama, J.; Tagahaya, K.; Nishino, H. Anti-tumor
Chem., 2001, 22, 4313-19. promoting effects of naphthoquinone derivatives on short term Ep-
[276] You, Y.J.; Kim, Y.; Song, G.Y.; Ahn, B.Z. (E)-6-(1- stein-Barr early antigen activation assay and in mouse skin car-
alkyloxyiminoalkyl)-5,8-dimethoxy-1,4-naphthoquinones: Synthe- cinogenesis. Cancer Lett., 1997, 113, 47-53.
sis, cytotoxic activity and antitumor activity. Bioorg. Med. Chem. [295] Koyama, J.; Morita, I.; Kobayashi, N.; Osakai, T.; Hotta, H.; Taka-
Lett., 2000, 10, 2301-03. yasu, J.; Nishino, H.; Tokuda, H. Correlation of redox potentials
[277] Chung, Y. S.; Shin, Y. K.; Zhan, C. G.; Lee, S.; Cho, H. Synthesis and inhibitory effects on Epstein-Baff virus activation of naphtho-
and evaluation of antitumor activity of 2-and 6-[(1,3-benzothiazol- quinones. Cancer Lett., 2003, 201, 25-30.
2-yl)aminomethyl]-5,8-dimethoxy-1, 4-naphthoquinone deriva- [296] Stagliano, K.W.; Emadi, A.; Lu, Z.H.; Malinakova, H.C.; Twenter,
tives. Archiev. Pharm. Res., 2004, 27, 893-900. B.; Yu, M.; Holland, L.E.; Rom, A.M.; Harwood, J.S.; Amin, R.;
[278] Afrasiabi, Z.; Sinn, E.; Chen, J.N.; Ma, Y.F.; Rheingold, A.L.; Johnson, A.A.; Pommier, Y. Regiocontrolled synthesis and HIV
Zakharov, L.N.; Rath, N.; Padhye, S. Appended 1,2-naphthoqui- inhibitory activity of unsymmetrical binaphthoquinone and trimeric
nones as anticancer agents 1: synthesis, structural, spectral and an- naphthoquinone derivatives of conocurvone. Bioorg. Med. Chem.,
titumor activities of ortho-naphthaquinone thiosemicarbazone and 2006, 14, 5651-5665.
its transition metal complexes. Inorg. Chim. Acta, 2004, 357, 271- [297] Singh, S.B.; Jayasuriya, H.; Dewey, R.; Polishook, J.D.; Dom-
78. browski, A.W.; Zink, D.L.; Guan, Z.Q.; Collado, J.G.P.; Pelaez, F.;
Felock, P.J.; Hazuda, D.J. Isolation, structure, and HIV-1-integrase
Noteworthy Secondary Metabolites Naphthoquinones Current Pharmaceutical Analysis, 2009, Vol. 5, No. 1 67

inhibitory activity of structurally diverse fungal metabolites. J. Ind. [318] Grevenstuk, T.; Goncalves, S.; Nogueira, J.M.F.; Romano, A.
Microbiol. Biotechnol., 2003, 30, 721-31. Plumbagin recovery from field specimens of Drosophyllum lusi-
[298] Min, B.S.; Miyashiro, H.; Hattori, M. Inhibitory effects of quinones tanicum (L.) link. Phytochem. Anal., 2008, 19, 229-35.
on RNase H activity associated with HIV-1 reverse transcriptase. [319] Lin, L.C.; Yang, L.L.; Chou, C.J. Cytotoxic naphthoquinones and
Phytoter. Res., 2002, 16, S57-S62. plumbagic acid glucosides from Plumbago zeylanica. Phytochemis-
[299] Yardley, V.; Snowdon, D.; Croft, S.; Hazra, B. In vitro activity of try, 2003, 62, 619-22.
diospyrin and derivatives against Leishmania donovani, Trypano- [320] Rodrigues, S.V.; Viana, L.M.; Baumann, W. UV/VIS spectra and
soma cruzi and Trypanosoma brucei brucei. Phytoter. Res., 1996, solubility of some naphthoquinones, and the extraction behavior of
10, 559-62. plumbagin from Plumbago scandens roots in supercritical CO2.
[300] Ray, S.; Hazra, B.; Mittra, B.; Das, A.; Majumder, H.K. Diospyrin, Anal. Bioanal. Chem., 2006, 385, 895-900.
a bisnaphthoquinone: A novel inhibitor of type DNA topoi- [321] Sidhu, G.S.; Sankaram, A.V.B. New biplumbagin and 3-
somerase of Leishmania donovani. Mol. Pharmacol., 1998, 54, chloroplumbagin from Plumbago zeylanica. Tetrahedron Lett.,
994-99. 1971, 26, 2385-88.
[301] Danoun, S.; Baziard-Mouysset, G.; Stigliani, J.L.; Ane-Margail, [322] Sagratini, G.; Cristalli, G.; Giardina, D.; Gioventu, G.; Maggi, F.;
M.; Payard, M.; Leger, J.M.; Canron, X.; Vial, H.; Loiseau, P.M.; Ricciutelli, M.; Vittori, S. Alkannin/shikonin mixture from roots of
Bories, C.; Recoche, C. Synthesis and protozoocidal activity of Onosma echioides (L.) L.: Extraction method study and quantifica-
new 1,4-naphthoquinones. Heterocycl. Commun., 1999, 5, 343-48. tion. J. Sep. Sci., 2008, 31, 945-52.
[302] Kayser, O.; Kiderlen, A.F.; Laatsch, H.; Croft, L.S. In vitro [323] Kapadia, N.S.; Isarani, S.A.; Shah, M.B. A simple method for
leishmanicidal activity of monomeric and dimeric naphthoqui- isolation of plumbagin from roots of Plumbago rosea. Pharm. Biol.,
nones. Acta Trop., 2000, 77, 307-14. 2005, 43, 551-53.
[303] Hazra, B.; Sarkar, R.; Bhattacharyya, S.; Ghosh, P.K.; Chel, G.; [324] Hsieh, Y.J.; Lin, L.C.; Tsai, T.H. Measurement and pharmacoki-
Dinda, B. Synthesis of plumbagin derivates and their inhibitory Ac- netic study of plumbagin in a conscious freely moving rat using
tivities against ehrlich ascites carcinoma in vivo and Leishmania liquid chromatography/tandem mass spectrometry. J. Chromatogr.
donovani Promastigotes in vitro. Phytoter. Res., 2002, 16, 133-37. B, 2006, 844, 1-5.
[304] Teixiera, M.J.; de Almeida, Y.M.; Viana, J.R.; Holanda, J.G.; Rod- [325] Gilgenast, E.; Zuk, L.; Skrzypczak, A.; Krolicka, A.; Lojkowska,
rigues, T.P.; Prata, J.R.C.; Coelho, I.V.B.; Rao, V.S.; Pompeu, E.; Kaminski, M. Optimal conditions of ramentaceone and plum-
M.M. L. In vitro and in vivo leishmanicidal activity of 2-hydroxy- bagin separation and isolation from carnivorous plants extracts us-
3-(3-methyl-2-butenyl)-1,4-naphthoquinone (lapachol). Phytoter. ing normal phase-high performance liquid chromatography (NP-
Res., 2001, 15, 44-48. HPLC). Planta Med., 2008, 74, 1085-85.
[305] Salmon-Chemin, L.; Buisine, E.; Yardley, V.; Kohler, S.; Debreu, [326] Wang, Y.C.; Huang, T.L. High-performance liquid chromatogra-
M.A.; Landry, V.; Sergheraert, C.; Croft, S.L.; Krauth-Siegel, R.L.; phy for quantification of plumbagin, an anti-Helicobacter pylori
Davioud-Charvet, E. 2-and 3-substituted 1,4-naphthoquinone de- compound of Plumbago zeylanica L. J. Chromatogr. A, 2005,
rivatives as subversive substrates of trypanothione reductase and 1094, 99-104.
lipoamide dehydrogenase from Trypanosoma cruzi: Synthesis and [327] Nickel, S.L.; Carroll, T.F. Reversed-phase ion-pair high-per-
correlation between redox cycling activities and in vitro cytotoxic- formance liquid-chromatography of naphthazarins. J. Chromatogr.,
ity. J. Med. Chem., 2001, 44, 548-68. 1984, 295, 521-25.
[306] Oku, H.; Ishiguro, K. Cyclooxygenase-2 Inhibitory 1,4-Naphtho- [328] Assimopoulou, A.N.; Ganzera, M.; Stuppner, H.; Papageorgiou, V.
quinones from Impatiens balsamina L. Biol. Pharm. Bull., 2002, P. Simultaneous determination of monomeric and oligomeric al-
25, 658-60. kannins and shikonins by high-performance liquid chromatogra-
[307] Itoigawa, M.; Takeya, K.; Furukawa, H. Cardiotonic action of phy-diode array detection-mass spectrometry. Biomed. Chroma-
plumbagin on guinea-pig papillary muscle. Planta Med., 1991, 57, togr., 2008, 22, 173-90.
317-19. [329] Assimopoulou, A.N.; Karapanagiotis, I.; Vasiliou, A.; Kokkini, S.;
[308] Floreani, M.; Forlin, A.; Pandolfo, L.; Petrone, M.; Bellin, S. Papageorgiou, V.P. Analysis of alkannin derivatives from Alkanna
Mechanism of plumbagin action on guinea pig isolated atria. J. species by high-performance liquid chromatography/photodiode ar-
Pharmacol. Exp. Ther., 1996, 278, 763-70. ray/mass spectrometry. Biomed. Chromatogr., 2006, 20, 1359-74.
[309] Shen, Z.Q.; Dong, Z.J.; Cheng, P.; Li, L.; Chen, Z.H.; Liu, J. Ef- [330] Ikeda, Y.; Ishida, N.; Fukaya, C.; Yokoyama, K.; Tabata, M.; Fu-
fects of plumbagin on platelet aggregation and platelet-neutrophil kuji, H.; Honda, G. Determination of the ratio between optical iso-
interactions. Planta Med., 2003, 69, 605-09. mers, shikonin and alkannin by high-performance liquid-
[310] Vijayakumar, R.; Senthilvelan, M.; Ravindran, R.; Devi, R.S. chromatography analysis. Chem. Pharm. Bull., 1991, 39, 2351-52.
Plumbago zeylanica action on blood coagulation profile with and [331] Steinert, J.; Khalaf, H.; Rimpler, M. High-performance liquid
without blood volume reduction. Vasc. Pharmacol., 2006, 45, 86- chromatographic separation of some naturally occuring naphtho-
90. quinones and anthraquinones. J. Chromatogr. A, 1995, 723, 206-
[311] Sharma, I.; Gusain, D.; Dixit, V.P. Hypolipidaemic and antiathero- 09.
sclerosis effects of plumbagin in rabbits. Ind. J. Physiol. Pharma- [332] Awang, D.V.C.; Kindack, D.; Dawson, B.A. Tandem high-
col., 1991, 35, 10-14. performance liquid chromatography methods for resolution of la-
[312] An, S.; Park, Y.D.; Paik, Y.K.; Jeong, T.S.; Lee, W.S. Human lachol and related naphthoquinones. J. Chromatogr., 1986, 368,
ACAT inhibitory effects of shikonin derivatives from Lithosper- 439-43.
mum erythrorhizon. Bioorg. Med. Chem. Lett., 2007, 17, 1112-16. [333] Silva, R.S.F.; Leitao, G.G.; Brum, T.B.; Lobato, A.P.G.; Pinto, M.;
[313] Cavaleante, F.A.; Silva, J.L.; Carvalho, V.M.N.; Camara, C.A.; Pinto, A.V. Applications of counter-current chromatography in or-
Silva, T.M.S.; Pinto, A.C.; Vargas, M.D.; Silva, B.A. Spasmolytic ganic synthesis purification of heterocyclic derivatives of lapachol.
activity of lapachol and its derivatives, alpha and beta-lapachone, J. Chromatogr. A, 2007, 1151, 197-202.
on the guinea-pig ileum involves blockade of voltage-gated cal- [334] Marston, A.; Hoetettmann, K. High-performance liquid-chromato-
cium. Rev. Bras. Farmacogn.-Braz. J. Pharmacogn., 2008, 18, graphy of some naturally-occuring naphthoquinones. J. Chroma-
183-89. togr., 1984, 295, 526-29.
[314] Chowdhury, A.K.; Sushanta, K.C.; Azad Khan, A.K. Antifertility [335] Gupta, M.M.; Verma, R.K.; Uniyal, G.C.; Jain, S.P. Determination
activity of Plumbago zeylanica Linn. root. Ind. J. Med. Res., 1982, of plumbagin by normal-phase high-performance liquid-
76, 99-101. chromatography. J. Chromatogr., 1993, 637, 209-12.
[315] Bhargava, S.K. Effects of plumbagin on reproductive function of [336] Babula, P.; Mikelova, R.; Adam, V.; Kizek, R.; Havel, L.; Sladky,
male dog. Ind. J. Exp. Biol., 1984, 22, 153-56. Z. Using of liquid chromatography coupled with diode array detec-
[316] Bhargava, S.K. Effect of testoterone repacement therapy on quanti- tor for determination of naphthoquinones in plants and for investi-
tative spermatogenesis following plumbagin treatment in immature gation of influence of pH of cultivation medium on content of
rats. Acta Eur. Fertil., 1986, 17, 217-19. plumbagin in Dionaea muscipula. J. Chromatogr. B, 2006, 842,
[317] Santhakumari, G.; Suganthan, D. Antigonadotropic activity of 28-35.
plumbagin. Planta Med., 1980, 39, 244. [337] Meyer, J.J. M.; Van der Kooy, F.; Joubert, A. Identification of
plumbagin epoxide as a germination inhibitory compound through
a rapid bioassay on TLC. S. Afr. J. Bot., 2007, 73, 654-56.
68 Current Pharmaceutical Analysis, 2009, Vol. 5, No. 1 Babula et al.

[338] Ei-Shaer, N.S.; Badr, J.M.; Aboul-Ela, M.A.; Gohar, Y.M. Deter- [350] Adam, V.; Petrlova, J.; Potesil, D.; Zehnalek, J.; Sures, B.;
mination of lawsone in henna powders by high performance thin Trnkova, L.; Jelen, F.; Kizek, R. Study of metallothionein modified
layer chromatography. J. Sep. Sci., 2007, 30, 3311-15. electrode surface behaviour in the presence of heavy metal ions-
[339] Zalis, S.; Fiedler, J.; Pospisil, L.; Fanelli, N.; Lanza, C.; Lampug- biosensor. Electroanalysis, 2005, 17, 1649-57.
nani, L. Electron transfer in donor-acceptor molecules of substitued [351] Adam, V.; Zehnalek, J.; Petrlova, J.; Potesil, D.; Sures, B.;
naphthoquinones: spectral and redox properties of internal charge Trnkova, L.; Jelen, F.; Vitecek, J.; Kizek, R. Phytochelatin modi-
transfer complexes. Microchem. J., 1996, 54, 478-86. fied electrode surface as a sensitive heavy metal ion biosensor.
[340] Raoof, J.B.; Golabi, S.M. Electrochemical behaviour of some 1,4- Sensors, 2005, 5, 70-84.
naphthoquinone derivates at the surface of glassy carbon electrode [352] Babula, P.; Huska, D.; Hanustiak, P.; Baloun, J.; Krizkova, S.;
in acetonitrile. Iran. J. Sc. Technol., 1998, 22, 123-34. Adam, V.; Havel, L.; Zemlicka, M.; Horna, A.; Beklova, M.;
[341] Bodini, M.E.; Arancibia, V. Redox chemistry of 1,2-dihydroxy- Kizek, R. Flow Injection Analysis Coupled with Carbon Electrodes
naphthalene, 1,2-naphthosemiquinone and 1,2-naphthoquinone and as the Tool for Analysis of Napthoquinones with Respect to Their
their complexes with manganese(II) and manganese (III). Transit. Content and Functions in Biological Samples. Sensors, 2006, 6,
Met. Chem., 1997, 22, 150-55. 1466-82.
[342] Camus, J.; Meghea, A.; Anacona, J.R. Spectroscopic and electro- [353] Klejdus, B.; Vacek, J.; Adam, V.; Zehnalek, J.; Kizek, R.; Trnkova,
chemical studies of Ni-II, Cu-II, Pd-II and VOII complexes with L.; Kuban, V. Determination of isoflavones in soybean food and
naphthoquinonic ligands. Polyhedron, 1997, 16, 933-38. human urine using liquid chromatography with electrochemical de-
[343] Turovska, B.; Stradins, J.; Logins, J.; Strazdins, I.; Dregeris, J. tection. J. Chromatogr. B, 2004, 806, 101-11.
Electrochemical study of intramolecular charge-transfer complexes [354] Mikelova, R.; Hodek, P.; Hanustiak, P.; Adam, V.; Krizkova, S.;
derived from 1,4-naphthoquinone. 1. Electroreduction. J. Electro- Havel, L.; Stiborova, M.; Horna, A.; Beklova, M.; Trnkova, L.;
anal. Chem., 1995, 394, 229-38. Kizek, R. Determination of isoflavones using liquid chromatogra-
[344] Raoof, J.B.; Golabi, S.M. Electrochemical properties of carbon- phy with electrochemical detection. Acta Chim. Slov., 2007, 54, 92-
paste electrodes spiked with some 1,4-naphthoquinone derivates. 97.
Bull. Chem. Soc. Jpn., 1995, 68, 2253-61. [355] Koyama, J.; Morita, I.; Kino, A.; Tagahara, K. Micellar electroki-
[345] Glezer, V.; Stradins, J.; Freimanis, J.; Baider, L. The mechanism of netic chromatography (MEKC) separation of furanonaphthoqui-
electrochemical reduction of intramolecular charge-thansfer com- nones from Tabebuia impetiginosa. Chem. Pharm. Bull., 2000, 48,
plexes derived from 1,4-naphthoquinone. Electrochim. Acta, 1983, 873-75.
28, 87-95. [356] Abdollahi, H.; Bagheri, L. Simultaneous spectrophotometric de-
[346] Patriarche, G.H.; Lingane, J.J. Electrochemical characteristics of 2- termination of vitamin K-3 and 1,4-naphthoquinone after cloud
methyl-1,4-naphthoquinone (vitamin K3) : A coulometric micro- point extraction by using genetic algorithm based wavelength se-
method of determination. Anal. Chim. Acta, 1970, 49, 241-46. lection-partial least squares regression. Anal. Chim. Acta, 2004,
[347] Alonso, L.; Palmero, S.; Munoz, E.; Sanllorente, S.; Garcia-Garcia, 514, 211-18.
M. A. Electrochemical behaviour of menadione on glassy carbon [357] Sakamoto, S.; Putalun, W.; Tsuchihashi, R.; Morimoto, S.; Kinjo,
rotating disk electrode (RDE). Electroanalysis, 2000, 12, 757-62. J.; Tanaka, H. Development of an enzyme-linked immunosorbent
[348] Raoof, J.B.; Golabi, S.M. Electrochemical behaviour of Plumbagin assay (ELISA) using highly-specific monoclonal antibodies against
at solid electrodes in non-aqueous media. Bull. Electrochem., 1997, plumbagin. Anal. Chim. Acta, 2008, 607, 100-05.
13, 311-16. [358] Ardakani, M.M.; Karami, P.E.; Zare, H.R.; Hamzehloo, M. Elec-
[349] Wu, H.Q.; Huang, Z.S.; Bu, X.Z.; Shen, Y.D.; Zhang, Z.L.; Xie, trocatalytic reduction of oxygen on the surface of glassy carbon
B.F.; Liu, Z.C.; Gu, L.Q.; Chan, A.S.C. The molecular mechanisms electrodes modified with plumbagin. Microchim. Acta, 2007, 159,
involved in the cytotoxicity of alkannin derivatives. Eur. J. Med. 165-73.
Chem., 2005, 40, 1341-45. [359] Long, G.L.; Winefordner, J.D. Limit of Detection. Anal. Chem.,
1983, 55, A712-A724.

Received: 23 April, 2008 Revised: 25 August, 2008 Accepted: 14 October, 2008

Potrebbero piacerti anche