You are on page 1of 13

PHYTOTHERAPY RESEARCH

Phytother. Res. 20, 1023–1035 (2006)


Published online 14 August POTENTIAL
2006 in Wiley InterScience
MEDICINAL PLANTS FOR CNS DISORDERS 1023
(www.interscience.wiley.com) DOI: 10.1002/ptr.1970

REVIEW ARTICLE
Potential Medicinal Plants for CNS Disorders:
an Overview

Vikas Kumar*
Department of Pharmaceutical Sciences, Texas Tech University, Health Sciences Center, School of Pharmacy, Amarillo, TX
79106, USA

Although very few drugs are currently approved by regulatory authorities for treating multi-factorial ailments
and disorders of cognition such as Alzheimer’s disease, certain plant-derived agents, including, for example,
galantamine and rivastigmine (a semi-synthetic derivative of physostigmine) are finding an application
in modern medicine. However, in Ayurveda, the Indian traditional system of medicine which is more than
5000 years old, selected plants have long been classified as ‘medhya rasayanas’, from the Sanskrit words
‘medhya’, meaning intellect or cognition, and ‘rasayana’, meaning ‘rejuvenation’. These plants are used both
in herbal and conventional medicine and offer benefits that pharmaceutical drugs lack. In the present article,
an attempt has been made to review the most important medicinal plants, including Ginkgo biloba, St John’s
wort, Kava-kava, Valerian, Bacopa monniera and Convolvulus pluricaulis, which are widely used for their
reputed effectiveness in CNS disorders. Copyright © 2006 John Wiley & Sons, Ltd.

Keywords: medicinal plants; Alzheimer’s disease; depression; anxiety; stress; CNS disorders.

psychotherapeutics based on acquired knowledge from


INTRODUCTION herbal remedies continue to be rare. The fact that the
vast majorities of currently available psychoactive drugs
Herbs, which have always been the principal form of are not natural products or are not derived from
medicine in developing countries, are once again bioactive constituents of medicinal plants seems to be a
becoming popular throughout the developing and reflection of such a situation. This is not, however, the
developed world. Statistics show that more and more situation in many other therapeutic areas where exam-
people in Europe, North America and Australia are ples of drugs derived from secondary plant metabolites
consulting trained herbal professionals and using and their derivatives, or conceived from pharmacolog-
the plant medicines that their grandparents or great- ical knowledge gained from studies of herbal remedies,
grandparents took. Sparreboom et al. (2004) indicated are abundant. In a systematic analysis published during
that interest in the use of herbal products has grown 1997 it was assessed that 157 of 520 drugs (30%)
dramatically in the Western word. Ayurveda-based drug approved by Food and Drug Administration (FDA) in
discovery uses ‘reverse pharmacology’, in which drug the USA during 1983–94 (11 years) were natural prod-
candidates are first identified based on large-scale ucts or their derivatives (Cragg et al., 1997). This report
use in the population, then validated in clinical trials. revealed in addition, that when focused efforts were
Experts say that this approach can cut the time for made to discover natural products for clinical use, the
drug discovery from 12 years to 5 years or less, and success level rose dramatically. Thus during the same
for a fraction of the usual cost (Padma, 2005). period, 61% of anticancer agents approved were natu-
All critical analysis on commercial and other infor- ral products or their derivatives. In the absence of
mation available on traditionally known CNS active targeted programs for natural products, there was no
herbal remedies indicate that the most popular amongst success. Thus, there were no analgesics, antidepressants,
such remedies are those which are clinically and anxiolytics or other CNS active drugs derived from
preclinically the most well studied ones, and which are natural products which were approved during the 11
also recommended for therapeutic purposes by the year time period analysed.
health authorities of many Western and other coun- Our current analysis indicates that the situation
tries outside the USA. Although the clinical efficacy of has not changed much during more recent years.
several such herbal extract based remedies have been Although identification of hits and leads from second-
revealed repeatedly by proper clinical trials conducted ary plant metabolites continue to be a major goal of
during the past three decades, reports of concentrated many drug discovery projects, most such efforts do
efforts to develop structurally and functionally novel not concentrate on the search for agents potentially
useful for the treatment of CNS disorders. In addition,
the comparatively few reports on neuronal function
modulating activities of herbal extracts and their active
* Correspondence address: Pharmacology Laboratory, Department
of Pharmaceutics, Institute of Technology, Banaras Hindu Univerity,
constituents that do appear regularly are, in general,
Varanasi-221 005, UP, India. not subsequently evaluated adequately in terms of their
E-mail(s): neuropharmacologist@rediffmail.com, vikask@bhu.ac.in potential for identifying structurally or functionally
Copyright © 2006 John Wiley & Sons, Ltd. Received
Phytother. Res. 3 October(2006)
20, 1023–1035 2005
Accepted
DOI:1310.1002/ptr
June 2006
Copyright © 2006 John Wiley & Sons, Ltd.
1024 V. KUMAR

novel CNS active drugs. The main goals of such reports published information on such extracts is beyond the
continue to be the evaluation of traditionally known scope of this review, in the following an attempt will
CNS active herbal remedies in terms of our current be made to summarize only those potentially useful
understanding of brain functions, or to identify their for obtaining novel structurally and functionally novel
active constituents. In general, the main goal of these CNS active drugs based on our current knowledge on
reports has been to generate more evidence based Ginkgo extracts and their active constituents. Several
knowledge justifying their traditionally known thera- reviews and monographs (Boonkaew and Camper, 2005;
peutic uses. Nakanishi, 2005; Gertz and Kiefer, 2004; Sierpina et al.,
In view of the fact that the clinical efficacy of several 2003; Zimmermann et al., 2002; Z’Brun, 1995; Carrigan,
psychoactive herbal extract based remedies has been 1995; Kleijnen and Knipschild, 1992; Warburton, 1988;
demonstrated repeatedly in properly controlled clinical Boralle et al., 1988; Michel and Hosford, 1988) can be
trials, their efficacy can not be easily interpreted in terms consulted for more detailed information.
of current concepts of psychopharmacology, they Some researchers showed that bilobalide, a consti-
deserve proper attention as readily available sources tuent of Ginkgo biloba was successful in inhibiting
for structurally and functionally novel classes of CNS phospholipid breakdown and choline release under
active drugs. This communication summarizes our hypoxic conditions (Klein et al., 1997). This group
current knowledge of the major bioactivities and clini- has further established that bilobalide also inhibited
cal efficacy of some currently popular CNS active herbal glutamatergic excitotoxic membrane breakdown both
remedies, which could stimulate interest in evaluating in vitro and in vivo, an effect which may be beneficial
these herbs as potential sources for structurally and in the treatment of brain hypoxia and/or neuronal
functionally novel CNS active drug leads or hits. hyperactivity (Weichel et al., 1999). Recently, a similar
group has also showed that bilobalide inhibited an
NMDA-induced chloride flux through glycine/GABA-
operated channels, thereby preventing NMDA-induced
GINKGO BILOBA breakdown of membrane phospholipids (Klein et al.,
2003). This effect is expected to contribute to the
Since early pharmacological studies revealed that neuroprotective effects of Ginkgo biloba extracts.
Ginkgo biloba leaf specific flavonoids modulated the Chatterjee et al. (2003) demonstrated that ginkgolides
contractile functions of vascular smooth muscles, A, B, C and J were also effective blockers of glycine-
efforts were subsequently made to obtain a well stand- activated chloride channels in the hippocampal
ardized extract enriched in flavonoids. This extract is neurons of rat, whereas Kondratskaya et al. (2004a)
now often referred to as EGb 761, and until the 1990s, pointed out that Ginkgolide B preferentially blocks
almost all preclinical and clinical studies reported in chloride channels formed by heteromeric glycine
the literature were conducted with this extract only. receptors in hippocampal pyramidal neurons of rat.
It was soon realized though that flavonoids can not In our laboratory, other studies are in progress to
be the sole therapy relevant bioactive constituents and investigate bilobalide and its mechanism of action
that its clinical efficacy or pharmacological activity focused on its neuroprotective effects.
is not easily interpretable in terms of its effects on blood Dementia of the Alzheimer’s type (DAT), together
vessels only. During the 1980s, based on the results of with vascular dementia, is the most important indica-
extensive studies made in several German and French tion for Ginkgo biloba extract (EGb). The therapeutic
laboratories, it was suggested that the structurally unique efficacy of this extract is founded on its neuroprotective,
and Ginkgo biloba specific terpenic lactones, i.e. metabolic and rheological effects. In addition to these
bilobalide and ginkgolides, could be the CNS function mechanisms – which also form the basis of the activity
modulating components, and that it could be, pharmaco- of the older synthetic nootropics – the hypothesis that
logically, a novel type of so-called ‘noo-tropic’ agent DAT is due to a ‘cholinergic deficit’ at central synapses
with cognitive function modulating as well as neuro- has led, over the past decade, to the development of a
protective properties. Preclinical and clinical efficacy new group of drugs for this indication, the cholinesterase
data generated during these efforts, eventually led the (ChE) inhibitors. Thus nowadays, EGb is competing,
German and French health authorities to accept EGb on the synthetic side, with the ChE inhibitors tacrine,
761, and some other analogous Ginkgo extracts (EGbs), donepezil, rivastigmine and galantamine. In addition,
as a phytotherapeutic drug which could also be the benefits of treatment were rapidly reversed after
prescribed by registered medicinal practitioners. More ending the administration of ChE inhibitors, which did
recent preclinical and clinical studies have continued not occur to the same extent with EGb (Schulz, 2003a).
to add further evidence in support of the conviction Adverse drug reactions were more than ten times more
that Ginkgo leaf extracts are useful remedies for cop- common with the ChE inhibitors and the treatment
ing with several age-related mental health problems costs about five times higher than with EGb. Given the
of the elderly, including those associated with AD. limited therapeutic options for DAT, treatment with
Although, during the past two decades, Ginkgo extracts EGb still appears to be the method of choice compared
have remained the most well studied herbal remedies with the ChE inhibitors (Schulz, 2003a). A few another
commercialized to date in the Western world, many studies also confirmed the role of Ginkgo biloba or its
questions concerning active constituents and their modes isolated constituents in dementia and other memory
of actions can not yet be answered with certainty. disorders (Pan, 2005; Haan and Horr, 2004; Yao et al.,
In any case, it seems certain now that their pharmaco- 2004; Lee et al., 2004; Kurz and Van Baelen, 2004; Evans
logical activity profiles, active constituents and their et al., 2004; Williams et al., 2004; Colciaghi et al., 2004;
modes of actions are certainly not like those of any Gertz and Kiefer, 2004; Kanowski and Hoerr, 2003;
known CNS active drug. Since describing all the Stackman et al., 2003; Smith and Luo, 2003; Ahlemeyer
Copyright © 2006 John Wiley & Sons, Ltd. Phytother. Res. 20, 1023–1035 (2006)
DOI: 10.1002/ptr
POTENTIAL MEDICINAL PLANTS FOR CNS DISORDERS 1025

and Krieglstein, 2003a; 2003b; Le Bars, 2003; Muller functionally novel pharmacophore moiety identified in
and Chatterjee, 2003; Andrieu et al., 2003; Birks et al., the previously mentioned report could be a starting
2002; Nathan et al., 2002; Mix and Crews, 2002; Gasser point as well.
and Gasser, 2001; Wettstein, 2000; van Dongen et al.,
2000; Bastianetto et al., 2000).
During recent years, several cases of hemorrhage have
been reported to occur in coincidence with the use of HYPERICUM PERFORATUM (ST JOHN’S
Ginkgo products. Although a clear causality between WORT)
Ginkgo intake and bleeding could not be established,
these observations have generally been explained by As in the case of Ginkgo biloba, current interest in the
the platelet activating factor (PAF)-antagonistic action use of Hypericum perforatum (HP) for the treatment
of ginkgolides, which represent characteristic constitu- of mental health conditions was also triggered by
ents of Ginkgo extracts. A recent study raises serious extensive efforts made by German physicians during
doubts that the PAF antagonistic effect of Ginkgolides the 1980s. Soon after it became apparent that HP
could be responsible for bleeding in patients taking EGb extracts could be as useful for the treatment of mild
761® (Koch, 2005). Rather it appears more likely that to moderately depressed patients as imipramine and
the reported cases are isolated incidences which have that HP extracts were better tolerated by the patients
happened by chance in connection with the administra- than many synthetic antidepressants, extensive efforts
tion of Ginkgo extracts and whose frequencies are were initiated in many German industrial and academic
within the statistical variation with which such events laboratories. Observations resulting from such efforts
occur. Indeed, it has recently been suggested that have been chronologically published and mentioned
frequently other motivations may be behind the publi- elsewhere in this review. In addition, more recently
cation of possible herb–drug interactions and the side books in the Milestones in Drug Therapy series by
effects of phytomedicine (Ernst, 2003b). Another Muller (2005) and Medicinal and Aromatic Plants-
recent study indicates that the effects of EGb 761 on Industrial Profiles series by Ernst (2003a) have summa-
drug metabolizing enzymes are specific for rats and may rized nicely our current knowledge on CNS function
not be extrapolated to man (Chatterjee et al., 2005). modulating effects of HP extracts, and several other
Although our current knowledge on the active monographs and reviews have appeared (Izzo, 2004;
constituents and modes of actions of Ginkgo extracts Muller, 2003; Rodriguez-Landa and Contreras, 2003;
remain far from being satisfactory, existing preclinical Kumar et al., 2000b; Challem, 2001; Gaster and Holroyd,
and clinical evidences available now are in agreement 2000; Linde et al., 1996; Bombardelli and Morazzoni,
with the conviction that more than one active constitu- 1995). Therefore, in the following some more impor-
ent and mode of action is involved in its observed tant developments leading to the suggestion that this
clinical effects, and that the activity profile of a given herb could be used as a starting point in the search for
Ginkgo extract can not be predicted by the knowledge novel CNS active drugs are summarized only.
of its already known or proposed bioactive constituents Hyperforin, a prenylated phloroglucinol present
only. Of the known CNS active constituents of Ginkgo in this plant, has been focused on as being primarily
biloba leaf extracts, bilobalide seems to be unique, not responsible for the antidepressant activity of HP
only structurally, but also pharmacologically. Although (Bhattacharya et al., 1998a; Chatterjee et al., 1998a;
much has been published on the pharmacological prop- 1998b; Biber et al., 1998; Laakmaann et al., 1998; Muller
erties of ginkgolides, by far the vast majority of them et al., 1998). Many of the experimental and clinical
deal with their PAF antagonistic activity and that studies have confirmed the antidepressant activity of
too with that of ginkgolide B only. Since therapeutic hyperforin (Treiber et al., 2005; Vitiello et al., 2005;
relevance of modulating PAF-receptor system still Zanoli, 2004; Roz and Rehavi, 2004; Eckert et al., 2004;
remains controversial, and only a few reports on the Butterweck et al., 2003; Verotta, 2003; Zanoli et al.,
CNS function modulating effects of ginkgolides are 2002; Cervo et al., 2002; Marsh and Davies, 2002).
available at present, it is still too early to consider them Indian Hypericum perforatum (IHp) extract standard-
as lead molecules for drug discovery purposes. It must ized for hyperforin lacked MAO-A and B inhibitory
be pointed out, that despite certain structural similari- activity (Kumar et al., 2001c). Additionally, IHp showed
ties between ginkgolides and bilobalide, few analogies antidepressant, anxiolytic, antiamnestic, antiinflam-
between their CNS activity profiles could be detected. matory and analgesic and antistress activities in various
The results of a recently reported structure–activity animal models (Kumar et al., 1999; 2000a; 2000b; 2000c;
study indicate that differences in the existing molecular 2001a; 2001b; 2002; 2003; Zhang, 2004). Some research-
space around their common pharmacophore moiety (i.e. ers recently found that HP had neuroprotective effects
t-butyl substituted cyclo-pentane ring) dictate their and diminished cognitive impairment and improved
activity profile (Chatterjee et al., 2003). One of the spatial learning and memory (Trofimiuk et al., 2005;
molecules synthesized and tested to confirm this possi- Silva et al., 2004; Kumar et al., 2003; Widy-Tyszkiewicz
bility led to the identification of a structurally and func- et al., 2002; Kumar et al., 2002; Klusa et al., 2001; Kumar
tionally novel molecule (NV-31) with a therapeutically et al., 2000c). Other studies confirmed that hyperforin
interesting combination of neuro-protective, memory released hippocampal acetylcholine (Kiewert et al., 2004;
enhancing and other CNS function modulating prop- Buchholzer et al., 2002) and HP reduced the degrada-
erties with a broad safety margin. Thus it seems rea- tion rate of acetylcholine (Re et al., 2003). A recent
sonable to suggest that structure–activity studies with study from our laboratory indicated that hyperforin may
known bioactive Ginkgo constituents could be a suit- be a potential neuroprotective agent that blocks activa-
able starting point in the search for urgently needed tion of NMDA-type glutamate receptors (Kumar et al.,
novel types of CNS active drugs. The structurally and 2006; Kumar et al., 2005). Further studies testing the
Copyright © 2006 John Wiley & Sons, Ltd. Phytother. Res. 20, 1023–1035 (2006)
DOI: 10.1002/ptr
1026 V. KUMAR

possible effect of hyperforin in protein kinase C- predominantly metabolized by CYP3A4 and P-


mediated responses and in vivo experiment(s) to glycoprotein at the same time (Mannel, 2004; Ernst,
evaluate hyperforin as a potential inhibitor of water 2002; 1999; Drewe, 2000). Through these mechanisms,
transport are in progress (unpublished). HP can decrease the plasma range of prescribed drugs
The current widespread medicinal uses of HP (such as anticoagulants, oral contraceptives and antivi-
extracts concentrate mainly on their therapeutic effects ral drugs), with possible clinically serious consequences
in patients with mild to moderately severe depression, (Ruschitzka et al., 2000; Piscitelli et al., 2000; Yue et al.,
and that too with hydro-alcoholic extracts only. The 2000). Some evidence indicates that HP and selective
very first report on the efficacy of such an extract serotonin reuptake inhibitors may lead to serotonin
(Daniel, 1935) dates back much earlier than the for- overload or the serotonin syndrome, particularly in
tuitous discovery of these effects of the synthetic tri- elderly patients (Ernst, 2002; Ernst, 1999). Although
cyclic antidepressant imipramine or the MAO inhibitor these reports strongly suggest that co-medications of
iproniazide. Interestingly, however, the discovery of some other drugs with HP extracts should be made
these two synthetic antidepressants during the 1950s with caution, it can not be ignored that many known
continues to have important impacts on the develop- psychoactive drugs possess, in addition to other more
ment of modern psychotherapeutics, whereas the simi- serious side effects, potential for drug–drug interaction.
lar clinical efficacy of HP extracts remained unexplored Since all the properly controlled clinical trials of
until the last decades of the past millennium. Only very diverse types of HP extracts in patients with mild to
recently, some scattered reports on efforts to develop a moderately severe depression have consistently demon-
hyperforin derivative as an antidepressant as discussed strated efficacy and a very high safety margin, and
above or on other possible psychotherapeutic potentials that drug–drug interaction is their only potential side
of hyperforin derivatives or HP extracts have appeared. effect, such extracts can be considered as one of the
In any case, the vast majority of reports on the poten- safest known psychotherapeutic agents with proven
tial psychotherapeutic uses of this herb and its CNS clinical efficacy according to the modern concept of
active constituents appearing during the past decade ‘evidence based medicine’.
concentrate around hyperforin only. It can not be over-
emphasized though, that several other therapeutically
interesting CNS-active HP constituents have already
been identified, and that none of the therapeutically PIPER METHYSTICUM (KAVA KAVA)
interesting pharmacological properties of HP extracts
can yet be properly explained by their content of the Piper methysticum Forst. (Family Piperaceae) called
known bioactive constituents alone. In addition, avail- kava, continues to occupy a central place in everyday
able quantitative data on antidepressant like and other life in the Polynesian islands concerned. The term ‘kava’
CNS function modulating effects of hyperforin and HP and the variant ‘kawa’ are used for both the plant and
extracts strongly suggest that, quantitatively, the con- the beverage made from it. The origins of kava usage
tribution of hyperforin to the observed antidepressant- in Oceania are not known. The first pharmacological
like efficacy of HP extracts must not be a major one. evaluation of the kava pyrones was published in Lewin’s
Indeed, the clinical efficacy of HP extracts almost admirable monograph but because limited quantities
devoid of hyperforin has been reported (Käufeler of pure compounds – methysticin and yongonin – were
et al., 2001), and it has been demonstrated also that at his disposal, his data now are deemed of only histori-
hyperforin and HP extracts are more potent cognitive cal significance (Lewin, 1886). A study showed that kava
function modulating agents than antidepressants (Klusa helped to induce deep sleep that set in lasting from 2 to
et al., 2001). Thus, as often suggested in the literature, 10 h. The birds used in the study appeared to be fully
the initiation of drug discovery projects based on our recovered upon awakening (Van Veen, 1938). Many
current knowledge on hyperforin seems desirable, researchers demonstrated Kava’s effectiveness in anxi-
concentrating such efforts for obtaining novel anti- ety disorders and enhancing sleep quality through
depressants only seems not very justifiable. preclinical or placebo-controlled double-blind studies
HP has an excellent safety profile clearly superior to (Capasso and Sorrentino, 2005; Brown and Gerbarg,
many conventional antidepressants (Stevinson and 2001; Conner et al., 2001; Witte et al., 2005; Shinomiya
Ernst, 2000). The only potentially serious adverse et al., 2005b; Jorm et al., 2004a; 2004b; Barnett et al.,
effects are photosensitization, which is extremely rare, 2001; Cote et al., 2004; Abraham et al., 2004; Geier
and the induction of manic symptoms in predisposed and Konstantinowicz, 2004; Feltenstein et al., 2003;
patients (Schulz, 2000). In recent years, multiple case Wheatley, 2001a; 2001b; Singh and Singh, 2002; Ernst,
reports and clinical studies in herb–drug interactions 2002; Smith et al., 2001; Beaubrun and Gray, 2000; Pittler
have been published (Izzo, 2004). In addition, several and Ernst, 2000). Thompson et al. (2004) investigated
preclinical reports have demonstrated that many the acute effects of the herbal anxiolytic Kava-kava on
bioactive constituents of HP-extracts can modulate the emotional reactivity and cognitive performance through
activities of hepatic drug metabolizing enzymes (Singh, a double-blind randomized placebo-controlled trial
2005, Muller et al., 2004b; Mills et al., 2004; Fugh-Berman involving healthy volunteers. The results of this study
and Ernst, 2001; Izzo, 2004, and that hyperforin is a indicate that the mood-elevating effects of Kava were
very potent modulator of a therapeutically important most prominent in trait cheerful subjects, indicating that
drug transport protein, which is encoded by the multi- trait cheerfulness moderated the drug-induced increase
ple drug resistance-gene MDR-1 (Bodo et al., 2003). in cheerful mood. Furthermore, Kava improved the
There is consistent evidence that clinically relevant drug accuracy and the speed of performing the partial
interactions may occur when St John’s wort is co- report and the item recognition task, indicative of a
administered with other drugs, in particular with agents beneficial effect of the phytopharmacon on visual
Copyright © 2006 John Wiley & Sons, Ltd. Phytother. Res. 20, 1023–1035 (2006)
DOI: 10.1002/ptr
POTENTIAL MEDICINAL PLANTS FOR CNS DISORDERS 1027

attention and short-term memory retrieval, respectively. authors also stated that liver function changes in users
Thus, unlike conventional benzodiazepine-type anxi- of the aqueous kava extracts at moderate levels of con-
olytics, which tend to impair cognitive performance and sumption appear to be reversible and begin to return
to increase the occurrence of negative affective states, to baseline after 1 to 2 weeks abstinence from kava. No
Kava is a potent anxiolytic agent, which, additionally, evidence for irreversible liver damage has been found
can facilitate cognitive functioning and can increase (Clough et al., 2003).
positive affectivity related to exhilaration. Toxicological and clinical studies have shown that
In a 1996 randomized placebo-controlled double-blind kava extracts are virtually devoid of toxic effects with
study, two groups of 29 patients were treated with a the exception of the rare hepatotoxic side effects re-
standardized special extract made from kava rhizome ported in few patients (Teschke et al., 2003). In recent
or placebo. The dose was 100 mg of dry extract stand- years, kava kava has been implicated in a number of
ardized to 70 mg kava lactones three times daily for liver failure cases. Ever since this has kept the scientific
4 weeks. The study concluded that kava was clinically world busy. Even though, on closer inspection, the
effective in the reduction of states of anxiety, tension majority of the case reports are probably not connected
and excitedness of non-mental origin. No adverse re- to kava intake, the hepatotoxic effects of kava cannot
actions were noted (Lehmann et al., 1996). Kava con- generally be ruled out (Anke and Ramzan, 2004a). In
tinues to occupy a central place in South Pacific spite of all these hypotheses, there is still no satisfac-
communities, and its importance there recently has tory answer. In any case, further studies, that might
increased with a surge of interest in the ethnic culture hopefully restore the reputation of kava, are required.
and customs of native people. Kava products were sold However, a recommendation has already been made
in the USA in many health food stores, Polynesian by Natural Standard Research Collaboration, Cam-
grocery stores, by mail order and also in mass market bridge, MA, USA to consolidate and analyse available
outlets. Because of its reputed lack of addictive proper- reports and to continue postmarket surveillance in an
ties, legal availability and claims by various authors (it international repository to prevent duplication and to
induces ‘deep restful sleep and clear, epic-length dreams’ promote the collection of thorough details at the time
(Tierra, 1990), ‘enhances psychic powers and visions’ of each report so that any association with kava is clearly
(Cunningham, 1991) and ‘relieves insomnia and ner- defined (Ulbricht et al., 2005).
vousness’ (Santillos, 1991) and because earlier it has Wheatley (2005) demonstrated that Kava-kava is
been recognized by European health authorities as a a well-established hypnotic drug, with a rapid onset
relatively safe remedy for anxiety (Blumental et al., of effect, adequate duration of action and minimal
1998), kava had rapidly gained popularity. The present morning after-effects. However, reports of serious
interest and widespread use of herbal medicines has hepatotoxicity with this preparation have led to it
created the possibility of interaction between them and being banned in most countries worldwide. In his
pharmaceutical drugs if they are used concomitantly. personal trial (not double-blind) in stress-induced
Before the recent reports of apparent hepatotoxicity insomnia, kava and valerian improved sleep and the
associated with the use of Kava, it was one of the top ill-effects of stress, and the combination of the two was
10 selling herbal remedies in Europe and North America even more effective for the control of insomnia. The
(Singh, 2005). This adverse effect had not been pre- author concluded there is a need for longer-term con-
viously encountered with the traditional beverage trolled studies.
prepared as a water infusion in contrast to the com- During the early 1950s some concentrated efforts
mercial products extracted with organic solvents. were made to evaluate the antiepileptic-like efficacy of
Kavalactones, the active principles in kava, are potent pure methysticine and one of its synthetic analogs,
inhibitors of several of the CYP 450 enzymes, sug- ethysticine. The results of clinical trials conducted
gesting a high potential for causing pharmacokinetic during these efforts demonstrated their efficacy only
interactions with drugs and other herbs which are after a very high or near toxic dose, and thus no further
metabolized by the same CYP 450 enzymes (Singh, efforts were made to develop it as a drug. However,
2005; Anke and Ramzan, 2004b; Zou et al., 2004). later extensive efforts (supported by ADD project of
Furthermore, some kavalactones have been shown to NIMH, USA) to obtain a therapeutically useful
possess pharmacological effects, such as blockade of kavalactone derivative led to the identification of
GABA receptors and sodium and calcium ion chan- losigamone as a potential antiepileptic drug (Chatterjee
nels, which may lead to pharmacodynamic interac- and Nöldner, 1997). Its efficacy and safety as an
tions with other substances which possess similar add-on therapy for drug resistant epilepsy have been
pharmacological properties (Singh, 2005). Hu et al. demonstrated in several clinical trials, and its pre-
(2005) showed that Kava enhanced the hypnotic effect clinical activity profile revealed that in addition to
of alcohol in mice, but this was not observed in anticonvulsant activity it possessed a broad range of
humans. The authors further suggested that herbs should therapeutically interesting CNS function modulating
be appropriately labeled to alert consumers to poten- properties. Despite extensive efforts, no definitive
tial interactions when concomitantly used with drugs, statements can yet be made on its mode of action,
and to recommend a consultation with their general which is also the case for the neuroactive kavalactones
practitioners and other medical carers. In fact, hepatic (Chatterjee and Nöldner, 1997). Therefore, it seems
toxicity from manufactured herbal remedies that con- reasonable to suggest that further concentrated
tain kava lactones has been reported in Europe, North efforts to understand the proper therapeutic use and
America and Australia (Clough et al., 2003). There is mode of actions of kava-lactones, can not only lead
no evidence for serious liver damage in kava-using to novel drugs, but also to urgently needed novel
populations in Pacific Island societies or in indigenous pharmacological targets potentially useful for drug
Australians who have used aqueous kava extracts. The discovery purposes.
Copyright © 2006 John Wiley & Sons, Ltd. Phytother. Res. 20, 1023–1035 (2006)
DOI: 10.1002/ptr
1028 V. KUMAR

limited as they are all relevant to uncontrolled trials,


VALERIANA OFFICINALIS conducted with a variety of valerian preparations.
Reference is here made for historical reason to the
Valeriana officinalis L. (Valerian) is a perennial her- paper of Schultz and Mueller (1960), where the reports
baceous plant, belonging to the Valerianaceae family. of different valerian investigators are reviewed. A
Etymologically, Valeriana derives from the Latin word double-blind clinical study, comparing a valerian
‘Valere’ (well being); ‘officinalis’ is a term indicating aqueous extract, placebo and a commercial prepara-
the pharmaceutical use of the plant. Valerian has been tion containing extracts of valerian and hop flowers,
used therapeutically since the Greek and Roman times. showed that valerian produced a significant increase in
Discorides in the first century AD described its use as sleep quality. The improvement in sleep quality was
a mild sedative. V. officinalis is a plant widely distrib- most notable among people who considered themselves
uted in Europe and Asia; nowadays, for medicinal use, poor and irregular sleepers (Leathwood et al., 1982;
it is largely cultivated in Europe. Complementary thera- Leathwood and Chauffard, 1985). Another double-blind
pies are becoming increasingly popular, particularly for study (Lindahl and Lindwall, 1989) was carried out on
symptoms such as sleep disturbance. The herb valerian a valerian preparation standardized for sesquiterpenes.
may be useful as a mild sleep aid in the clinical popu- Volunteers with self-reported sleep difficulties were
lation, such as for persons with rheumatoid arthritis selected. The study showed significant improvement
(Taibi et al., 2004). in subjective sleep quality, 44% reporting perfect sleep
In Unani, Ayurvedic and Homoeopathic systems of and 89% reporting improved sleep with the prepara-
medicine, it is used as a cardiotonic, sedative, stimulat- tion. Other clinical trials and animal studies also
ing, cure for epilepsy, hysteria and convulsive affec- established the effect of valerian on improving sleep
tions, in increased heart palpitation, in intestinal colic, quality and its role as a sedative (Shinomiya et al., 2005a;
for late scanty menses and as an antiseptic. Valerian Fernandez et al., 2004; Oliva et al., 2004; Trevena, 2004;
has been used in China for over 1000 years for similar Diaper and Hindmarch, 2004; Hadley and Petry,
purposes. The essential oils in valerian appear to pro- 2003; Hallam et al., 2003; Ziegler et al., 2002; Cropley
vide its sedative activity, while the valepotriates exert a et al., 2002; Poyares et al., 2002; Larzelere and Wiseman,
regulatory effect on the autonomic nervous system. 2002; Andreatini et al., 2002; Krystal and Ressler, 2001;
Although more than 150 constituents have been identi- Wheatley, 2001a; Fussel et al., 2000; Dominguez et al.,
fied, none appear to be solely responsible for valerian’s 2000; Donath et al., 2000).
effects, suggesting that many compounds may act Valerian is on the FDA’s Generally Recognized
synergistically (Houghton, 1999; Hendriks et al., 1981). as Safe (GRAS) list. The results of a recent phase I
Despite the well-known sedative properties of clinical study show that valerian is a safe herb with
valerian, little information exists on the activity of the respect to effects on human male sterility when admin-
different GABA receptors and their modulatory sites. istered at dose levels corresponding to approximately
The total extract of valerian, as well as the isolated three times the human daily dose (Mkrtchyan et al.,
sesquiterpene alcohols and ketones and valepotriates 2005). Many studies reported in this review confirm the
were reported to induce a significant displacement of validity of valerian preparations, in spite of the doubts
fluorodiazepam in the guinea-pig brain (Thies and raised in the past.
Funke, 1966). A slight inhibition of 3H-flunitrazepam
binding to benzodiazepine-like immunoreactivity in the
TLC of the extract fractions was observed, thus sug-
gesting that the binding inhibition could be due to in- BACOPA MONNIERA
terfering substances in the total extract (Medina et al.,
1989). A recent study indicates that valerian and valeric Though pharmaceutical companies continue to invest
acid are new partial agonists of the 5-HT (5a) receptor enormous resources in identifying agents that could
(Dietz et al., 2005). Another in vitro study showed that be used to alleviate debilitating disorders and retard
valerian had binding affinity at the melatonin (ML 1 mental deterioration afflicting numerous people around
and ML 2) and serotonin (5-HT4e, 5-HT6 and 5-HT7) the world, a source of potentially beneficial agents,
receptor subtypes (Abourashed et al., 2004). A few stud- namely phytochemicals, would appear to have signifi-
ies also established valerian’s role through GABA cant benefits that have yet to be fully exploited (Russo
mediation (Yuan et al., 2004; Santos et al., 1994c). and Borrelli, 2005). Bacopa monniera (BM) in India is
Schellenberg et al. (2004) indicated in a clinical trial locally known as Brahmi or Jalanimba (Chopra et al.,
that valerian extract acts via a central adenosine mecha- 1956). The name Brahmi is derived from the word
nism which is possibly the reason for its sleep-inducing ‘Brama’, the mythical ‘creator’ in the Hindu pantheon.
and maintaining activity. Because the brain is the centre for creative activity,
The clinical use of valerian as a sedative is well known any compound that improves the brain health is
and widespread. The drug is used in the composition of called Brahmi. ‘Brahmi’ which also means ‘bringing
many proprietary medicinal products in the form of its knowledge of the Supreme Reality’ and it has long been
galenic preparations, mostly as an extract and tincture. used there medicinally and as an aid to meditation. In
Valerian was used as a sedative in various forms of India, BM is largely treasured as a revitalizing herb
nervous disorders, non-severe hysterical conditions, that strengthens nervous function and memory.
anxiety, neurasthenia, all cases of nervous sensory or BM has been used by Ayurvedic medical practition-
cerebral erethism, menopausal disturbances, nervous ers in India for almost 3000 years and is classified as
gastralgia and infantile anorexy. The drug was usually a medhyarasayana, a drug used to improve memory
administered half an hour before meals (Auster and and intellect (medhya). BM has been mentioned in
Schaefer, 1958). The value of past clinical studies is several ancient Ayurvedic treatises including the Charaka
Copyright © 2006 John Wiley & Sons, Ltd. Phytother. Res. 20, 1023–1035 (2006)
DOI: 10.1002/ptr
POTENTIAL MEDICINAL PLANTS FOR CNS DISORDERS 1029

Samhita (6th century AD), in which it is recommended crude extracts of BM in rats were 1000 mg and 15 g/kg
in formulations for the management of a range of by the intraperitoneal route, respectively (Martis et al.,
mental conditions including anxiety, poor cognition 1992). It has been reported that antiepileptic drugs, such
and lack of concentration, and the Bhavprakash as phenytoin, can result in cognitive impairment (Smith,
Var-Prakarana (16th century AD). In certain parts of 1991). Vohora et al. (2000) reported that BM reversed
India, Brahmi is believed to be an aphrodisiac; in Sri the phenytoin-induced cognitive impairment when
Lanka, under the name of Loonooweella, Brahmi is administered concomitantly with phenytoin which sug-
prescribed for fevers; in the Philippines, it is used as a gests a potential corrective effect of BM extract in
diuretic (Uphof, 1968). phenytoin-induced cognitive deficits. Diverse studies
The BM extracts and isolated bacosides have been indicated that interactions between herbal medicines
investigated extensively for their neuropharmacological and synthetic drugs exist and can have serious conse-
effects and their nootropic action or antiamnestic quences (Izzo and Ernst, 2001). Therefore, it is neces-
effect confirmed (Kishore and Singh, 2005; Anbarasi sary to consider the possibility of BM–drug interaction.
et al., 2005a; 2005b; 2005c; Jorm et al., 2004; Achliya The mechanism of action behind various reported
et al., 2004; Nathan et al., 2004; Sumathi et al., 2002a; preclinical studies indicating a cognitive enhancing ef-
2002b; Das et al., 2002; Nathan et al., 2001; Kidd, 1999; fect is still uncertain, as its multiple active constituents
Malhotra and Das, 1959; Singh and Dhawan, 1982; Singh make its pharmacology complex. In light of the many
and Dhawan, 1992; Singh and Dhawan, 1997). Singh reports showing important activities of BM extracts or
et al. (1988) suggested that bacosides induce membrane bacosides, further research is required to ascertain the
dephosphorylation, with a concomitant increase in pro- findings mentioned in this review.
tein and RNA turnover in specific brain areas. The other
proposal is that BM enhances protein kinsae activity
in the hippocampus which may also contribute to its
nootropic action (Singh and Dhawan, 1997). Loss of CONVOLVULUS PLURICAULIS
cholinergic neuronal activity in the hippocampus is the
primary feature of Alzheimer’s disease (Enz et al., 1993). Convolvulus pluricaulis (CP) is also known as
A team of other researchers reported that a stand- Shankhpushpi (or shankapushpi), is an herb that has
ardized bacosides-rich extract of BM, reversed the been used in India for hundreds of years for nervous
cognitive deficits induced by intracerebroventricularly disorders such as stress, anxiety and insomnia. It pro-
administered colchicines and injection of ibotenic acid duces a feeling of peace and calm, reduces stress, anxi-
into the nucleus basalis magnocellularis (Bhattacharya ety and mental fatigue. Shankhpushpi is a morning-glory
et al., 1999). In the same study, BM also reversed the like perennial that grows on the plains of India. It has
depletion of acetylcholine, the reduction in choline been widely used in Ayurvedic medicine to treat ner-
acetylase activity and the decrease in muscarinic vous disorders, in the same way that kava-kava and
cholinergic receptor binding in the frontal cortex and valerian are prescribed by American herbalists. It is
hippocampus. only recently that Shankhpushpi has been brought to
BM extract or bacosides have also shown anxiolytic American stores for medicinal use. Herbalists believe
effects (Bhattacharya and Ghosal, 1998b; Shankar and that Shankhpushpi calms the nerves by regulating the
Singh, 2000), antidepressant activity (Sairam et al., 2002), body’s production of the stress hormones, adrenaline
anticonvulsive action (Shanmugasundaram et al., 1991; and cortisol.
Martis and Rao, 1992; Ganguly and Malhotra, 1967), In Ayurvedic medicine, it is also believed that
antioxidant activity (Singh et al., 2006; Bafna and Shankhpushpi is an anti-aging remedy called Rasayan.
Balaraman, 2005; Rohini et al., 2004; Russo et al., 2003a; Even though Ayurvedic practitioners have used
2003b; Sumathy et al., 2001; Pawar et al., 2001; Shankhpushpi for centuries, there is no hard scientific
Bhattacharya et al., 2000c; Tripathi et al., 1996) anti- evidence as to the positive effects of this herb, beside
stress (Rai et al., 2003; Chowdhuri et al., 2002) a few Indian studies performed in the 1970s and
and antiulcerogenic activity (Dorababu et al., 2004; 1980s and most of them were published locally. In those
Jain et al., 1994; Rao et al., 2000; Sairam et al., 2001; studies, people suffering from anxiety were given
Goel et al., 2003). Singh et al. (1996) suggest an involve- Shankhpushpi for 6 weeks and claimed to have slept
ment of the GABA-ergic system in the mediation of better, have more energy and better concentration. In
central nervous system effects of BM. one of these studies, published in an Indian Medical
Various clinical studies have also been carried out to Journal in 1982, researchers gave 28 people diagnosed
establish the efficacy of BM in memory and attention with anxiety 50 mg daily of an herbal formula with
disorders (Maher et al., 2002; Roodenrys et al., 2002; shankhpushpi as a primary ingredient. After 6 weeks
Stough et al., 2001; Dave et al., 1993; Sharma et al., of treatment, 91% of the patients had more energy
1987; Singh and Singh, 1980; Ghosh and Kar, 1966; and 60% to 70% could sleep and concentrate better
Mukherjee and Dey, 1966). In view of the positive (Shaughnessy, 2002). Today this herb is still the pre-
results of such clinical trials, BM has been introduced ferred method for reducing symptoms associated with
in the Indian market and in other countries, alone or in anxiety, panic attacks, nervousness and insomnia. The
combination with other phytocomplexes, and utilized leaves of Shankhpushpi are used to treat chronic
in the treatment of memory and attention disorders bronchitis and asthma. The root is used for childhood
(Shukia et al., 1987). fever, and the oil stimulates the growth of hair. Using
BM has been found to be well tolerated and without the whole plant in the form of a decoction with cumin
any untoward reaction or side effects in many regula- and milk is used to treat fever, debility, memory loss,
tory pharmacological and toxicological studies (Russo syphilis and scrofula. Ganju et al. (2003) has showed
and Borrelli, 2005). The LD50 of aqueous and alcohol that Shankhpushpi has immunomodulatory effects.
Copyright © 2006 John Wiley & Sons, Ltd. Phytother. Res. 20, 1023–1035 (2006)
DOI: 10.1002/ptr
1030 V. KUMAR

Another study indicated that CP has an antiulcerogenic strated, reports on attempts to identify and develop a
effect due to augmentation of mucosal defensive fac- better reserpine-like agent have been rare. Concentrated
tors such as mucin secretion, lifespan of mucosal cells efforts made to obtain a better chlorpromazine-like drug
and glycoproteins rather than on the offensive factors by structure activity studies led, not only to many still
such as acid-pepsin (Sairam et al., 2001c). therapeutically used antipsychotics, but also to the first
CP shows promise as a safe, effective remedy for generation of modern antidepressants. Reports on
anxiety, but controlled human studies are needed attempts to determine whether similar concentrated
to establish scientifically its efficacy in various CNS efforts to better clarify the structure–activity relation-
disorders with special emphasis on memory enhancing ship with reserpine could have analogous therapeutic
properties. Indurwade and Biyani (2000) found that impacts, have been rare to date.
shankhpushpi has potent depressive action in mice. It is now becoming exceedingly apparent that the
A study indicated that CP has antiepileptic activity available psychotherapeutics do not properly meet the
and researchers further investigated the CP interaction therapeutic demands of the vast majority of patients
with phenytoin from both pharmacokinetic (serum with mental health problems, and that herbal remedies
levels) and pharmacodynamic (electroshock seizure remain the ultimate therapeutic hope for many such
prevention) aspects. They advised that clinical combina- patients in the Western world and elsewhere. Critical
tion of CP with phenytoin should be avoided (Dandekar analysis of our current understanding of the most popu-
et al., 1992). lar, and most well studied, CNS active medicinal plants
reveal that many therapy relevant questions have not
yet been properly answered for any of them. However,
for many (or most) of them clinical efficacy has been
CONCLUSIONS AND PERSPECTIVES demonstrated repeatedly in properly controlled clinical
trials. Consequently, therefore, many phyto-pharmaceu-
Although the therapeutic possibilities offered by herbs tical laboratories are now concentrating their efforts on
for diverse types of CNS disorders have been known to identifying the active constituents and modes of actions
mankind for centuries, until now little concentrated of these herbs. The ultimate goal of most such efforts
effort has been made to define and understand their has been to obtain a patentable, or better therapeuti-
most appropriate therapeutic uses, or to exploit them cally useful, or better standardized extract. However,
for identifying and developing CNS active drugs. As until now, very little attention has been paid to the
can be inferred from the facts mentioned in the intro- development of structurally and/or functionally novel
duction, researchers interested in CNS active drug dis- CNS active drugs from psychoactive medicinal plants.
covery projects have been, until recently, very negligent The available information summarized in this review
in using existing knowledge on herbal remedies as the strongly suggests that such a situation could be ham-
starting point for their endeavours. Such a situation pering more rapid progress of CNS active drug discov-
is surprising because several modern therapeutically ery projects.
interesting concepts of modern pharmacology, in gen-
eral, and psychopharmacology in particular, could be
conceived through studies and efforts made to better Acknowledgements
understand the traditionally known herbal remedies
only. Hereupon studies conducted with reserpine (an
active principle of Raulwolfia serpentina), amphetamine The author would like to dedicate this article to his PhD mentors Dr
P. N. Singh, Reader, Department of Pharmaceutics, Institute of Tech-
(a congener of ephedrine, an active component of the nology, Banaras Hindu University (BHU), India and late Professor
Chinese herbal agent ma huang) and with many other S. K. Bhattacharya, Department of Pharmacology, Institute of Medi-
well known psychoactive secondary plant metabolites cal Sciences, BHU. The author is grateful to his postdoc mentor
played a crucial role. Thus, although the clinical anti- Dr Jochen Klein, Associate Professor, Department of Pharmaceuti-
cal Sciences, Texas Tech School of Pharmacy, Amarillo, TX, USA
psychotic effects of reserpine were described during and Dr Shyam S Chatterjee, Former Head, Section of Pharmacology,
the early 1950s, i.e. the same period during which the Dr. Willmar Schwabe, Arzneimittel, Karlsruhe D-76207, Germany
therapeutic efficacy of chlorpromazine was first demon- for their critical review and constructive comments.

REFERENCES

Abourashed EA, Koetter U, Brattstrom A. 2004. In vitro binding Anbarasi K, Vani G, Balakrishna K, Devi CS. 2005a. Effect of
experiments with a Valerian, hops and their fixed combina- bacoside A on membrane-bound ATPases in the brain of
tion extract (Ze91019) to selected central nervous system rats exposed to cigarette smoke. J Biochem Mol Toxicol 19:
receptors. Phytomedicine 11: 633 – 638. 59 – 65.
Abraham KC, Connor KM, Davidson JR. 2004. Explanatory Anbarasi K, Vani G, Balakrishna K, Devi CS. 2005b. Creatine
attributions of anxiety and recovery in a study of kava. kinase isoenzyme patterns upon chronic exposure to ciga-
J Altern Complement Med 10: 556 – 559. rette smoke: protective effect of bacoside A. Vascul
Achliya GS, Wadodkar SG, Dorle AK. 2004. Evaluation of seda- Pharmacol 42: 57–61.
tive and anticonvulsant activities of Unmadnashak Ghrita. Anbarasi K, Vani G, Devi CS. 2005c. Protective effect of bacoside
J Ethnopharmacol 94: 77– 83. A on cigarette smoking-induced brain mitochondrial
Ahlemeyer B, Krieglstein J. 2003a. Neuroprotective effects of dysfunction in rats. J Environ Pathol Toxicol Oncol 24: 225 –
Ginkgo biloba extract. Cell Mol Life Sci 60: 1779 –1792. 234.
Ahlemeyer B, Krieglstein J. 2003b. Pharmacological studies Andreatini R, Sartori VA, Seabra ML, Leite JR. 2002. Effect of
supporting the therapeutic use of Ginkgo biloba extract valepotriates (valerian extract) in generalized anxiety disor-
for Alzheimer’s disease. Pharmacopsychiatry 36: 8 – der: a randomized placebo-controlled pilot study. Phytother
14. Res 16: 650 – 654.

Copyright © 2006 John Wiley & Sons, Ltd. Phytother. Res. 20, 1023–1035 (2006)
DOI: 10.1002/ptr
POTENTIAL MEDICINAL PLANTS FOR CNS DISORDERS 1031

Andrieu S, Gillette S, Amouyal K et al. 2003. Association of Chatterjee SS, Bhattacharya SK, Wonnemann M, Singer A,
Alzheimer’s disease onset with Ginkgo biloba and other Muller WE. 1998a. Hyperforin as a possible antidepressant
symptomatic cognitive treatments in a population of women component of Hypericum extracts. Life Sci 63: 499 – 510.
aged 75 years and older from the EPIDOS study. J Gerontol Chatterjee SS, Doelman CJ, Noldner M, Biber A, Koch E. 2005.
A Biol Sci Med Sci 58: 372 – 377. Influence of the Ginkgo extract EGb 761 on rat liver
Anke J, Ramzan I. 2004a. Kava hepatotoxicity: Are we any closer cytochrome P450 and steroid metabolism and excretion in
to the truth? Planta Med 70: 193 –196. rats and man. J Pharm Pharmacol 57: 641–650.
Anke J, Ramzan I. 2004b. Pharmacokinetic and pharmaco- Chatterjee SS, Kondratskaya EL, Krishtal OA. 2003. Structure-
dynamic drug interactions with Kava (Piper methysticum activity studies with Ginkgo biloba extract constituents as
Forst. f.). J Ethnopharmacol 93: 153 –160. receptor-gated chloride channel blockers and modulators.
Auster F, Schaefer J. 1958. Valeriana officinalis L., Arzneipflan- Pharmacopsychiatry 36: S68 – S77.
zen. G. Thieme: Lepzig. Chatterjee SS, Noldner M, Koch E, Erdelmeier C. 1998b. Anti-
Bafna PA, Balaraman R. 2005. Antioxidant activity of DHC-1, an depressant activity of Hypericum perforatum and hyperforin:
herbal formulation, in experimentally-induced cardiac and the neglected possibility. Pharmacopsychiatry 31: 7–15.
renal damage. Phytother Res 19: 216 –221. Chatterjee SS, Nöldner M. 1997. Losigamone: From plant ex-
Barnett SD, Connor KM, Davidson JR. 2001. The self-assess- tract to antiepileptic drug. CNS Drug Rev 3: 225–244.
ment of resilience and anxiety: psychometric properties. Chopra RN, Nayar SL, Chopra IC. 1956. Glossary of Indian
CNS Spectr 10: 854 – 857. Medicinal Plants. Council of Scientific and Industry Research:
Bastianetto S, Ramassamy C, Dore S, Christen Y, Poirier J, New Delhi, 32.
Quirion R. 2000. The Ginkgo biloba extract (EGb 761) pro- Chowdhuri DK, Parmar D, Kakkar P, Shukla R, Seth PK, Srimal
tects hippocampal neurons against cell death induced by RC. 2002. Antistress effects of bacosides of Bacopa monnieri:
beta-amyloid. Eur J Neurosci 12: 1882 –1890. modulation of Hsp70 expression, superoxide dismutase and
Beaubrun G, Gray GE. 2000. A review of herbal medicines for cytochrome P450 activity in rat brain. Phytother Res 16:
psychiatric disorders. Psychiatr Serv 51: 1130 –1134. 639 – 645.
Bhattacharya SK, Bhattacharya A, Kumar A, Ghosal S. 2000c. Clough AR, Bailie RS, Currie B. 2003. Liver function test abnor-
Antioxidant activity of Bacopa monniera in rat frontal cor- malities in users of aqueous kava extracts. J Toxicol Clin
tex, striatum and hippocampus. Phytother Res 14: 174 –179. Toxicol 41: 821– 829.
Bhattacharya SK, Chakrabarti A, Chatterjee SS. 1998a. Activity Colciaghi F, Borroni B, Zimmermann M et al. 2004. Amyloid
profiles of two hyperforin-containing Hypericum extracts in precursor protein metabolism is regulated toward alpha-
behavioural models. Pharmacopsychiatry 31: 22–29. secretase pathway by Ginkgo biloba extracts. Neurobiol Dis
Bhattacharya SK, Ghosal S. 1998b. Anxiolytic activity of a stand- 16: 454 – 460.
ardized extract of Bacopa monniera: an experimental study. Connor KM, Davidson JR, Churchill LE. 2001. Adverse-effect
Phytomedicine 5: 77–82. profile of kava. CNS Spectr 6: 850 – 853.
Bhattacharya SK, Kumar A, Ghosal S. 1999. Effect of Bacopa Cote CS, Kor C, Cohen J, Auclair K. 2004. Composition and
monniera on animal models of Alzheimer’s disease and biological activity of traditional and commercial kava ex-
perturbed central cholinergic markers of cognition in rats. tracts. Biochem Biophys Res Commun 10: 147–152.
In Molecular Aspects of Asian Medicines, Siva Sankar DV Cragg GM, Newman DJ, Sanders KM. 1997. Natural products in
(ed.). PJD Publications: New York. drug discovery and development. J Nat Prod 60: 52 – 60.
Biber A, Fischer H, Romer A, Chatterjee SS. 1998. Oral Cropley M, Cave Z, Ellis J, Middleton RW. 2002. Effect of kava
bioavailability of hyperforin from Hypericum extracts in rats and valerian on human physiological and psychological
and human volunteers. Pharmacopsychiatry 31: 36 – 43. responses to mental stress assessed under laboratory con-
Birks J, Grimley EV, Van Dongen M. 2002. Ginkgo biloba for ditions. Phytother Res 16: 23 –27.
cognitive impairment and dementia. Cochrane Database Cunningham S. 1991. Encyclopedia of Magical Herbs. Lewellyn
Syst Rev 4: CD003120. Publications: St Paul, 133.
Blumenthal M, Busse WR, Goldberg A et al. (eds), Klein S and Dandekar UP, Chandra RS, Dalvi SS et al. 1992. Analysis of
Rister RS, trans. 1998. The Complete German Commission a clinically important interaction between phenytoin and
E Monographs. American Botanical Council: Austin, TX, 156. Shankhapushpi, an Ayurvedic preparation. J Ethnopharma-
Bodo A, Bakos E, Szeri F. 2003. The role of multidrug transport- col 35: 285–288.
ers in drug availability, metabolism and toxicity. Toxicol Daniel K. 1935. Johanniskraut (Hypericum perforatum) bei
Lett 141: 133 –143. psychische Störungen. Hippokrates 10: 929 – 932.
Bombardelli E, Morazzoni P. 1995. Hypericum perforatum. Das A, Shanker G, Nath C, Pal R, Singh S, Singh H. 2002. A
Fitoterapia 66: 43 – 68. comparative study in rodents of standardized extracts of
Boonkaew T, Camper ND. 2005. Biological activities of Ginkgo Bacopa monniera and Ginkgo biloba: anticholinesterase and
extracts. Phytomedicine 12: 318 –323. cognitive enhancing activities. Pharmacol Biochem Behav
Boralle E, Braquet P, Gottlieb. 1988. A review of its chemical 73: 893–900.
composition. In Ginkgolides Chemistry, Biology, Pharma- Dave UP, Chauvan V, Dalvi J. 1993. Evaluation of BR-16 A
cology and Clinical Perspective. Vol 1. JP Process Science (Mentat) in cognitive and behavioural dysfunction of men-
Publishers: Barcelona, Spain, 9–25. tally retarded children: a placebo-controlled study. Indian J
Brown RP, Gerbarg PL. 2001. Herbs and nutrients in the treat- Pediatr 60: 423–428.
ment of depression, anxiety, insomnia, migraine, and Diaper A, Hindmarch I. 2004. A double-blind, placebo-controlled
obesity. J Psychiatr Pract 7: 75 – 91. investigation of the effects of two doses of a valerian pre-
Buchholzer ML, Dvorak C, Chatterjee SS, Klein J. 2002. Dual paration on the sleep, cognitive and psychomotor function
modulation of striatal acetylcholine release by hyperforin, a of sleep-disturbed older adults. Phytother Res 18: 831–
constituent of St. John’s wort. J Pharmacol Exp Ther 301: 836.
714 –719. Dietz BM, Mahady GB, Pauli GF, Farnsworth NR. 2005. Valerian
Butterweck V, Christoffel V, Nahrstedt A, Petereit F, Spengler B, extract and valerenic acid are partial agonists of the 5-HT(5a)
Winterhoff H. 2003. Step by step removal of hyperforin and receptor in vitro. Brain Res Mol Brain Res 138: 191–197.
hypericin: activity profile of different Hypericum prepara- Dominguez RA, Bravo-Valverde RL, Kaplowitz BR, Cott JM. 2000.
tions in behavioral models. Life Sci 73: 627– 639. Valerian as a hypnotic for Hispanic patients. Cultur Divers
Capasso A, Sorrentino L. 2005. Pharmacological studies on the Ethnic Minor Psychol 6: 84 – 92.
sedative and hypnotic effect of Kava kava and Passiflora Donath F, Quispe S, Diefenbach K, Maurer A, Fietze I, Roots I.
extracts combination. Phytomedicine 12: 39–45. 2000. Critical evaluation of the effect of valerian extract on
Carrigan D. 1995. Fossil medicine – Ginkgo biloba. Eur J Herbal sleep structure and sleep quality. Pharmacopsychiatry 33:
Med 1: 16 –18. 47–53.
Cervo L, Rozio M, Ekalle-Soppo CB, Guiso G, Morazzoni P, Caccia Dorababu M, Prabha T, Priyambada S, Agrawal VK, Aryya NC,
S. 2002. Role of hyperforin in the antidepressant-like activ- Goel RK. 2004. Effect of Bacopa monniera and Azadirachta
ity of Hypericum perforatum extracts. Psychopharmacol indica on gastric ulceration and healing in experimental
(Berl) 164: 423–428. NIDDM rats. Indian J Exp Biol 42: 389–397.
Challem J. 2001. St. John’s wort vs drugs. Nutr Sci News 6: Drewe J. 2000. Mechanisen der Interaktionen mit Johanniskraut
212–218. [Abstract]. In Phytopharmaka VI Forschung und Klinsche

Copyright © 2006 John Wiley & Sons, Ltd. Phytother. Res. 20, 1023–1035 (2006)
DOI: 10.1002/ptr
1032 V. KUMAR

Anwendung. Deutsche Gesellschaft fur Klinische Pharmako- Izzo AA. 2004. Drug interactions with St. John’s Wort (Hypericum
logie und Therapie: Berlin. perforatum): a review of the clinical evidence. Int J Clin
Eckert GP, Keller JH, Jourdan C et al. 2004. Hyperforin modifies Pharmacol Ther 42: 139 –148.
neuronal membrane properties in vivo. Neurosci Lett 367: Izzo AA, Ernst E. 2001. Interactions between herbal medicines
139 –143. and prescribed drugs: a systematic review. Drugs 61: 2163 –
Enz A, Amstutz R, Boddeke H et al. 1993. Brain selective inhibi- 2175.
tion of acetylcholinesterase: a novel approach to therapy Jain P, Khanna NK, Trehan T, Pendse VK, Godhwani JL. 1994.
for Alzheimer’s disease. Prog Brain Res 98: 431– 438. Antiinflammatory effects of an Ayurvedic preparation,
Ernst E (ed.). 2003a. In Hypericum. The genus Hypericum. Brahmi Rasayan, in rodents. Indian J Exp Biol 32: 633–
Medicinal and Aromatic Plants-Industrial Profiles Series, 636.
Hardman R (series ed), Taylor & Francis: London and New Jorm AF, Christensen H, Griffiths KM, Parslow RA, Rodgers B,
York. Blewitt KA. 2004a. Effectiveness of complementary and
Ernst E. 1999. Second thoughts about safety of St John’s wort. self-help treatments for anxiety disorders. Med J Aust 181:
Lancet 354: 2014 – 2016. 529–46.
Ernst E. 2002. The risk-benefit profile of commonly used herbal Jorm AF, Rodgers B, Christensen H. 2004b. Use of medications
therapies: Ginkgo, St. John’s Wort, Ginseng, Echinacea, Saw to enhance memory in a large community sample of 60–64
Palmetto, and Kava. Ann Intern Med 136: 42–53. year olds. Int Psychogeriatr 16: 209–217.
Ernst E. 2003b. Herb-drug interactions: not a clinically relevant Kanowski S, Hoerr R. 2003. Ginkgo biloba extract EGb 761
risk. Focus Alter Complem Ther 8: 189. in dementia: intent-to-treat analyses of a 24-week, multi-
Evans JG, Wilcock G, Birks J. 2004. Evidence-based pharmaco- center, double-blind, placebo-controlled, randomized trial.
therapy of Alzheimer’s disease. Int J Neuropsychopharmacol Pharmacopsychiatry 36: 297–303.
7: 351–369. Käufeler R, Meier B, Brattström A. 2001. Efficacy and tolerabil-
Feltenstein MW, Lambdin LC, Ganzera M et al. 2003. Anxiolytic ity of Ze117 St. John’s wort extract in comparison with
properties of Piper methysticum extract samples and placebo, imipramine and fluoxetine for the treatment of
fractions in the chick social-separation-stress procedure. mild to moderate depression according to ICD-10. An over-
Phytother Res 17: 210–216. view. Pharmacopsychiatry 34: S49–S50.
Fernandez S, Wasowski C, Paladini AC, Marder M. 2004. Seda- Kidd PM. 1999. A review of nutrients and botanicals in the
tive and sleep-enhancing properties of linarin, a flavonoid- integrative management of cognitive dysfunction. Altern
isolated from Valeriana officinalis. Pharmacol Biochem Med Rev 4: 144 –161.
Behav 77: 399 – 404. Kiewert C, Buchholzer ML, Hartmann J, Chatterjee SS, Klein J.
Fugh-Berman A, Ernst E. 2001. Herb-drug interactions: review 2004. Stimulation of hippocampal acetylcholine release by
and assessment of report reliability. Br J Clin Pharmacol hyperforin, a constituent of St. John’s Wort. Neurosci Lett
52: 587– 595. 364: 195–198.
Fussel A, Wolf A, Brattstrom A. 2000. Effect of a fixed valerian- Kishore K, Singh M. 2005. Effect of bacosides, alcoholic extract
Hop extract combination (Ze 91019) on sleep polygraphy in of Bacopa monniera Linn. (brahmi), on experimental amne-
patients with non-organic insomnia: a pilot study. Eur J sia in mice. Indian J Exp Biol 43: 640–645.
Med Res 5: 385 – 390. Kleijnen J, Knipschild P. 1992. Ginkgo biloba for cerebral insuf-
Ganguly DK, Malhotra CL. 1967. Some behavioural effects of ficiency. Br J Clin Pharmacol 34: 352–358.
an active fraction from Herpestis monniera, Linn. (Brahmi). Klein J, Chatterjee SS, Loffelholz K. 1997. Phospholipid break-
Indian J Med Res 55: 473–482. down and choline release under hypoxic conditions: inhibi-
Ganju L, Karan D, Chanda S, Srivastava KK, Sawhney RC, tion by bilobalide, a constituent of Ginkgo biloba. Brain Res
Selvamurthy W. 2003. Immunomodulatory effects of agents 755: 347–350.
of plant origin. Biomed Pharmacother 57: 296 – 300. Klein J, Weichel O, Hilgert M, Rupp J, Chatterjee SS, Nawrath
Gasser US, Gasser T. 2001. Alzheimer dementia. Comparison H. 2003. Excitotoxic hippocampal membrane breakdown
of the effectiveness of cholinesterase inhibitors and gingko. and its inhibition by bilobalide: role of chloride fluxes.
MMW Fortschr Med 143: 40 – 41. Pharmacopsychiatry 36: S78 – 83.
Gaster B, Holroyd J. 2000. St. John’s wort for depression: a Klusa V, Germane S, Noldner M, Chatterjee SS. 2001. Hypericum
systemic review. Arch Int Med 160: 152–156. extract and hyperforin: memory-enhancing properties in
Geier FP, Konstantinowicz T. 2004. Kava treatment in patients rodents. Pharmacopsychiatry 34: S61–S69.
with anxiety. Phytother Res 18: 297–300. Koch E. 2005. Inhibition of platelet activating factor (PAF)-
Gertz HJ, Kiefer M. 2004. Review about Ginkgo biloba special induced aggregation of human thrombocytes by ginkgolides:
extract EGb 761 (Ginkgo). Curr Pharm Des 10: 261–264. considerations on possible bleeding complications after
Ghosh S, Kar SK. 1966. Clinical trial on Brahmi. II. Psycho- oral intake of Ginkgo biloba extracts. Phytomedicine 12:
logical investigation with normals. J Exp Med Sci 10: 10 –16.
12 –13. Kondratskaya EL, Fisyunov AI, Chatterjee SS, Krishtal OA. 2004a.
Goel RK, Sairam K, Babu MD, Tavares IA, Raman A. 2003. In Ginkgolide B preferentially blocks chloride channels formed
vitro evaluation of Bacopa monniera on anti-Helicobacter by heteromeric glycine receptors in hippocampal pyrami-
pylori activity and accumulation of prostaglandins. dal neurons of rat. Brain Res Bull 63: 309–314.
Phytomedicine 10: 523 – 527. Kondratskaya EL, Pankratov YV, Lalo UV, Chatterjee SS, Krishtal
Haan J, Horr R. 2004. Delay in progression of dependency OA. 2004b. Inhibition of hippocampal LTP by ginkgolide B
and need of care of dementia patients treated with Ginkgo is mediated by its blocking action on PAF rather than glycine
special extract EGb 761. Wien Med Wochenschr 154: 511– receptors. Neurochem Int 44: 171–177.
514. Krystal AD, Ressler I. 2001. The use of valerian in neuro-
Hadley S, Petry JJ. 2003. Valerian. Am Fam Physician 67: 1755 – psychiatry. CNS Spectr 6: 841–847.
1758. Kumar V, Jaiswal AK, Singh PN, Bhattacharya SK. 2000a.
Hallam KT, Olver JS, McGrath C, Norman TR. 2003. Compara- Anxiolytic activity of Indian Hypericum perforatum Linn: an
tive cognitive and psychomotor effects of single doses of experimental study. Indian J Exp Biol 38: 36 – 41.
Valeriana officianalis and triazolam in healthy volunteers. Kumar V, Khanna VK, Seth PK, Singh PN, Bhattacharya SK.
Hum Psychopharmacol 18: 619–625. 2002. Brain neurotransmitter receptor binding and nootropic
Hendriks H, Bos R, Allersma DP, Malingre TM, Koster AS. 1981. studies on Indian Hypericum perforatum Linn. Phytother
Pharmacological screening of valerenal and some other Res 16: 210–216.
components of essential oil of Valeriana officinalis. Planta Kumar V, Kiewert C, Klein J. 2005. Hyperforin inhibits NMDA-
Med 42: 62–68. induced edema in hippocampal slices. Poster # T3283
Houghton PJ. 1999. The scientific basis for the reputed activity presented at American Association of Pharmaceutical
of Valerian. J Pharm Pharmacol 51: 505–512. Scientist’s annual meeting and exposition, Nashville, TN,
Hu Z, Yang X, Ho PC et al. 2005. Herb-drug interactions: a USA, November 6 –10.
literature review. Drugs 65: 1239–1282. Kumar V, Mazinarishvili A, Kiewert C, Abbruscato T, Bickel U,
Indurwade NH, Biyani KR. 2000. Evaluation of comparative and Schyf CJ, Klein J. 2006. NMDA receptor-antagonistic
combined depressive effect of Brahmi, Shankhpushpi and properties of hyperforin, a constituent of St. John’s work.
Jatamansi in mice. Indian J Med Sci 54: 339–341. J Pharmacol Sci : In press.

Copyright © 2006 John Wiley & Sons, Ltd. Phytother. Res. 20, 1023–1035 (2006)
DOI: 10.1002/ptr
POTENTIAL MEDICINAL PLANTS FOR CNS DISORDERS 1033

Kumar V, Singh PN, Bhattacharya SK. 2000b. Hypericum mon constituents of plants. Biochem Biophys Res Commun
perforatum: nature’s mood stabilizer. Indian J Exp Biol 38: 165: 547–553.
1077–1085. Michel PF, Hosford D. 1988. Ginkgo biloba: From ‘Living Fossil’
Kumar V, Singh PN, Bhattacharya SK. 2001a. Anti-inflammatory to Modern Therapeutic Agent. In Ginkgolled-Chemistry, Bio-
and analgesic activity of Indian Hypericum perforatum L. logy, Pharmacology and Clinical Perspectives, Braquet P
Indian J Exp Biol 39: 339–343. (ed.), Vol 1. JP Process Publishers: Barcelona, Spain, 1–8.
Kumar V, Singh PN, Bhattacharya SK. 2001b. Anti-stress Mills E, Montori VM, Wu P, Gallicano K, Clarke M, Guyatt G.
activity of Indian Hypericum perforatum Linn. Indian J Exp 2004. Interaction of St John’s wort with conventional drugs:
Biol 39: 344 – 349. systematic review of clinical trials. Br Med J 329: 27–30.
Kumar V, Singh PN, Bhattacharya SK. 2001c. Neurochemical Mix JA, Crews WD Jr. 2002. A double-blind, placebo-controlled,
studies on Indian Hypericum perforatum Linn. Indian J Exp randomized trial of Ginkgo biloba extract EGb 761 in a
Biol 39: 334 – 338. sample of cognitively intact older adults: neuropsychological
Kumar V, Singh PN, Bhattacharya SK. 2003. Neuropsychopharm- findings. Hum Psychopharmacol 17: 267–277.
acological studies on Indian Hypericum perforatum Linn. In Mkrtchyan A, Panosyan V, Panossian A, Wikman G, Wagner H.
Medicinal and Aromatic Plants – Industrial Profile. Volume 2005. A phase I clinical study of Andrographis paniculata
Genus Hypericum, Ernst E (ed.), 1st edn. Taylor & Francis: fixed combination Kan Jang versus ginseng and valerian
London and simultaneously published by Taylor & Francis: on the semen quality of healthy male subjects. Phyto-
New York, USA and Canada, 179–226. medicine 12: 403–409.
Kumar V, Singh PN, Jaiswal AK, Bhattacharya SK. 1999. Anti- Mukherjee GD, Dey CD. 1966. Clinical trial on Brahmi. I. J Exp
depressant activity of Indian Hypericum perforatum Linn in Med Sci 10: 5 –11.
rodents. Indian J Exp Biol 37: 1171–1176. Muller T, Mannel M, Murck H, Rahlfs VW. 2004b. Treatment of
Kumar V, Singh PN, Muruganandam AV, Bhattacharya SK. somatoform disorders with St. John’s wort: a randomized,
2000c. Effect of Indian Hypericum perforatum Linn on double-blind and placebo-controlled trial. Psychosom Med
animal models of cognitive dysfunction. J Ethnopharmacol 66: 538 – 547.
72: 119 –128. Muller WE. 2003. Current St John’s wort research from mode
Kurz A, Van Baelen B. 2004. Ginkgo biloba compared with of action to clinical efficacy. Pharmacol Res 47: 101–109.
cholinesterase inhibitors in the treatment of dementia: a Muller WE (ed.). 2005. In St. John’s Wort and its Active Princi-
review based on meta-analyses by the Cochrane collabora- ples in Depression and Anxiety. Milestones in Drug Therapy
tion. Dement Geriatr Cogn Disord 18: 217–226. Series, Parnham MJ, Bruinvels J (series eds). Springer:
Laakmann G, Schule C, Baghai T, Kieser M. 1998. St. John’s Germany.
wort in mild to moderate depression: the relevance of Muller WE, Chatterjee SS. 2003. Cognitive and other behavioral
hyperforin for the clinical efficacy. Pharmacopsychiatry 31: effects of EGb 761 in animal models. Pharmacopsychiatry
54–59. 36: 24 –31.
Larzelere MM, Wiseman P. 2002. Anxiety, depression, and in- Muller WE, Singer A, Wonnemann M, Hafner U, Rolli M, Schafer
somnia. Prim Care 29: 339 – 360. C. 1998. Hyperforin represents the neurotransmitter reuptake
Le Bars PL. 2003. Response patterns of EGb 761 in Alzheimer’s inhibiting constituent of Hypericum extract. Pharmaco-
disease: influence of neuropsychological profiles. Pharmaco- psychiatry 31: 16 –21.
psychiatry 36: 50–55. Nakanishi K. 2005. Terpene trilactones from Gingko biloba: From
Leathwood PD, Chauffard F. 1985. Aqueous extract of valerian ancient times to the 21st century. Bioorg Med Chem 13:
reduces latency to fall asleep in man. Planta Med 4: 144 – 4987– 5000.
148. Nathan PJ, Clarke J, Lloyd J, Hutchison CW, Downey L, Stough
Leathwood PD, Chauffard F, Heck E, Munoz-Box R. 1982. C. 2001. The acute effects of an extract of Bacopa monniera
Aqueous extract of valerian root (Valeriana officinalis L.) (Brahmi) on cognitive function in healthy normal subjects.
improves sleep quality in man. Pharmacol Biochem Behav Hum Psychopharmacol 16: 345 – 351.
17: 65–71. Nathan PJ, Ricketts E, Wesnes K, Mrazek L, Greville W, Stough
Lee TF, Chen CF, Wang LC. 2004. Effect of ginkgolides on C. 2002. The acute nootropic effects of Ginkgo biloba in
beta-amyloid-suppressed acetylcholine release from rat healthy older human subjects: a preliminary investigation.
hippocampal slices. Phytother Res 18: 556 – 560. Hum Psychopharmacol 17: 45–49.
Lehmann E, Kinzler E, Friedemann J. 1996. Efficacy of a special Nathan PJ, Tanner S, Lloyd J et al. 2004. Effects of a combined
Kava extract (Piper methysticum) in patients with states of extract of Ginkgo biloba and Bacopa monniera on cognitive
anxiety, tension and excitedness of non-mental origin – A function in healthy humans. Hum Psychopharmacol 19: 91–
double-blind placebo-controlled study of four weeks treat- 96.
ment. Phytomedicine 3: 113 –119. Oliva I, Gonzalez-Trujano ME, Arrieta J, Enciso-Rodriguez
Lewin L. 1886. Uber Piper methysticum (Kawa). A Hirschwald: R, Navarrete A. 2004. Neuropharmacological profile of
Berlin. hydroalcohol extract of Valeriana edulis ssp. procera roots
Lindahl O, Lindwall L. 1989. Double blind study of a valerian in mice. Phytother Res 18: 290–296.
preparation. Pharmacol Biochem Behav 32: 1065 –1066. Padma TV. 2005. Ayurveda. Nature 436: 486.
Linde K, Ramirez G, Mulrow CD, Pauls A, Weidenhammer Pan HP. 2005. Progress in studies of pharmacological activities
W, Melchart D. 1996. St John’s wort for depression – an and clinical applications of preparations of dried leaf of
overview and meta-analysis of randomised clinical trials. Ginkgo biloba. Zhongguo Zhong Yao Za Zhi 30: 93–96.
Br Med J 313: 253–258. Pawar R, Gopalakrishnan C, Bhutani KK. 2001. Dammarane
Maher BF, Stough C, Shelmerdine A, Wesnes K, Nathan PJ. triterpene saponin from Bacopa monniera as the superoxide
2002. The acute effects of combined administration of inhibitor in polymorphonuclear cells. Planta Med 67: 752–
Ginkgo biloba and Bacopa monniera on cognitive function 754.
in humans. Hum Psychopharmacol 17: 163 –164. Piscitelli SC, Burstein AH, Chaitt D, Alfaro RM, Falloon J. 2000.
Malhotra CL, Das PK. 1959. Pharmacological studies of Herpestis Indinavir concentrations and St. John’s wort [Letter].
monniera Linn. (Brahmi). Indian J Med Res 47: 294 – 305. Lancet 355: 547– 548.
Mannel M. 2004. Drug interactions with St John’s wort. Drug Pittler MH, Ernst E. 2000. Efficacy of kava extract for treating
Safety 27: 773–797. anxiety: systematic review and meta-analysis. J Clin
Marsh WL, Davies JA. 2002. The involvement of sodium and Psychopharmacol 20: 84 – 89.
calcium ions in the release of amino acid neurotransmitters Poyares DR, Guilleminault C, Ohayon MM, Tufik S. 2002.
from mouse cortical slices elicited by hyperforin. Life Sci Can valerian improve the sleep of insomniacs after
71: 2645 –2655. benzodiazepine withdrawal? Prog Neuropsychopharmacol
Martis G, Rao A. 1992. Neuropharmacological activity of Biol Psychiatry 26: 539–545.
Herpestis monniera. Fitoterapia 63: 399 – 404. Rai D, Bhatia G, Palit G, Pal R, Singh S, Singh HK. 2003.
Martis G, Rao A, Karanth KS. 1992. Neuropharmaological Adaptogenic effect of Bacopa monniera (Brahmi). Pharmacol
activity of Herpestis monniera. Fitoterapia 63: 399 – 404. Biochem Behav 75: 823 – 830.
Medina JH, Pena C, Levi de Stein M, Wolfman C, Paladini AC. Rao CHV, Sairam K, Goel RK. 2000. Experimental evaluation of
1989. Benzodiazepine-like molecules, as well as other ligands Bacopa monniera on rat gastric ulceration and secretion.
for the brain benzodiazepine receptors, are relatively com- Indian J Physiol Pharmacol 44: 435–441.

Copyright © 2006 John Wiley & Sons, Ltd. Phytother. Res. 20, 1023–1035 (2006)
DOI: 10.1002/ptr
1034 V. KUMAR

Re L, Corneli C, Sturani E et al. 2003. Effects of Hypericum Silva BA, Dias AC, Ferreres F, Malva JO, Oliveira CR. 2004.
extract on the acetylcholine release: a loose patch clamp Neuroprotective effect of H. perforatum extracts on beta-
approach. Pharmacol Res 48: 55 – 60. amyloid-induced neurotoxicity. Neurotox Res 6: 119 –130.
Rodriguez-Landa JF, Contreras CM. 2003. A review of clinical Singh HK, Dhawan BN. 1982. Effect of Bacopa monniera
and experimental observations about antidepressant actions Linn. (brahmi) extract on avoidance responses in rat. J
and side effects produced by Hypericum perforatum ex- Ethnopharmacol 5: 205–214.
tracts. Phytomedicine 10: 688–699. Singh HK, Dhawan BN. 1992. Drugs affecting learning and
Rohini G, Sabitha KE, Devi CS. 2004. Bacopa monniera Linn. memory. In Lectures in Neurobiology, Tandon PN, Bijiani
extract modulates antioxidant and marker enzyme status in V, Wadhwa S (eds), vol. 1. Wiley Eastern: New Delhi, 189–
fibrosarcoma bearing rats. Indian J Exp Biol 42: 776–780. 207.
Roodenrys S, Booth D, Bulzomi S, Phipps A, Micallef C, Smoker Singh HK, Dhawan BN. 1997. Neuropsychopharmacological
J. 2002. Chronic effects of Brahmi (Bacopa monnieri) on effects of the Ayurvedic nootropic Bacopa monniera Linn
human memory. Neuropsychopharmacology 27: 279–281. (Brahmi). Indian J Pharmacol 29: S359–S365.
Roz N, Rehavi M. 2004. Hyperforin depletes synaptic vesicles Singh HK, Shanker G, Patnaik GK. 1996. Neuropharmacological
content and induces compartmental redistribution of nerve and anti-stress effects of bacosides: a memory enhancer.
ending monoamines. Life Sci 75: 2841–2850. Indian J Pharmacol 28: 47.
Ruschitzka F, Meier PJ, Turina M, Lüscher TF, Noll G. 2000. Singh HK, Rastogi RP, Srimal RC, Dhawan BN. 1988. Effect of
Acute heart transplant rejection due to Saint John’s wort. bacosides A and B on avoidance responses in rats.
Lancet 355: 548–549. Phytothey Res 2: 70–75.
Russo A, Borrelli F. 2005. Bacopa monniera, a reputed nootropic Singh RH, Singh L. 1980. Studies on the anti-anxiety effect of
plant: an overview. Phytomedicine 12: 305–317. the medyha rasayana drug Brahmi (Bacopa monniera
Russo A, Borrelli F, Campisi A, Acquaviva R, Raciti G, Vanella Wettst.). Res Ayur Siddha 1: 133 –148.
A. 2003b. Nitric oxide-related toxicity in cultured astrocytes: Singh S, Eapen S, D’Souza SF. 2006. Cadmium accumulation
effect of Bacopa monniera. Life Sci 73: 1517–1526. and its influence on lipid peroxidation and antioxidative
Russo A, Izzo AA, Borrelli F, Renis M, Vanella A. 2003a. Free system in an aquatic plant, Bacopa monnieri L.
radical scavenging capacity and protective effect of Bacopa Chemosphere 62: 233–246.
monniera L. on DNA damage. Phytother Res 17: 870–875. Singh YN. 2005. Potential for interaction of kava and St. John’s
Sairam K, Dorababu M, Goel RK, Bhattacharya SK. 2002. Anti- wort with drugs. J Ethnopharmacol 100: 108 –113.
depressant activity of standardized extract of Bacopa Singh YN, Singh NN. 2002. Therapeutic potential of kava in the
monniera in experimental models of depression in rats. treatment of anxiety disorders. CNS Drugs 16: 731–743.
Phytomedicine 9: 207–211. Smith DB. 1991. Cognitive effects of anti-epileptic drugs. Adv
Sairam K, Rao CV, Babu MD, Goel RK. 2001. Prophylactic and Neurol 55: 197–212.
curative effects of Bacopa monniera in gastric ulcer mod- Smith JV, Luo Y. 2003. Elevation of oxidative free radicals in
els. Phytomedicine 8: 423–430. Alzheimer’s disease models can be attenuated by Ginkgo
Sairam K, Rao CV, Goel RK. 2001c. Effect of Convolvulus biloba extract EGb 761. J Alzheimers Dis 5: 287–300.
pluricaulis Chois on gastric ulceration and secretion in rats. Smith KK, Dharmaratne HR, Feltenstein MW et al. 2001.
Indian J Exp Biol 39: 350–354. Anxiolytic effects of kava extract and kavalactones in the
Santillos H. 1991. Natural Healing with Herbs. Hohn Press: chick social separation-stress paradigm. Psychopharma-
Prescott, Arizona. cology 155: 86 – 90.
Santos MS, Ferreira F, Faro C et al. 1994c. The amount of GABA Sparreboom A, Cox MC, Acharya MR, Figg WD. 2004. Herbal
present in aqueous extracts of valerian is sufficient to remedies in the United States: potential adverse interac-
account for [3H]GABA release in synaptosomes. Planta Med tions with anticancer agents. J Clin Oncol 22: 2489–2503.
60: 475–476. Stackman RW, Eckenstein F, Frei B, Kulhanek D, Nowlin J, Quinn
Schellenberg R, Sauer S, Abourashed EA, Koetter U, Brattstrom JF. 2003. Prevention of age-related spatial memory deficits
A. 2004. The fixed combination of valerian and hops in a transgenic mouse model of Alzheimer’s disease by
(Ze91019) acts via a central adenosine mechanism. Planta chronic Ginkgo biloba treatment. Exp Neurol 184: 510 – 520.
Med 70: 594–597. Stevinson C, Ernst E. 2000. A pilot study of Hypericum
Schultz OE, Mueller F. 1960. Investigations on valerian active perforatum for the treatment of premenstrual syndrome. Br
substances with the Haffner test. Arzneimittelforschung 10: J Obstet Gynaecol 107: 870–876.
78 – 88. Stough C, Lloyd J, Clarke J et al. 2001. The chronic effects of an
Schulz V. 2000. Incidence and clinical relevance of the interac- extract of Bacopa monniera (Brahmi) on cognitive function
tions and side effects of Hypericum preparations. Perfusion in healthy human subjects. Psychopharmacology (Berl) 156:
13: 486 – 494. 481– 484.
Schulz V. 2003a. Ginkgo extract or cholinesterase inhibitors in Sumathi T, Nayeem M, Balakrishna K, Veluchamy G, Devaraj
patients with dementia: what clinical trials and guidelines SN. 2002a. Alcoholic extract of ‘Bacopa monniera’ reduces
fail to consider. Phytomedicine 10: 74 – 79. the in vitro effects of morphine withdrawal in guinea-pig
Shankar G, Singh HK. 2000. Anxiolytic profile of standardized ileum. J Ethnopharmacol 82: 75 – 81.
Brahmi extract. Indian J Pharmacol 32: 152. Sumathy T, Govindasamy S, Balakrishna K, Veluchamy G. 2002b.
Shanmugasundaram ER, Akbar GK, Shanmugasundaram KR. Protective role of Bacopa monniera on morphine-induced
1991. Brahmighritham, an Ayurvedic herbal formula for the brain mitochondrial enzyme activity in rats. Fitoterapia 73:
control of epilepsy. J Ethnopharmacol 33: 269–276. 381–385.
Sharma R, Chaturvedi C, Tewari PV. 1987. Efficacy of Bacopa Sumathy T, Subramanian S, Govindasamy S, Balakrishna K,
monniera in revitalizing intellectual functions in children. J Veluchamy G. 2001. Protective role of Bacopa monniera on
Res Educ Indian Med 1: 12. morphine induced hepatotoxicity in rats. Phytother Res 15:
Shaughnessy F. 2002. Ease anxiety with shankhpushpi: 643–645.
Ayurvedic doctors prefer this mood-soother over kava-kava Taibi DM, Bourguignon C, Taylor AG. 2004. Valerian use for
– Herb Brief. Natural Health Sept.: 1 as available on http:// sleep disturbances related to rheumatoid arthritis. Holist
www.findarticles.com/p/articles/mi_m0NAH/is_7_32/ Nurs Pract 18: 120 –126.
ai_91563259 Teschke R, Gaus W, Loew D. 2003. Kava extracts: safety and
Shinomiya K, Fujimura K, Kim Y, Kamei C. 2005a. Effects of risks including rare hepatotoxicity. Phytomedicine 10: 440 –
valerian extract on the sleep-wake cycle in sleep-disturbed 446.
rats. Acta Med Okayama 59: 89–92. Thies PW, Funke S. 1966. On the active ingredients in baldrian.
Shinomiya K, Inoue T, Utsu Y et al. 2005b. Effects of kava-kava 1. Detection and isolation of isovalerian acid esters
extract on the sleep-wake cycle in sleep-disturbed rats. with sedative effect from roots and rhizomes of various
Psychopharmacology 180: 564 – 569. valerian and kentranthus species. Tetrahedron Lett 11: 1155 –
Shukia B, Khanna NK, Godhwani JL. 1987. Effect of Brahmi 1162.
Rasayan on the central nervous system. J Ethnopharmacol Thompson R, Ruch W, Hasenohrl RU. 2004. Enhanced cognitive
21: 65 –74. performance and cheerful mood by standardized extracts
Sierpina VS, Wollschlaeger B, Blumenthal M. 2003. Ginkgo of Piper methysticum (Kava-kava). Hum Psychopharmacol
biloba. Am Fam Physician 68: 923 – 926. 19: 243–250.

Copyright © 2006 John Wiley & Sons, Ltd. Phytother. Res. 20, 1023–1035 (2006)
DOI: 10.1002/ptr
POTENTIAL MEDICINAL PLANTS FOR CNS DISORDERS 1035

Tierra M. 1990. The Way of Herbs. Pocket Books: New York, Wheatley D. 2001a. Kava and valerian in the treatment of stress-
177–178. induced insomnia. Phytother Res 15: 549–551.
Treiber K, Singer A, Henke B, Muller WE. 2005. Hyperforin Wheatley D. 2001b. Stress-induced insomnia treated with kava
activates nonselective cation channels (NSCCs). Br J and valerian: singly and in combination. Hum Psychopharm-
Pharmacol 145: 75–83. acol 16: 353–356.
Trevena L. 2004. Sleepless in Sydney – is valerian an effective Wheatley D. 2005. Medicinal plants for insomnia: a review of
alternative to benzodiazepines in the treatment of insom- their pharmacology, efficacy and tolerability. J Psycho-
nia? ACP J Club 141: A14–A16. pharmacol 19: 414 –421.
Tripathi YB, Chaurasia S, Tripathi E, Upadhyay A, Dubey GP. Widy-Tyszkiewicz E, Piechal A, Joniec I, Blecharz-Klin K. 2002.
1996. Bacopa monniera Linn. as an antioxidant: mechanism Long term administration of Hypericum perforatum im-
of action. Indian J Exp Biol 34: 523–526. proves spatial learning and memory in the water maze.
Trofimiuk E, Walesiuk A, Braszko JJ. 2005. St John’s wort Biol Pharm Bull 25: 1289–1294.
(Hypericum perforatum) diminishes cognitive impairment Williams B, Watanabe CM, Schultz PG, Rimbach G, Krucker T.
caused by the chronic restraint stress in rats. Pharmacol 2004. Age-related effects of Ginkgo biloba extract on
Res 51: 239–246. synaptic plasticity and excitability. Neurobiol Aging 25: 955 –
Ulbricht C, Basch E, Boon H et al. 2005. Safety review of kava 962.
(Piper methysticum) by the Natural Standard Research Witte S, Loew D, Gaus W. 2005. Meta-analysis of the efficacy of
Collaboration. Expert Opin Drug Saf 4: 779–794. the acetonic kava-kava extract WS1490 in patients with non-
Uphof JC. 1968. The Dictionary of Economic Plants, vol. 62. psychotic anxiety disorders. Phytother Res 19: 183 –188.
Verlag von J Cramer: New York. Yao ZX, Han Z, Drieu K, Papadopoulos V. 2004. Ginkgo biloba
van Dongen MC, van Rossum E, Kessels AG, Sielhorst HJ, extract (Egb 761) inhibits beta-amyloid production by low-
Knipschild PG. 2000. The efficacy of ginkgo for elderly ering free cholesterol levels. J Nutr Biochem 15: 749–756.
people with dementia and age-associated memory impair- Yuan CS, Mehendale S, Xiao Y, Aung HH, Xie JT, Ang-Lee MK.
ment: new results of a randomized clinical trial. J Am Geriatr 2004. The gamma-aminobutyric acidergic effects of valerian
Soc 48: 1183–1194. and valerenic acid on rat brainstem neuronal activity. Anesth
Van Veen AG. 1938. (Over the bedwelmende stof vit de Analg 98: 353–358.
kawakawa of wati-plant) (Piper methysticum). Geneeskundig Yue QY, Bergquist C, Gerden B. 2000. Safety of St. John’s wort
tijdschrift voor Nederlandsch-Indie 78: 194 –195. (Hypericum perforatum) [Letter]. Lancet 355: 576–577.
Verotta L. 2003. Hypericum perforatum, a source of neuroactive Zanoli P. 2004. Role of hyperforin in the pharmacological activi-
lead structures. Curr Top Med Chem 3: 187–201. ties of St. John’s Wort. CNS Drug Rev 10: 203–218.
Vitiello B, Shader RI, Parker CB et al. 2005. Hyperforin plasma Zanoli P, Rivasi M, Baraldi C, Baraldi M. 2002. Pharmacological
level as a marker of treatment adherence in the National activity of hyperforin acetate in rats. Behav Pharmacol 13:
Institutes of Health Hypericum Depression Trial. J Clin 645 – 651.
Psychopharmacol 25: 243–249. Z’Brun A. 1995. Ginkgo – myth and reality. Schweiz Rundsch
Vohora D, Pal SN, Pillai KK. 2000. Protection from phenytoin- Med Prax 84: 1–6.
induced cognitive deficit by Bacopa monniera, a reputed Zhang ZJ. 2004. Therapeutic effects of herbal extracts and con-
Indian nootropic plant. J Ethnopharmacol 71: 383 – 390. stituents in animal models of psychiatric disorders. Life Sci
Warburton DM. 1988. Clinical Psychopharmacology of Ginkgo 75: 1659–1699.
biloba extract. In Rokan (Ginkgo biloba): Recent Results in Ziegler G, Ploch M, Miettinen-Baumann A, Collet W. 2002. Effi-
Pharmacology and Clinic, Flmfgeld EW (ed.), Springer cacy and tolerability of valerian extract LI 156 compared
Verlag, Berlin-Heidelberg, 327–345. with oxazepam in the treatment of non-organic insomnia –
Weichel O, Hilgert M, Chatterjee SS, Lehr M, Klein J. 1999. a randomized, double-blind, comparative clinical study. Eur
Bilobalide, a constituent of Ginkgo biloba, inhibits NMDA- J Med Res 7: 480–486.
induced phospholipase A2 activation and phospholipid Zimmermann M, Colciaghi F, Cattabeni F, Di Luca M. 2002.
breakdown in rat hippocampus. Naunyn Schmiedebergs Ginkgo biloba extract: from molecular mechanisms to the
Arch Pharmacol 360: 609–615. treatment of Alzheimer’s disease. Cell Mol Biol (Noisy-le-
Wettstein A. 2000. Cholinesterase inhibitors and Gingko grand) 48: 613–623.
extracts – are they comparable in the treatment of demen- Zou L, Henderson GL, Harkey MR, Sakai Y, Li A. 2004. Effects
tia? Comparison of published placebo-controlled efficacy of kava (Kava-kava, ’Awa, Yaqona, Piper methysticum) on
studies of at least six months’ duration. Phytomedicine 6: c-DNA-expressed cytochrome P450 enzymes and human
393 –401. cryopreserved hepatocytes. Phytomedicine 11: 285–294.

Copyright © 2006 John Wiley & Sons, Ltd. Phytother. Res. 20, 1023–1035 (2006)
DOI: 10.1002/ptr

You might also like