You are on page 1of 13

ARTICLE IN PRESS

Phytomedicine 12 (2005) 305–317


www.elsevier.de/phymed

Bacopa monniera, a reputed nootropic plant: an overview


A. Russoa,, F. Borrellib
a
Department of Biological Chemistry, Medical Chemistry and Molecular Biology, University of Catania, Catania, Italy
b
Department of Experimental Pharmacology, University of Naples ‘Federico II’, Naples, Italy

Received 22 October 2003; accepted 15 December 2003

Abstract
Bacopa monniera (BM), a traditional Ayurvedic medicine, used for centuries as a memory enhancing, anti-
inflammatory, analgesic, antipyretic, sedative and antiepileptic agent. The plant, plant extract and isolated bacosides
(the major active principles) have been extensively investigated in several laboratories for their neuropharmacological
effects and a number of reports are available confirming their nootropic action. In addition, researchers have evaluated
the anti-inflammatory, cardiotonic and other pharmacological effects of BM preparations/extracts. Therefore, in view
of the important activities performed by this plant, investigation must be continued in the recently observed actions
described in this paper. Moreover, other clinical studies have to be encouraged, also to evidence any side effects and
possible interactions between this herbal medicine and synthetic drugs.
r 2004 Elsevier GmbH. All rights reserved.

Keywords: Bacopa monniera; Ayurvedic; Saponins; Nootropic action; Antioxidant activity

Introduction act as nootropic agents. One plant that has been used as
brain tonic and restorative in debilitated conditions is
Cerebral abilities have been observed to diminish Bacopa monniera (BM). BM, family Scrophulariaceae, is
significantly with advancing age and factors such as a creeping annual plant found throughout the Indian
emotional stress could precipitate these effects. subcontinent in wet, damp and marshy areas (Chune-
While pharmaceutical companies continue to invest kar, 1960; Satyavati et al., 1976). This medicinal plant is
enormous resources in identifying agents that could be locally known as Brahmi. The name Brahmi is derived
used to alleviate debilitating disorders and retard mental from the word ‘‘Brama’’, the mythical ‘‘creator’’ in the
deterioration afflicting numerous people around the Hindu pantheon. Because the brain is the centre for
world, a source of potentially beneficial agents, namely creative activity, any compound that improves the brain
phytochemicals, would appear to have significant health is called Brahmi.
benefits that have yet to be fully exploited. Therefore, BM has been used by Ayurvedic medical practitioners
several plants have been selected based on their use in in India for almost 3000 years and is classified as a
traditional systems of medicine, and research has medhyarasayana, a drug used to improve memory and
identified a number of natural compounds that could intellect (medhya). The earliest chronicled mention of
BM is in several ancient Ayurvedic treatises including
Corresponding author. Tel.: +39 095 7384073; the Caraka Samhita (6th century A.D.), in which it is
fax: +39 095 7384220. recommended in formulations for the management of a
E-mail address: alrusso@unict.it (A. Russo). range of mental conditions including anxiety, poor

0944-7113/$ - see front matter r 2004 Elsevier GmbH. All rights reserved.
doi:10.1016/j.phymed.2003.12.008
ARTICLE IN PRESS
306 A. Russo, F. Borrelli / Phytomedicine 12 (2005) 305–317

cognition and lack of concentration, and the Bravpra- Chemical constituents


kash Var-Prakarana (16th century A.D.). Brahmi is
currently recognized as being effective in the treatment In view of the importance of this plant in the
of mental illness and epilepsy. After clinical trials in indigenous system of medicine, systematic chemical
human volunteers, a chemically standardized extract of examinations of the plant have been carried out by
BM has now been made available for clinical use by the several groups of researchers. Detailed investigations
central Drug Research Institute in India (Dhawan and were first documented in 1931, when Bose and Bose
Singh, 1996). reported the isolation of the alkaloid ‘‘brahmine’’ from
In certain parts of India, Brahmi is believed to be an BM. Later, other alkaloids like nicotine and herpestine
aphrodisiac; in Sri Lanka, under the name of Loonoo- have also been reported (Chopra et al., 1956). The
weella, Brahmi is prescribed for fevers; in the Philip- isolation of D-mannitol, and a saponin, hersaponin and
pines, it is used as a diuretic (Uphof, 1968). potassium salts by Sastri et al. (1959) provided further
details of the chemical components of BM.
The major chemical entity shown to be responsible for
Botanical aspects the memory-facilitating action of BM, bacoside A, was
assigned as 3-(a-L-arabinopyranosyl)-O-b-D-glucopyra-
Family: Scrophulariaceae noside-10, 20-dihydroxy-16-keto-dammar-24-ene (Chat-
The genus Bacopa includes over 100 species of aquatic terji et al., 1965). Bacoside A usually co-occurs with
herbs distributed throughout the warmer regions of the bacoside B, the latter differing only in optical rotation
world. In the United States, the herbs are recognized as and probably an artefact produced during the process of
weeds in rice fields and found growing abundantly in isolating bacoside A (Rastogi, 1990). The chemical
marshes and wetlands of warmer regions (Barrett and composition of bacosides, contained in the polar fraction,
Strother, 1978). has been established on the basis of chemical and physical
BM is a small herb with purple flowers. It grows in degradation studies. On acid hydrolysis, bacosides
wet and sandy areas and near streams in tropical yield a mixture of aglycones, bacogenin A1, A2, A3
regions. It is a creeping herb with numerous branches (Kulshreshtha and Rastogi, 1973, 1974; Chandel et al.,
and small fleshy, oblong leaves. Flowers and fruits 1977), which are artefacts, and two genuine sapogenins,
appear in summer. The stem and the leaves of the plant jujubogenin and pseudojujubogenin (Rastogi et al.,
are used (Mathew, 1984). 1994). Another bacogenin, A4 was identified as ebelin
lactone pseudojujubogenin (Rastogi et al., 1994). Succes-
Salient botanical features sively, a minor saponin bacoside A1 was isolated and
characterized as 3-O-[a-L-arabinofuranoyl(1-3)-b-L-ara-
Stem: prostrate, (sub)succulent, herbaceous; leaves: binopyranosyl]jujubogenin (Rastogi et al., 1994). Rastogi
decussate, simple, oblong, 1  0.4 cm, succulent, punc- et al. (1994) evidenced a new triperpenoid saponin,
tuate, penninerved, margin entire, apex obtuse, sessile; bacoside A3. Its structure was established as 3-b-[O-b-D-
flower: axillar, solitary, bracteate, linear, pedicel to glucopyranosyl(1-3)-O-[a-L-arabinofuranosy(1-2)]-O-
0.5 cm, purple in colour; calyx: 5 lobes (unequal); outer 2 b-D-glucopyranosyl)oxy] jujubogenin by chemical and
lobes larger, oval, 7  3.5 mm; inner 2 lobes linear, spectral analyses. Garay et al. (1996a) isolated from
5.5  0.7 mm; median 1 lobe oblong, 5.5  2 mm, im- BM three new dammarane-type triterpenoid saponins
bricate, (sub)succulent, punctuate, obtuse, acute; cor- of biological interest, bacopasaponins A, B and C,
olla: white with violet and green bands inside the throat, identified as 3-O-a-L-arabinopyranosyl-20-O-a-L-arabino-
0.8 cm across, 5 mm tube; 5 lobes, obscurely 2-lipped, pyranosyl-jujubogenin, 3-O-[a-L-arabinofuranosyl (I-2)
2+3, (sub)equal, obtuse or emarginated; stamens: 4, a-L-arabinopyranosyl]pseudojujubogenin and 3-O-[b-
D-glucopyranosyl(1-3){a- L-arabinofuranosyl(1-2)}a-L-
didynamous; filament pairs 1 and 2.5 mm anthers
oblong, contiguous, 1.5 mm; ovary: oblong-globose, arabinopyranosyl]pseudojujubogenin by spectroscopic
2 mm; style slightly deflexed, 5.5 mm; stigma flat capsule, and chemical transformation methods. The hitherto
oblong-globose, 5  2.5 cm septicidal or locilicidal or 4 undetermined configurations at C-20 and C-22 of
valved. pseudojujubogenin were elucidated by phase-sensitive
ROESY, and 1H and 13C signals of the saponins were
assigned by DEPT, 1H–1H COSY, HSQC and HMBC
Synonyms for BM wettst.
techniques. Successively, the same authors (Garay et al.,
(1) B. monniera (L.) Pennell yes 1996b) isolated a new dammarane-type pseudojujubo-
(2) Lysimachia monnieri L. Cent. genin glycoside, bacopasaponin D, defined as 3-O-[a-L-
(3) Graticola monnieri L. arabinofuranosyl(I-2)b-D-glucopiranosyl]pseudojuju-
(4) Monniera cunefolia Michaux bogenin by spectroscopic and chemical transformation
(5) Herpestis monniera L. Kunth. methods. It is noteworthy that dammarane-type triter-
ARTICLE IN PRESS
A. Russo, F. Borrelli / Phytomedicine 12 (2005) 305–317 307

penoid saponins are major constituents of a number of their neuropharmacological effects and a number of
reputed herbal drugs, including ginseng. Although reports are available confirming their nootropic action.
jujubogenin glycosides have been isolated from several Preliminary studies established that the treatment
reputed medicinal plants (i.e. Rhamnaceae and Scro- with the plant (Malhotra and Das, 1959) and with the
phulariaceae), pseudojubogenin glycosides have only alcoholic extract of BM plant (Singh and Dhawan,
been reported in this Indian herbal drug. 1982) enhanced learning ability in rats. Subsequent
In view of the increasing interest on this herbal drug, studies indicated that the cognition-facilitating effect
Chakravarty et al. (2001) undertook a thorough chemical was due to two active saponins, bacosides A and B,
reinvestigation of the glicosidic fraction of the methanol present in the ethanol extract (Singh and Dhawan,
extract of the plant and were able to isolate two new 1992). These active principles, apart from facilitating
pseudojujubogenin glycosides designated as bacopaside I learning and memory in normal rats, inhibited the
and II. Their structures have been elucidated as 3-O-a-L- amnesic effects of scopolamine, electroshock and
arabinofuranosyl-(1-2)-[6-O-sulphonyl-b-D-glucopyrano- immobilization stress (Dhawan and Singh, 1996). The
sil-(1-3)]-a-L-arabinopyranosyl pseudojujubogenin and mechanism of these pharmacological actions remains
3-O-a-L-arabinofuranosyl-(1-2)-[b-D-glucopyranosil-(1- conjectural. It has been suggested that the bacosides
3)]b-D-glucopyranosyl pseudojujubogenin, mainly on the induce membrane dephosphorylation, with a concomi-
basis of dimensional (2D) NMR and other spectral tant increase in protein and RNA turnover in specific
analyses. Subsequently, the same authors (Chakravarty brain areas (Singh et al., 1990). Further, BM has been
et al., 2003) isolated three new saponins from BM, shown to enhance protein kinase activity in the
designated as bacopasides III, IV and V, with structures hippocampus which could also contribute to its noo-
3-O-a-L-arabinofuranosyl-(1-2)-b-D-glucopyranosyl juju- tropic action (Singh and Dhawan, 1997). A study of
bogenin, 3-O-b-D-glucopyranosyl-(1-3)-a-L-arabinopyra- Bhattacharya et al. (1999) reported that a standardized
nosyl jujubogenin, 3-O-b-D-glucopyranosyl-(1-3)-a-L- bacoside-rich extract of BM, administered for 2 weeks in
arabinofuranosyl pseudojujubogenin, on the basis of rats, reversed cognitive deficits induced by intracereb-
dimensional (3D) NMR and other spectral analyses. roventricularly administered colchicines and by injection
In addition, Hou et al. (2002) have isolated a new of ibotenic acid into the nucleus basalis magnocellularis.
saponin, 3-O-[a-1-arabinofuranosyl-(1-2)]-a-L-arabino- The central cholinergic system is considered the most
pyranosyl jujubogenin, named bacopasaponin G, a new important neurotransmitter involved in the regulation of
matsutaka alcohol derivative, (3R)-1-octan-3yl-(6-O-sulfo- cognitive functions. Cholinergic neuronal loss in hippo-
nyl)-b-D-glucopyranoside, a new phenylethanoid glycoside, campal area is the major feature of Alzheimer’s disease
3,4-dihydroxyphenylethyl alcohol (2-O-feruloyl)-b-D-glu- (AD) and enhancement of central cholinergic activity by
copyranoside, and a new glycoside, phenylethyl alcohol [5- anticholinesterase is presently the mainstay of the
O-p-hydroxybenzoyl-b-D-apiofuranosyl-(1-2)]-b-D-gluco- pharmacotherapy of AD-type senile dementia. Admin-
pyranoside. istration of BM for two weeks, also reversed the
Moreover, three new phenylethnoid glycosides, viz. depletion of acetylcholine, the reduction in choline
monnierasides I–III along with the known analogue acetylase activity and the decrease in muscarinic
plantainoside B have been isolated from the glycosidic cholinergic receptor binding in the frontal cortex
fraction of BM (Chakravarty et al., 2002). and hippocampus, induced by neurotoxin, colchicines
In Fig. 1, the chemical structure of some saponins (Bhattacharya et al., 1999).
isolated from BM are reported. It has been suggested that the behavioural effects of
cholinergic degeneration can be alleviated by a reduc-
tion in noradrenergic function (Sara, 1989). BM is
Biological activity known to lower norepinephrine and increase 5-hydro-
xytryptamine levels in the hippocampus, hypothalamus
The biological effects of BM are documented in and cerebral cortex (Singh and Dhawan, 1997). BM may
traditional as well as scientific literature. The most thus, also indirectly, modify Ach concentrations,
important of these are the effects of the plant, plant through its influence on other neurotransmitter systems.
extracts and isolated bacosides on cognition and In a recent study, standardized extracts of BM and
memory functions, their anxiolytic effects and their role Ginkgo biloba (GB) were used to evaluate the anti-
in management of convulsive disorders. dementic and anticholinesterase activities in adult male
Swiss mice (Das et al., 2002). Antidementic activity was
Neuropharmacological activity tested against scopolamine (3 mg/kg ip)-induced deficits
in passive avoidance (PA) test. Three different extracts
Beneficial effects on learning and memory of BM (30 mg/kg) and of GB (15, 30 and 60 mg/kg) were
The plant, plant extracts and isolated bacosides have administered daily for 7 days and 60 min after the last
been extensively investigated in several laboratories for dose of scopolamine, i.e. transfer latency time (TLT)
ARTICLE IN PRESS
308 A. Russo, F. Borrelli / Phytomedicine 12 (2005) 305–317

OH

OH OH
OH
O OH
O O O

O O
O
OH OH
O O
O O
O CH OH
OH O
OH O
O OH
O O O
O O
OH OH
OH OH
OH
CH OH CH OH
OH OH
OH

Bacosaponin A Bacosaponin B Bacosaponin C

OH
OH OH

O
O O

O
O
O
OH
O OH O
OH O O
O
HO SO O O OH O
O HO O
O
OH O
O OH
O OH OH
OH
OH OH O
OH O OH
OH OH OH
OH
OH OH
OH
OH

Bacopaside I Bacopaside II Bacopaside IV

OH
OH OH

O O
O

O O O
CH OH
OH
O O
O O O
OH
O OH O
OH
OH
O OH
OH O
OH O
OH OH O
HOH C O
OH
OH
OH HO

OH

Bacopaside V Bacopasaponin D Bacosaponin G


Fig. 1. Chemical structure of some saponins isolated from BM.

and no transfer response (NTR) were taken as criteria that, unlike the amnesic action of benzodiazepine
for learning. BM and GB treatments produced sig- anxiolytics, BM promotes cognition. One study, using
nificant increases in TLT (Fig. 2) and NTR (Fig. 3), thus experimentally validated rat models of clinical anxiety,
attenuating the dementic effect. All the extracts showed compared to the anxiolytic effect of standardized BM
a dose-dependent inhibitory effect on acetylcholinester- extract with that of benzodiazepine. The effects of BM
ase/AchE activity in vitro, but none of BM extracts extract at levels of 5, 10, and 20 mg/kg administered
showed more than 50% inhibition, suggesting that the orally to rats were compared to those elicited by
extracts of GB and BM have potent cognitive enhancing lorazepam (LZP) (0.5 mg/kg administered intraperito-
properties, but with different mechanisms of action. neally). The higher doses of BM extract produced
significantly greater anxiolytic effects compared to
Anxiolytic effect LZP, a standard benzodiazepine (Tables 1 and 2)
The current interest in the anxiolytic properties of BM (Bhattacharya and Ghosal, 1998). However, BM has a
extract assumes greater relevance in view of the fact distinct advantage over LZP since it does not induce
ARTICLE IN PRESS
A. Russo, F. Borrelli / Phytomedicine 12 (2005) 305–317 309

Fig. 2. Comparative antidementic effect of GB extract and three different extracts of BM at 30 mg/kg po on TLT in single-trial PA
test, significant difference from first trial, *po0:001: Scopolamine (3 mg/kg ip) 5 min prior to first trial in all the groups except
control (vehicle-treated) to induce dementia (no significant increase in TLT on second trial).

Fig. 3. Comparative antidementic effect of GB extract and three different extracts of BM at 30 mg/kg po on the percent NTR
parameter in single-trial PA test, significant difference in second trial from dementia group, *po0:05: Dementia and control groups
are the same as in Fig. 2.

amnesia and has, instead, a memory-promoting action forced swim and learned helplessness models of depres-
in animals and man (Singh and Dhawan, 1992; Dhawan sion and was comparable to that of imipramine (Sairam
and Singh, 1996). These results were confirmed by et al., 2002).
Shanker and Singh (2000) who reported that BM extract
possessed an anxiolytic effect. Anticonvulsive action
Another important use of BM in traditional medicine
Antidepressant activity is anticonvulsive action, as reported in different experi-
More recently, the standardized methanolic extract of mental studies. Shanmugasundaram et al. (1991) have
BM was investigated for potential antidepressant reported that the crude water extract of BM controls
activity in rodent models of depression. The effect was epilepsy in experimental animals. Successively, BM
compared with the standard antidepressant drug imi- was studied in mice and rats, at oral doses ranging
pramine (15 mg/kg ip). When given in the dose of 20 and between 1 and 30 g/kg, for its effect on the central
40 mg/kg, orally once daily for 5 days, the extract was nervous system. The test material was studied for its
found to have significant antidepressant activity in effect on pentobarbitone hypnosis, motor co-ordination,
ARTICLE IN PRESS
310 A. Russo, F. Borrelli / Phytomedicine 12 (2005) 305–317

Table 1. Effects of BM and LZP on the open-field test in rats (data are means7SEM)

Groups (mg/kg) n Squares crossed (N) Immobility (s) Rears (N) Faecal pellets (N)

Control (vehicle) 14 123.479.8 29.475.8 21.373.5 5.270.9


BM (5, p.o.) 8 193.778.5 23.272.8 28.474.0 4.470.8
BM (10, p.o.) 8 166.278.3* 18.473.6* 36.373.2* 3.170.7*
BM (20, p.o.) 8 176.276.6* 18.972.6* 44.273.3* 2.470.6*
LZP (0.5, i.p.) 8 169.474.9* 16.372.0* 38.172.6* 3.070.8*

*po0:05 different from control group.

Table 2. Effects of BM and LZP on the elevated plus-maze test in rats (data are means7SEM)

Groups n Total time spent on Total entries on % time spent on % entries on open
both open and both open and open arms (s) arms (N)
closed arms (s) closed arms (N)

Control (vehicle) 15 229.4716.8 23.271.4 22.974.0 36.274.6


BM (5, p.o.) 8 216.0712.3 27.972.6 31.674.8 45.475.1
BM (10, p.o.) 8 239.6711.2 27.472.9 46.473.9* 53.674.4*
BM (20, p.o.) 8 239.579.6 30.374.2 59.473.9* 66.272.8*
LZP (0.5, i.p.) 6 232.0710.1 29.673.4 53.973.5* 68.773.8*

*po0:05 different from control group.

tail-withdrawal reaction time, electroshock, chemocon- Antioxidant activity


vulsions, haloperidol-induced catalepsy and conditioned
avoidance response. The natural material exhibited a Data from different laboratories suggest that the
sedative effect and significantly prolonged the hypnotic cognition-promoting functions of BM may be partially
action of pentobarbitone. It produced a variable attributed to the antioxidant effects of the bacosides.
blockage of conditioned avoidance response. The One study determined the effects of alcohol and hexane
presence of a significant antinociceptive effect, coupled extract of BM on lipid peroxidation by ferrous sulphate
with the ability of BM to offer protection against and cumene hydroperoxide in rat liver homogenate
electroshock seizures and chemoconvulsions plus the (Tripathi et al., 1996). The alcohol fraction exhibited a
ability to antagonize haloperidol-induced catalepsy, greater protection against both inducers. The results
suggests an involvement of the GABA-ergic system in were also compared with known antioxidants, tris (2-
the mediation of central nervous system effects of BM amino-2-hydroxymethyl-1,3-propanediol) a hydroxyl
(Singh et al., 1996). Substances which stimulate GABA trapper, EDTA, a metal chelator, and the natural
are known to possess anticonvulsant, pain relieving and antioxidant, vitamin E (100 mg of the alcoholic extract
sedative effects (Shanker and Singh, 2000). of BM was equivalent to 247 mg of EDTA and 58 mg of
On the basis of these studies, BM was evaluated vitamin E). EDTA and Vitamin E, offered dose-
alone and in combination with phenytoin (PHT) (one dependent protection against ferrous sulphate-induced
of the widely used anticonvulsants known to adver- peroxidation, BM only slightly protected against the
sely affect cognitive function), for its effect on PA oxidation of reduced glutathione suggesting that BM
task, maximal electroshock seizures and locomotor mechanism of antioxidant action could be attributed to
activity in mice (Vohora et al., 2000). PHT adversely metal chelation at the initiation level of the free radical-
affected cognitive function in the PA task. BM reversed induced chain reaction or to the quenching of free
PHT-induced impairment (Tables 3 and 4). Both radicals at propagation level.
acquisition and retention of memory showed improve- A more recent study explored the effect of BM extract
ment without affecting PHT anticonvulsivant activity. on the rat brain frontal cortical, striatal, and hippo-
Further investigations using BM alone or in com- campal superoxide dismutase (SOD), catalase (CAT)
bination with other antiepileptic drugs are warranted and glutathione peroxidase (GPX) activities, following
to explore the full potential of BM in epilepsy and administration for 7, 14 or 21 days (Bhattacharya et al.,
in correcting antiepileptic drug-induced cognitive 2000). The results were compared with the effect
impairment. induced by deprenyl, a well-known neurological anti-
Table 3. Effect of phenytoin, BM extract and their combination (chronic treatment) on acquisition and retention performance in a passive-avoidance paradigm in mice

Treatments (per Trials required Retention (2 h) Retention (24 h)


kg po; n ¼ 10) for acquisition
(number)
SDL (s) SDE (number) TSZ (s) SDL (s) SDE (number) TSZ (s)

Distilled water 3.470.25 189.9732.40 4.270.487 51.3710.02 424.0745.69 1.070.24 6.473.07


(10 ml  14 days)
PHT (25 mg  14 4.770.20*** 60.8715.52** 6.871.33b 259.1757.23c** 101.4710.61C*** 2.670.35C** 7.5730.37b*
days)
BM (40 mg  7 2.370.14d*** 203.0725.83d** 2.670.2** 28.972.53b* 489.0746.97b 0.770.24 1.070.28*
days)
PHT (25 mg  14 4.070.31 71.6712.70** 3.070.20* 74.1716.46 290.7754.85 1.170.17 11.275.23
days)+BM

A. Russo, F. Borrelli / Phytomedicine 12 (2005) 305–317


(40 mg  7 days)
a
PHT, phenytoin; BM, Bacopa monniera extract; SDL, step down latency; SDE, step-down errors; and TSZ, time in shock zone.
*po0.05.

ARTICLE IN PRESS
**po0.01.
***po0:001 versus control; b, po0:05; c, po0:01; d, po0:001 versus combined treatment. Significant by one-way ANOVA and students’s t-test.

Table 4. Effect of phenytoin, BM extract and their combination (chronic treatment) on maximal electroshock-induced seizure in mice

Sl. no. Treatment (per kg; po) n Duration (mean7SEM, s)

Flexion Extension Clonus Stupor

I Distilled water (10 ml  14 days) 10 1.270.12 13.070.51 8.670.69 210.775.10


II PHT (25 mg  14 days) 10 1.770.20* 0.070.00*** 16.571.05*** 98.9728.96**
III BM (40 mg  7 days) 10 1.270.12 12.570.74d 9.170.55d 201.577.30d
IV PHT (25 mg  14 days)+BM (40 mg  7 days) 10 1.670.21 0.070.00*** 16.870.85*** 89.6726.95***
V PHT (10 mg  14 days) 5 1.470.21 6.072.31 7.471.34 108.0713.20
VI PHT (25 mg  14 days)+BM (40 mg  7 days) 5 1.270.17 5.072.74 7.271.03 123.0728.75
a
n ¼ number of animals.
*po0.05.
**po0.01.
***po0.001 versus I; d: po0.001 versus IV (Significance by ANOVA and student’s t-test).

311
ARTICLE IN PRESS
312 A. Russo, F. Borrelli / Phytomedicine 12 (2005) 305–317

oxidant for the same time period. BM induced a dose- the marker 20 ,70 -dichlorodihydrofluorescein diacetate,
related increase in SOD, CAT, and GPX activities in all and caused a fragmentation of genomic DNA (examined
brain regions investigated, after treatment for 14 or 21 by COMET assay) compared to control astrocytes.
days. On the contrary, deprenyl induced an increase in Methanolic extract of BM (3–6–12 mg/ml) added to the
SOD, CAT, GPX activities in the frontal cortex and cells during SNAP treatment, reduced the intracellular
striatum, but not in the hippocampus, after 12 or 21 oxidants, consequently preventing DNA damage (Russo
days. Pawar et al. (2001) demonstrated that the et al., 2003b).
hydroalcoholic extract of the whole BM plant exhibited
an inhibitory effect on superoxide released from
polymorphonuclear cells in nitroblue tetrazolium assay. Other biological effects
The bacoside A3 was found to be responsible for this
effect. Besides mental functioning, Ayurvedic texts advocate
Sumathy et al. (2001) investigated the hepato- the use of BM in other physiological conditions. As a
protective activity of BM alcohol extract, administered result, researchers have evaluated the anti-inflamma-
orally, on the liver antioxidant status of morphine- tory, cardiotonic and other pharmacological effects of
treated rats. The results obtained demonstrated that BM preparations/extracts.
coadmnistration of BM exerted a protective influence The effect of BM in inhibiting experimentally induced
against the inhibition of antioxidant enzymes and inflammation was compared with that of indomethacin,
reduction in GSH level in rats. Further, BM inhibited a known anti-inflammatory drug. The results of this
the lipid peroxide formation of morphine administra- study showed that BM effectively suppressed experi-
tion, thereby clearly establishing its protective role in mentally induced inflammatory reactions, by inhibiting
morphine toxicity in rat liver. A more recent study, prostaglandin synthesis and partly by stabilizing lyso-
affected by the same research group, indicated the somal membranes, and did not cause gastric irritation at
protective effect of BM extract on morphine-decreased anti-infiammatory doses (Jain et al., 1994).
brain mitochondrial enzyme activity in rats (Sumathy Fresh BM juice (BMJ) has been reported to have
et al., 2002). significant antiulcerogenic activity (Rao et al., 2000).
In a study to test the hypothesis that BM directly The antiulcerogenic effect of BMJ was examined using
decreases the production of free radicals, we used the gastric ulcer models induced by ethanol, aspirin, 2 h
Paoletti assay, which excludes the Fenton-type reaction cold-restraint stress and 4 h pylorus ligation. BMJ
and the xanthine–xanthine oxidase system (Russo et al., 100–300 mg/kg produced significant antiulcer activity
2003a). Our results indicate that BM is able to directly in all the experimental gastric ulcer models except in the
inhibit the superoxide anion formation in a dose- case of ethanol-induced ulcers where 100 mg/kg was not
dependent manner. The free radical scavenging activity found to have a significant effect. BMJ (100–300 mg/kg)
of this methanolic extract was also evaluated through its was found to have little or no effect on offensive
ability to quench the synthetic radical 1,1-diphenyl-2- acid–pepsin secretion, while cell shedding and mucin
picryl-hydrazyl (DPPH). The free radical scavenging secretion in terms of total carbohydrates and protein
capacity of BM, demonstrated in the Paoletti and in ratio (TC:P), the two important parameters of defensive
DPPH tests, was confirmed by protection against factors, respectively, decreased and increased signifi-
plasmid DNA strand scission, induced by  OH radicals, cantly, indicating enhancement of protective mucosal
generated from UV photolysis of H2O2. In fact, this factors. Both BMJ (300 mg/kg) and sucralphate, used as
extract suppressed the formation of linDNA and the reference compound, showed a tendency to increase
induced a partial recovery of scDNA. Moreover, this the mucosal glycoproteins in terms of TC:P. Thus, ulcer
extract exhibited a protective effect on H2O2-induced protective effect of BMJ may be due to its effect on
cytotoxicity and DNA damage in human non-immorta- mucosal defensive factors such as enhanced mucin
lized fibroblasts. secretion, mucosal glycoprotein and decreased cell
There is growing evidence that high concentrations of shedding, rather than on offensive factors such as acid
nitric oxide (NO), generated (enzymatically and non- and pepsin. Moreover, Sairam et al. (2001) reported that
enzymatically) by activated astrocytes, might be involved BME, standardized to bacoside A content, when given
in a variety of neurodegenerative diseases, such as AD, in the dose of 10–50 mg/kg, twice daily for 5 days,
ischemia and epilepsy (Colasanti and Suzuki, 2000). showed dose-dependent antiulcerogenic action on var-
Therefore, a study was designed to examine the effect of a ious gastric ulcer models induced by ethanol, aspirin, 2 h
methanolic BM extract on toxicity induced by the NO cold-restraint stress, and 4 h pylorus ligation. BME
donor, S-nitroso-N-acetyl-penicillamine (SNAP), in cul- 20 mg/kg showed no effect on acid–pepsin secretion,
ture of rat astrocytes. The results indicated that after 18 h but increased mucin secretion and decreased cell
of treatment, SNAP determined an increase in the shedding with no effect on cell proliferation. In the
intracellular oxidants, as demonstrated by oxidation of same study, it was reported that BME effectively
ARTICLE IN PRESS
A. Russo, F. Borrelli / Phytomedicine 12 (2005) 305–317 313

alleviated stress-induced ulcers with a marked decrease alkaloids (Yano et al., 1991), terpenes (Hwang et al.,
in LPO, increasing SOD and CAT activity. More 1987) and flavonoids (Di Carlo et al., 1999). The plant
recently, Goel et al. (2003) reported that a standardized extract inhibited the spontaneous movements of both
BM extract in the dose of 1000 mg/ml showed anti- guinea-pig ileum (IC50 ¼ 2474 mg/ml) and rabbit jeju-
Helicobacter pylori activity in vitro. num (IC50 ¼ 13679 mg/ml). A marked reduction in
As stress is a factor in many diseases, research on an acetylcholine and histamine-induced responses in the
effective antistress agent obtained from plants has gained ileum was evident in the presence of extract (260 mg/ml).
importance. Rai et al. (2003) have evaluated the effect of Acetylcholine (1 mM)-induced contraction in the ileum
a standardized extract of BM against acute (AS) and was also inhibited by the extract in a dose-dependent
chronic stress (CS) models in rats. Pretreatment with BM manner. Calcium chloride-induced responses in rabbit
at 40 mg/kg per os significantly reduced the AS-induced blood vessels and jejunum were attenuated in the
changes in adrenal gland weight, spleen weight, plasma presence of plant extract (10–700 mg/ml) implying a
glucose, alanine aminotransferase and aspartate amino- direct effect of the extract on the influx of calcium ions
transferase (AST). CS exposure resulted in a significant into cells. However, the absence of any modification of
increase in the ulcer index, adrenal gland weight, plasma either noradrenaline or caffeine-induced contractions in
AST, and creatine kinase (CK) with a significant decrease the presence of BM extract suggests that this natural
in thymus and spleen weight, plasma triglyceride, and compound has no detectable effect on mobilization of
cholesterol. Pretreatment with low dose of BM extract at intracellular calcium. The author, therefore hypothe-
40 mg/kg significantly reversed changes in ulcer index and sized that the spasmolytic effect of BM extract in
plasma AST only, whereas the pretreatment with higher smooth muscles is predominantly due to inhibition of
dose significantly reversed CS-induced changes in ulcer calcium influx via both voltage- and receptor-operated
index, adrenal gland weight, CK, and AST. On the basis calcium channels of the cell membrane. A recent study
of these results, therefore, the authors suggested that the by Channa et al. (2003) demonstrates that various
standardized extract of BM possesses an adaptogenic fractions and subfractions isolated from BM produced
activity. significant inhibition of carbachol-induced bronchocon-
The anticancer nature of this plant extract was striction, hypotension and bradycardia in anaesthetized
reported in Walker carcinoma; a subsequent study rats, and that methanol fraction and CHCl3/MeOH
showed that BME significantly inhibited Sarcoma-180 subfraction caused a marked reduction in calcium
cell growth in vitro (Elangovan et al., 1995). chloride-induced contraction on guinea-pig ileum, in-
Dar and Channa (1997a) demonstrated that BME dicating their interference with Ca2+ ion movement.
possessed relaxant properties in blood vessels and In addition, it has been reported that BM methanolic
tracheal preparations from rabbit and guinea-pigs with extract exhibited a potent activity comparable to
a partial contribution by b-adrenoreceptors and pros- disodium cromoglycate, a known mast cell stabilizer,
taglandins. BM also produced bronchodilation in indicating the potential usefulness of BM leaves in
anaesthetized rats (Dar and Channa, 1997b), supporting allergic conditions (Samiulla et al., 2001).
the traditional use of this plant in folklore medicine for
various respiratory ailments (Chopra et al., 1956;
Nadkarni, 1976). This study demonstrated that the Clinical studies
BME possessed a bronchodilatatory property, clearly
reflected by antagonism of carbachol-induced effects on Supporting the preclinical investigations, several
inspiratory and expiratory pressures. Additionally, the clinical studies, summarized in Table 5, have been
plant extract at 37 and 50 mg/kg also inhibited carried out. Two singe-blind open clinical studies have
carbachol-induced hypotension. The plant extract ex- reported memory and learning enhancing effects of
hibited a dual action on bronchoconstriction induced by chronic BM treatment in both patients with anxiety
carbachol. At low doses (25 and 37 mg/kg), it predomi- neurosis (Singh and Singh, 1980) and in children
nantly inhibited inspiratory pressure, but at a high dose (Sharma et al., 1987). Therefore, BM has been
(50 mg/kg) inhibited only expiratory pressure. This introduced onto the market in India and other
property of the plant extract implies that more than countries, alone or in association with other phytocom-
one mechanism of action may be responsible for plexes, and utilized in the treatment of memory and
bronchodilation. Some of the possible mechanisms attention disorders (Shukia et al., 1987). The commer-
include b-adrenoceptor activation, muscarinic receptor cial preparation has shown a remarkable nootropic
antagonism, prostaglandin release or interference with activity, above all in very young subjects (Dave et al.,
calcium mobilization. A more recent study by the same 1993). After clinical trials on human volunteers, a
authors demonstrates the calcium antagonistic activity standardized extract of BM has now been made
in ethanol extract of BM (Dar and Channa, 1999), as available for clinical use by the Central Drug Research
reported in various groups of natural products such as Institute, Lucknow, India (Dhawan and Singh, 1996).
ARTICLE IN PRESS
314 A. Russo, F. Borrelli / Phytomedicine 12 (2005) 305–317

Table 5. Clinical studies of BM

Substances/treatments Subjects/patients Results/comments References

BM crude extract; chronically Patients with anxiety neurosis Enhancing of memory Singh and Singh (1980)
for 4 weeks
BM extract; chronically for Children Enhancing memory and Sharma et al. (1987)
12 weeks learning
BM standardized extract Mentally retarded children Effective in enhancing Dave et al. (1993)
learning and in controlling
abnormal behaviour
BM standardized extract Healthy adult subjects Improving early information Stough et al. (2001)
(300 mg); chronically for 12 processing, verbal learning
weeks and memory consolidation
BM standardized extract Healthy adult subjects No significant changes were Nathan et al. (2001)
(300 mg); acute treatment for found
2h
BM standardized extract Healthy adult subjects No acute effect on cognitive Maher et al. (2002)
(300 mg) combined with was found on any of the tests
Ginkgo biloba extract
(120 mg); acute treatment for
90 and 180 min
BM standardized extract Healthy adult subjects Significant effect on a test for Roodenrys et al. (2002)
(300 mg); one trial after retention of new information.
chronic treatment for three Tasks assessing attention,
months and an other trial and verbal and visual short term
for 6 weeks after the memory and the retrieval of
completion of the trial pre-experimental knowledge
were unaffected

Recently, Stough et al. (2001) undertook a study, Follow-up tests showed that the rate of learning was
using a double-blind placebo-controlled design and a unaffected, suggesting that it decreases the rate of
battery of well-validated neuropsychological tests, to forgetting of newly acquired information. Tasks asses-
examine the chronic effects of an extract of BM on sing attention, verbal and visual short-term memory and
cognitive function in healthy human subjects. The the retrieval of pre-experimental knowledge were
authors showed that BM (300 mg) given chronically unaffected. Questionnaire assessments of every day
for 12 weeks improved early information processing, memory function and anxiety levels were also unaf-
verbal learning and memory consolidation in humans. fected.
This is consistent with the study of Sharma et al. (1987),
who also observed positive effects on learning and
memory following 12 weeks administration. However,
as reported for GB extract (Nathan et al., 2002), no Toxicological study
acute effects on cognitive function were found at a lower
dose (300 mg) (Nathan et al., 2001), even when BM It has been found to be well tolerated and without any
extract was combined with GB extract (Maher et al., untoward reaction or side effects in regulatory pharma-
2002). Therefore, as suggested by the authors, it is cological and toxicological studies.
possible that the combined administration of both these The LD50 of aqueous and alcoholic crude extracts of
extracts chronically may exert more potent effects on BM in rats were 1000 mg and 15 g/kg by intraperitoneal
cognitive function. route, respectively (Martis et al., 1992). The aqueous
Roodenrys et al. (2002), again based on behavioural crude extract given orally at a dose of 5 g/kg did not
studies with rats, examined the effects of an extract of show any toxicity. The LD50 of the alcoholic crude
BM on cognitive function. Seventy-six adults aged extract was 17 g/kg given orally.
between 40 and 65 years took part in a double-blind The safety of pharmacological doses of isolated
randomized, placebo control study in which various bacosides has been tested in healthy, male, human
memory functions were tested and levels of anxiety volunteers. A phase I single-dose tolerance study was
measured. The results showed a significant effect of conducted in 31 healthy human volunteers. The trial was
Brahmi on a test for the retention of new information. double-blind, placebo controlled and non-crossover.
ARTICLE IN PRESS
A. Russo, F. Borrelli / Phytomedicine 12 (2005) 305–317 315

The dose of bacosides A and B ranged from 20 to can have serious clinical consequences (Izzo and Ernst,
300 mg (6 volunteers in placebo group and 4 each in 20, 2001). GB raised blood pressure when combined
50, 75, 100, 150, 200 mg group, respectively, only 1 with a thiazide diuretic and coma in a patient with
volunteer received 300 mg of bacosides). A multiple dose AD taking low-dose trazodone. It is therefore extremely
study was completed in 20 healthy human subjects. Ten important to also consider the possibility of BM–drug
healthy human volunteers received 100 mg bacosides A interaction.
and B orally per day for 4 weeks, while 10 were given
200 mg bacosides A and B. A detailed pre- and post-
drug monitoring of clinical, haematological and bio-
chemical laboratory investigations did not reveal any References
drug-related abnormality. Single as well as repeated
doses of bacosides A and B/ placebo were tolerated well, Barrett, S.C.H., Strother, J.L., 1978. Taxonomy and natural
no side effects were experienced by any subject. Thus history of Bacopa in California. Syst. Bot. 5, 408–419.
single doses of bacosides A and B (100 and 200 mg) Bhattacharya, S.K., Ghosal, S., 1998. Anxiolytic activity of a
administered for 4 weeks have been tolerated well and standardized extract of Bacopa monniera: an experimental
are devoid of any untoward reaction or side effects study. Phytomedicine 5, 77–82.
Bhattacharya, S.K., Kumar, A., Ghosal, S., 1999. Effect of
(Singh and Dhawan, 1997).
Bacopa monniera on animal models of Alzheimer’s disease
and perturbed central cholinergic markers of cognition in
rats. In: Siva Sankar, D.V. (Ed.), Molecular Aspects of
Asian Medicines. PJD Publications, New York.
Conclusion Bhattacharya, S.K., Bhattacharya, A., Kumar, A., Ghosal, S.,
2000. Antioxidant activity of Bacopa monniera in rat frontal
BM, a traditional Ayurvedic medicinal plant has been cortex, striatum and hippocampus. Phytother. Res. 14,
used for centuries as a memory-enhancing, anti-inflam- 174–179.
matory, analgesic, antipyretic, sedative, and antiepilep- Bose, K.C., Bose, N.K., 1931. Observations on the actions and
tic agent. More recently, preclinical studies have uses of Herpestis monniera. J. Indian Med. Assoc. 1, 60.
reported cognitive enhancing effects with various Chakravarty, A.K., Sarkar, T., Masuda, K., Shiojima, K.,
Nakane, T., Kawahara, N., 2001. Bacopa side I and II: two
extracts of BM, but the exact mechanism of its actions
pseudojujubogenin glycosides from Bacopa monniera.
is still uncertain, as its multiple active constituents make
Phytochemistry 58, 553–556.
its pharmacology complex. It has been suggested that Chakravarty, A.K., Sarkar, T., Nakane, T., Kawahara, N.,
BM, like GB, exhibits neuroprotective and cognitive Masuda, K., 2002. New phenylethanoid glycosides from
enhancing effects, in part due to its, capacity to Bacopa monniera. Chem. Pharm. Bull. 50, 1616–1618.
modulate the cholinergic system (Bhattacharya et al., Chakravarty, A.K., Garai, S., Masuda, K., Nakane, T.,
1999) and to contrast oxidative stress (Bhattacharya et Kawahara, N., 2003. Bacopasides III–V: three new
al., 2000; Russo et al., 2003a, b). Therefore, in view of triterpenoid glycosides from Bacopa monniera. Chem.
the important actions performed by this plant, further Pharm. Bull. 51, 215–217.
research must be undertaken into the recent findings Chandel, R.S., Kulshreshtha, D.K., Rastogi, R.P., 1977.
described here. Bacogenin A3: a new sapogenin from Bacopa monniera.
Phytochemistry 16, 141–143.
Although pre-clinical animal studies have shown that
Channa, S., Dar, A., Yaqoob, M., Anjum, S., Sultani, Z.,
BM, like GB, has nootropic effects in established
Rahman, A., 2003. Broncho-vasodilatatory activity of
learning and memory models, few clinical studies have fractions and pure constituents isolated from Bacopa
been performed to complement these findings. Litera- monniera. J. Ethnopharmacol. 86, 27–35.
ture data reported that BM given chronically improved Chatterji, N., Rastogi, R.P., Dhar, M.L., 1965. Chemical
early information processing and verbal learning and examination of Bacopa monniera Wettst.: parti-isolation of
memory consolidation in humans (Stough et al., 2001). chemical constituents. Indian J. Chem. 3, 24–29.
However, as reported for GB extract (Nathan et al., Chopra, R.N., Nayar, L., Chopra, I.C., 1956. Glossary of
2002), no acute effects on cognitive function were found Indian Medicinal Plants, vol. 32. Council of Scientific and
at a lower dose (Nathan et al., 2001), even when BM Industrial Research, New Delhi.
extract was combined with GB extract (Maher et al., Chunekar, K.C., 1960. Bhav Prakasa Nighantu, vol. 372.
Chaukhamba Bharati Publications, Varanasi (Hindi trans-
2002). As suggested by the authors, it is possible that the
lation).
combined administration of both these extracts chroni-
Colasanti, M., Suzuki, H., 2000. The dual personality of NO.
cally may exert more potent effects on cognitive TPS 21, 249–252.
function. Therefore, other clinical investigations have Dar, A., Channa, S., 1997a. Relaxant effect of ethanol extract
to be encouraged, also in order to evidence any possible of Bacopa monniera on trachea, pulmonary artery and
side effects. Different studies indicated that interactions aorta from rabbit ad guinea-pig. Phytother. Res. 11,
between herbal medicines and synthetic drugs exist and 323–325.
ARTICLE IN PRESS
316 A. Russo, F. Borrelli / Phytomedicine 12 (2005) 305–317

Dar, A., Channa, S., 1997b. Bronchodilatatory and cardio- Martis, G., Rao, A., Karanth, K.S., 1992. Neuropharmaologi-
vascular effects of an ethanol extract of Bacopa monniera in cal activity of Herpestis monniera. Fitoterapia 63, 399–404.
anaesthetized rats. Phytomedicine 4, 319–323. Mathew, K.M., 1984. The flora of Tamil Nadu and Carna.
Dar, A., Channa, S., 1999. Calcium antagonistic activity of Rapinat Herbarium St. Joseph’s College, Tiruchirapalli,
Bacopa monniera on vascular and intestinal smooth muscles India.
of rabbit and guinea-pig. J. Ethnopharmacol. 66, 167–174. Nadkarni, K.M., 1976. Indian Materia Medica. Popular
Das, A., Shanker, G., Nath, C., Pal, R., Singh, S., Singh, H., Prakashan Private, Bombay, pp. 624–625.
2002. A comparative study in rodents of standardized Nathan, P.J., Clarke, J., Lloyd, J., Hutchison, C.W., Downey,
extracts of Bacopa monniera and Ginkgo biloba. Pharmacol. L., Stough, C., 2001. The acute effect of an extract of
Biochem. Behav. 73, 893–900. Bacopa monniera (Brahmi) on cognitive function in healthy
Dave, U.P., Chauvan, V., Dalvi, J., 1993. Evaluation of BR-16 normal subjects. Hum. Psychopharmacol. 16, 345–351.
A (Mentat) in cognitive and behavioural dysfunction of Nathan, P.J., Ricketts, E., Wesnes, K., Mrazek, L., Greville, W.,
mentally retarded children: a placebo-controlled study. Stough, C., 2002. The acute nootropic effects of an extract of
Indian J. Pediatr. 60, 423–428. Gingko biloba in healthy elderly human subjects: a preliminary
Dhawan, B.N., Singh, H.K., 1996. Pharmacology of ayurvedic investigation. Hum. Psychopharmacol. 17, 45–49.
nootropic Bacopa monniera, Abstr. No. NR 59. Int. Conv. Pawar, R., Gopalakrishnan, C., Bhutani, K.K., 2001. Dam-
Biol. Psychiat. Bombay. marane triterpene saponin from Bacopa monniera as the
Di Carlo, G., Mascolo, N., Izzo, A.A., Capasso, F., 1999. superoxide inhibitor in polymorphonuclear cells. Planta
Flavonoids: olds and new aspects of a class of natural Med. 67, 752–754.
therapeutic drugs. Life Sci. 65, 337–353. Rai, D., Bhatia, G., Palit, G., Pal, R., Singh, S., Singh, H.K.,
Elangovan, V., Govindasamy, S., Ramamoorthy, N., Balasu- 2003. Adaptogenic effect of Bacopa monniera (Brahmi).
bramanian, K., 1995. In vitro studies on the anticancer Pharmacol. Biochem. Behav. 75, 823–830.
activity of Bacopa monnieri. Fitoterapia 66, 211–215. Rao, C.H.V., Sairam, K., Goel, R.K., 2000. Experimental
Garay, S., Mahato, S.B., Ohtani, K., Yamasaki, K., 1996a. evaluation of Bacopa monniera on rat gastric ulceration and
Dammarane-type triterpenoid saponins from Bacopa mon- secretion. Indian J. Physiol. Pharmacol. 44, 435–441.
niera. Phytochemistry 42, 815–820. Rastogi, R.P., 1990. Compendium of Indian Medicinal Plants,
Garay, S., Mahato, S.B., Ohtani, K., Yamaski, K., 1996b. vol. 1. CSIR, New Delhi, pp. 118–122.
Bacosaponin D-a pseudojujubogenin glycoside from Bacopa Rastogi, S., Pal, R., Kulshreshtha, D.K., 1994. Bacoside A3-a
monniera. Phytochemistry 43, 447–449. triterpenoid saponin from Bacopa monniera. Phytochem-
Goel, R.K., Sairam, K., Babu, M.D., Tavares, I.A., Raman, istry 36, 133–137.
A., 2003. In vitro evaluation of Bacopa monniera on anti- Roodenrys, S., Booth, D., Bulzoni, S., Phipps, A., Micallef, C.,
Helicobacter pylori activity and accumulation of prosta- Smoker, J., 2002. Chronic effects of Brahami (Bacopa
glandins. Phytomedicine 10, 523–527. monnieri) on human memory. Neuropsychopharmacology
Hou, C.C., Lin, S.J., Cheng, J.T., Hsu, F.L., 2002. Bacopaside 27, 279–281.
III, bacopasaponin G, and bacopasides A, B, and C from Russo, A., Izzo, A.A., Borrelli, F., Renis, M., Vanella, A.,
Bacopa monniera. J. Nat. Prod. 65, 1759–1763. 2003a. Free radical scavenging capacity and protective
Hwang, S.B., Chang, M.N., Garcia, M.L., Han, Q.Q., Huang, effect on DNA damage of Bacopa monniera L. Phytother.
L., King, V.F., Kaczorowski, G.J., Winquist, R.J., 1987. Res. 17, 870–875.
L652, 469-a dual receptor antagonist of platelet activating Russo, A., Borrelli, F., Campisi, A., Acquaviva, R., Raciti, G.,
factor and dihydropyridines from Tussilago farfara L. Eur. Vanella, A., 2003b. Nitric oxide-related toxicity in cultured
J. Pharmacol. 141, 269–281. astrocytes: effect of Bacopa monniera. Life Sci. 73, 1517–1526.
Izzo, A.A., Ernst, E., 2001. Interactions between herbal Sairam, K., Rao, C.V., Babu, M.D., Goel, R.K., 2001.
medicines and prescribed drugs: a systematic review. Drugs Prophylactic an curative effects of Bacopa monniera in
61, 2163–2175. gastric ulcer models. Phytomedicine 8, 423–430.
Jain, P., Khanna, N.K., Trehan, T., Pendse, V.K., Godhwani, Sairam, K., Dorababu, M., Goel, R.K., Bhattacharya, S.K.,
J.L., 1994. Antiinflammatory effects of an Ayurvedic 2002. Antidepressant activity of standardized extract of
preparation, Brahmi Rasayan, in rodents. Indian J. Exp. Bacopa monniera in experimental models of depression in
Biol. 32, 633–636. rats. Phytomedicine 9, 207–211.
Kulshreshtha, D.K., Rastogi, R.P., 1973. Bacogenin A1: a Samiulla, D.S., Prashanth, D., Amit, A., 2001. Mast cell
novel dammerane triterpene sapogenin from Bacopa stabilising activity of Bacopa monnieri. Fitoterapia 72,
monniera. Phytochemistry 12, 887–892. 284–285.
Kulshreshtha, D.K., Rastogi, R.P., 1974. Bacogenin A2: a new Sara, S.J., 1989. Noradrenergic–cholinergic interaction: its
sapogenin from bacosides. Phytochemistry 13, 1205–1206. possible role in memory dysfunction associated with senile
Maher, B.F., Stough, C., Shelmerdine, A., Wesnes, K., Nathan, dementia. Arch. Gerontol. Geriatr. 1S, 99–108.
P.J., 2002. The acute effects of combined administration of Sastri, M.S., Dhalla, N.S., Malhotra, C.L., 1959. Chemical
Ginkgo biloba and Bacopa monniera on cognitive function in investigation of Herpestis monniera Linn (Brahmi). Indian
humans. Hum. Psychopharmacol. 17, 163–164. J. Pharmacol. 21, 303–304.
Malhotra, C.L., Das, P.K., 1959. Pharmacological studies of Satyavati, G.V., Raina, M.K., Sharma, M., 1976. Medicinal
Herpestis monniera Linn. (Brahmi). Indian J. Med. Res. 47, Plants of India, vol I. Indian Council of Medical Research,
294–305. New Delhi, pp. 112–118.
ARTICLE IN PRESS
A. Russo, F. Borrelli / Phytomedicine 12 (2005) 305–317 317

Shanker, G., Singh, H.K., 2000. Anxiolytic profile of Singh, H.K., Shanker, G., Patnaik, G.K., 1996. Neurophar-
standardized Brahmi extract. Indian J. Pharmacol. 32, 152. macological and anti-stress effects of bacosides: a memory
Shanmugasundaram, E.R., Akbar, G.K., Shanmugasundar- enhancer. Indian J. Pharmacol. 28, 47.
am, K.R., 1991. Brahmighritham, an Ayurvedic herbal Stough, C., Lloyd, J., Clarke, J., Downey, A.L., Hutchison,
formula for the control of epilepsy. J. Ethnopharmacol. 33, C.W., Rodgers, T., Nathan, P.J., 2001. The chronic effects
269–276. of an extract of Bacopa monniera (Brahmi) on cognitive
Sharma, R., Chaturvedi, C., Tewari, P.V., 1987. Efficacy of function in healthy human subjects. Psychopharmacology
Bacopa monniera in revitalizing intellectual functions in 156, 481–484.
children. J. Res. Educ. Indian Med. 1, 12. Sumathy, T., Subramanian, S., Govidasamy, S., Balakrishna,
Shukia, B., Khanna, N.K., Godhwani, J.L., 1987. Effect of K., Veluchamy, G., 2001. Protective role of Bacopa
Brahmi Rasayan on the central nervous system. J. monniera on morphine induced hepatotoxicity in rats.
Ethnopharmacol. 21, 65–74. Phytother. Res. 15, 643–645.
Singh, H.K., Dhawan, B.N., 1982. Effect of Bacopa monniera Sumathy, T., Govindasamy, S., Balakrishna, K., Veluchamy,
extract on avoidance responses in rat. J. Ethnopharmacol. G., 2002. Protective role of Bacopa monniera on morphine-
5, 205–214. induced brain mitochondrial enzyme activity in rats.
Singh, H.K., Dhawan, B.N., 1992. Drugs affecting learning Fitoterapia 73, 381–385.
and memory. In: Tandon, P.N., Bijiani, V., Wadhwa, S. Tripathi, Y.B., Chaurasia, S., Tripathi, E., Upadhyay, A.,
(Eds.), Lectures in Neurobiology, vol. 1. Wiley Eastern, Dubey, G.P., 1996. Bacopa monniera Linn. as an antiox-
New Delhi, pp. 189–207. idant: mechanism of action. Indian J. Exp. Biol. 34,
Singh, H.K., Dhawan, B.N., 1997. Neuropsychopharmacolo- 523–526.
gical effects of the Ayurvedic nootropic Bacopa monniera Uphof, J.C., 1968. The dictionary of economic plants, vol. 62.
Linn (Brahmi). Indian J. Pharmachol. 29, S359–S365. Verlag von J Cramer, NewYork.
Singh, R.H., Singh, L., 1980. Studies on the anti-anxiety effect Vohora, D., Pal, S.N., Pillai, K.K., 2000. Protection from
of the medyha rasayana drug Brahmi (Bacopa monniera phenytoin-induced cognitive deficit by Bacopa monniera,
Wettst.). Res. Ayur. Siddha 1, 133–148. a reputed nootropic plant. J. Ethnopharmacol. 71,
Singh, H.K., Srimal, R.C., Srivastava, A.K., Garg, N.K., 383–390.
Dhan, B.N., 1990. Neuropsychopharmacological effects of Yano, S., Horiuchi, H., Horie, S., Aimi, N., Sakai, S.,
bacosides A and B. Proceedings of the Fourth Conference Watanabe, K., 1991. Ca2+ channel blocking effects of
on Neurobiology Learning Memory, Abstract No. 79. hirsutine, an indole alkaloid from Uncaria genus, in the
Irvine California. isolated rat aorta. Planta Med. 57, 403–405.

You might also like