Download as pdf or txt
Download as pdf or txt
You are on page 1of 84

Natural Products for

Neurodegenerative Diseases

Editors
Yung Hou Wong, Hong Kong
Joseph T.Y. Wong, Hong Kong

48 figures, 2 in color and 2 tables, 2005

Basel • Freiburg • Paris • London • New York •


Bangalore • Bangkok • Singapore • Tokyo • Sydney
S. Karger Drug Dosage All rights reserved.
Medical and Scientific Publishers The authors and the publisher have exerted every effort to en- No part of this publication may be translated into other
sure that drug selection and dosage set forth in this text are in languages, reproduced or utilized in any form or by any means,
Basel • Freiburg • Paris • London
accord with current recommendations and practice at the time electronic or mechanical, including photocopying, recording,
New York • Bangalore • Bangkok of publication. However, in view of ongoing research, changes microcopying, or by any information storage and retrieval
Singapore • Tokyo • Sydney in government regulations, and the constant flow of informa- system, without permission in writing from the publisher or, in
tion relating to drug therapy and drug reactions, the reader is the case of photocopying, direct payment of a specified fee to
urged to check the package insert for each drug for any change the Copyright Clearance Center (see ‘General Information’).
in indications and dosage and for added warnings and precau-
tions. This is particularly important when the recommended © Copyright 2005 by S. Karger AG,
agent is a new and/or infrequently employed drug. P.O. Box, CH–4009 Basel (Switzerland)
Printed in Switzerland on acid-free paper by
Reinhardt Druck, Basel
ISBN 3–8055–7933–0

Fax +41 61 306 12 34


E-Mail karger@karger.ch
www.karger.com
Vol. 14, No. 1–2, 2005

Contents

5 Editorial

Reviews

6 Natural Products and Derivatives Affecting Neurotransmission Relevant


to Alzheimer‘s and Parkinson‘s Disease
Houghton, P.J. (London); Howes, M.-J. (Kew)

23 Regulation of Neuroinflammation by Herbal Medicine and Its


Implications for Neurodegenerative Diseases. A Focus on Traditional
Medicines and Flavonoids
Suk, K. (Daegu)

34 Search for Natural Products Related to Regeneration of the Neuronal


Network
Tohda, C.; Kuboyama, T.; Komatsu, K. (Toyama)

46 Multifunctional Activities of Green Tea Catechins in Neuroprotection.


Modulation of Cell Survival Genes, Iron-Dependent Oxidative Stress and
PKC Signaling Pathway
Mandel, S.A.; Avramovich-Tirosh, Y.; Reznichenko, L.; Zheng, H.; Weinreb, O.; Amit, T.;
Youdim, M.B.H. (Haifa)

61 Unique Properties of Polyphenol Stilbenes in the Brain: More than


Direct Antioxidant Actions; Gene/Protein Regulatory Activity
Doré, S. (Baltimore, Md.)

71 Neuroprotective Effects of Huperzine A. A Natural Cholinesterase


Inhibitor for the Treatment of Alzheimer‘s Disease
Wang, R.; Tang, X.C. (Shanghai)

83 Author Index
84 Subject Index

© 2005 S. Karger AG, Basel

Fax +41 61 306 12 34 Access to full text and tables of contents,


E-Mail karger@karger.ch including tentative ones for forthcoming issues:
www.karger.com www.karger.com/nsg_issues
Neurosignals 2005;14:5

Editorial

Natural products have a long history of use for neurological symptoms.


For example, Crocus sativus has traditionally been used as an antispasmodic
and sedative. Yet the study of the mechanisms of action of neuroactive natu-
ral products began only recently. Armed with an arsenal of enabling tech-
nologies, researchers are now able to take a closer look at the complex mo-
lecular events associated with the therapeutic actions of natural products in
neurodegenerative diseases. Extensive information has been generated to in-
spire the design and development of more efficacious and safe therapies. This
issue brings together contemporary perspectives and research findings of lead-
ing experts on a vast array of mechanisms underlying the effects of natural
products on neuroprotection and neuronal regeneration. The multifunction-
al properties of constituents of botanicals and their derivatives are discussed
here to illustrate the possibilities toward the development of effective multi-
target therapeutics for neurodegenerative diseases. The study of neuroactive
natural products will indeed advance our understanding of the nervous sys-
tem, and open up exciting opportunities in drug discovery.
Yung Hou Wong, Hong Kong
Joseph T.Y. Wong, Hong Kong

© 2005 S. Karger AG, Basel


1424–862X/05/0142–0005$22.00/0
Fax +41 61 306 12 34
E-Mail karger@karger.ch Accessible online at:
www.karger.com www.karger.com/nsg
Review

Received: September 27, 2004


Neurosignals 2005;14:6–22
Accepted after revision: November 16, 2004
DOI: 10.1159/000085382

Natural Products and Derivatives


Affecting Neurotransmission Relevant
to Alzheimer’s and Parkinson’s Disease
Peter J. Houghton a Melanie-Jayne Howes b
a King’s College London, London, and b Jodrell Laboratory, Royal Botanic Gardens, Kew, UK

Key Words and by the administration of dopaminergic agonists,


Alzheimer’s disease W Parkinson’s disease W especially the ergot alkaloid derivatives. The use of
Cholinesterase inhibitors W Cholinergic receptors W inhibitors of enzymes that cause breakdown of DA is an
Dopaminergic receptors W Acetylcholine W Dopamine W avenue which is being explored. As well as the major
Galantamine W Huperzine W Physostigmine W L-DOPA W natural products of clinical interest, the paper discusses
Ergot alkaloids the chemistry, activity and usage of the constituents of
plants used in traditional medicine for the treatment of
diseases presenting symptoms similar to those charac-
Abstract teristic for Alzheimer’s or Parkinson’s disease.
The two major neurodegenerative diseases Alzheimer’s Copyright © 2005 S. Karger AG, Basel

disease (AD) and Parkinson’s disease (PD) are character-


ised by low levels in the brain of the neurotransmitters
acetylcholine (ACh) and dopamine (DA), respectively. Introduction
Clinical treatment of these two conditions is palliative
and relies, in most cases, on improving stimulation at the Neurodegenerative disease is a generic term applied to
relevant receptors by either increasing levels of the a variety of conditions arising from a chronic breakdown
endogenous neurotransmitter or by the use of sub- and deterioration of the neurons, particularly those of the
stances which have a similar agonist response. Natural central nervous system (CNS). In addition, these neurons
products continue to provide useful drugs in their own may accumulate aggregated proteins which cause dys-
right but also provide templates for the development of function. Alzheimer’s disease (AD) and Parkinson’s dis-
other compounds. The major advances in the treatment ease or parkinsonism (PD) are the two best-known dis-
of AD have been the use of acetylcholinesterase inhibi- eases of this type and will be the main diseases associated
tors such as galantamine, huperzine A, physostigmine with this review. However, some specialists would classify
and its derivatives to increase the levels of ACh rather multiple sclerosis and spongiform encephalopathies as
than the use of cholinergic compounds, although com- neurodegenerative diseases. The latter have received pub-
pounds with nicotinic properties have attracted some licity in recent years due to the link between Creutzfeldt-
interest. In contrast, the treatment of PD has relied on the Jacob disease (CJD) in humans and ‘mad cow disease’.
elevation of DA levels by use of L-DOPA, its precursor, Spongiform encephalopathies have been shown to be

© 2005 S. Karger AG, Basel Peter J. Houghton


ABC 1424–862X/05/0142–0006$22.00/0 Pharmacognosy Research Laboratories, Department of Pharmacy
Fax + 41 61 306 12 34 King’s College London, Franklin-Wilkins Building
E-Mail karger@karger.ch Accessible online at: 150 Stamford Street, London SE1 9NH (UK)
www.karger.com www.karger.com/nsg Tel. +44 20 7848 4775, Fax +44 20 7848 4800, E-Mail peter.houghton@kcl.ac.uk
caused by prions, proteinaceous particles synthesized Increasingly attention is being paid to the excitatory
within the cell which replicate and accumulate in cells and amino acid neurotransmitters and relevant receptors
so alter protein structure and therefore function but, as which are present in the CNS, e.g. the N-methyl-D-aspar-
yet, there are no drugs available to treat these conditions. tate (NMDA) receptor. The use of these in designing new
Most commonly, neurodegenerative disease manifests drugs or of explaining the use of traditional medicinal
in elderly people and, in advanced industrialised and plant extracts is in its infancy and no significant findings
post-industrialised societies, where life expectancy is long, have been made of natural products which either bind to
this group of conditions is a major cause of morbidity and the receptors or affect levels of the transmitters.
of death, as well as imposing severe strains on the social
welfare systems. To illustrate this it is reported that, in the
USA, AD now affects at least 5 million people and in the Alzheimer’s Disease
UK in 2001 it affected 750,000 people and was the fourth
most common cause of death [1]. However, it is increas- AD is the major disease of a group which is character-
ingly becoming recognized by the World Health Organisa- ised by loss of cognitive function leading to dementia. AD
tion as a global problem [2]. The common symptoms of is estimated to account for between 50 and 60% of
neurodegenerative diseases, such as loss of memory and dementia cases in persons over 65 years of age [5] and is a
tremor, have been recognised as a feature of increasing progressive, neurodegenerative disease that primarily af-
age for a long time and are acknowledged in many tradi- fects the elderly population. It is a major public health
tional medical systems. However, it is only comparatively concern in developed countries because of the strains
recently that, as distinctive diseases, they have been rec- imposed on carers and financial resources by the increas-
ognized and received much attention from mainstream ing numbers of sufferers. The main symptoms associated
medicine. This is most likely due to the fact that, in the with AD involve a decline in cognitive dysfunction, pri-
past, short life expectancy precluded many surviving to an marily memory loss [6, 7] and in the later stages of the
age where neurodegeneration was likely to affect a signifi- disease language deficits, depression, agitation, mood dis-
cant part of the population. turbances and psychosis are often seen [8].
The etiology of neurodegenerative diseases is still largely Although AD, as a defined medical condition, has only
unknown but, especially for AD and PD, postmortem stud- existed for about 100 years, age-related loss of memory
ies have shown clear links between the disease and a defi- and cognitive decline has been documented for thousands
ciency of neurotransmitters in parts of the brain. Thus, in of years in human history. In common with many other
AD there is a chronic shortage of acetylcholine (ACh) 1 and conditions, ancient writings which describe the symptoms
in PD a deficit of dopamine (DA) 2. Until very recently, the also suggest remedies, based usually on plant extracts.
only clinical treatment for both of these conditions was the Thus, Ashwagandha (Withania somnifera) is mentioned
reversal of these deficiencies by elevation of the levels of the in ancient Sanskrit writings from India as a ‘medhara-
transmitter by agonists, or by inhibition of enzymes in- sayan’ or promoter of learning and memory retrieval
volved in their removal from the immediate locality of the whilst in the sixteenth century Gerard’s Herbal from the
synapse. These approaches are discussed more thoroughly UK, sage Salvia officinalis, is described as being ‘good for
below. A variety of natural products have been shown to the memory’. In recent years, the value of a few of these
play roles which would have the desired effects and some of has been demonstrated scientifically and some of the
these, or their derivatives, have been brought into clinical compounds mentioned below have been isolated from
use. This paper gives an overview of such compounds, as traditional medicinal plants.
well as others, with interesting activity which have not been Postmortem studies have shown that AD is character-
developed as drugs or are still undergoing clinical trials. It ized by low amounts of the enzyme choline acetyl trans-
should not be forgotten that there are also many traditional ferase (ChAT) and enzyme abnormalities which would
medicinal plants with a reputation of alleviating or prevent- produce levels of the neurotransmitter ACh 1 (fig. 1) [9,
ing symptoms of neurodegeneration and that these are used 10] and there also appears to be a depletion of nicotinic
as crude extracts and mixtures [3, 4]. In some cases, such function. Attention deficit in AD is reversed with nicotine
extracts have been shown to relieve neurodegenerative 3, which is reported to upregulate nicotinic receptors and
symptoms in animal models or display relevant in vitro to increase ACh release, so enhancing cholinergic neuro-
activity, but both the modes of action and identity of any transmission [11, 12].
compounds responsible have not been fully determined.

Natural Products and Neurotransmission Neurosignals 2005;14:6–22 7


CH3 CH3 N
+ HO NH2
CH3 N CH3
O O H CH2 OH
CH3
O HO O

1 Acetylcholine 2 Dopamine O

4 Hyoscine (scopolamine)

N
H
CH3 Fig. 2. Scopolamine, a muscarinic receptor
N
antagonist.
3 Nicotine

Fig. 1. Neurotransmitters of the CNS relevant to neurodegenerative also being explored as leads for chemotherapeutic treat-
disease and the agonist nicotine. ment of AD. Activities being explored include antioxi-
dant, anti-inflammatory and inhibition of ß-amyloid syn-
thesis, but these are not covered to any extent in this
ACh is certainly associated with cognitive function, paper.
since situations where it is blocked from acting on the cho-
linergic receptors by drugs such as hyoscine (scopolamine)
4 (fig. 2), which is a muscarinic antagonist, result in severe AD: The Use of Cholinergic Compounds
cognitive impairment in the patient. It is still unclear if
the low levels of ACh in the CNS are cause or effect as far The rationale underlying the use of cholinergic com-
as AD is concerned, but the repletion of levels has been pounds is that they are agonists of the nicotinic choliner-
exploited therapeutically with some success in the last 15 gic receptor and so compensate for the low levels of ACh.
years in the symptomatic relief of AD. The synthetic com- The binding of ACh to the receptor is shown diagram-
pound tacrine was the first drug introduced into the clinic matically in figure 3. It can be seen that the important fea-
and it increased the levels of ACh by inhibition of acetyl- tures required in a molecule are an amine which becomes
cholinesterase (AChE), the enzyme responsible for fast positively charged at the pH of the immediate environ-
breakdown of ACh after its release from the nerve ending. ment and so binds to an aspartate in the receptor, a por-
This inhibition results in ACh having a longer half-life tion of the molecule able to form hydrogen bonds with the
and therefore increasing in concentration at the synapse. asparagine domain of the receptor and small groups able
Tacrine was the first of several AChE inhibitors which to bind to hydrophobic sites near the aspartate region. In
have come into clinical use, but it is no longer used since addition, the receptor lies in a pocket which allows only
the more recent introductions, generally named second small molecules to enter.
generation inhibitors, are safer and have longer-lasting Although these compounds have been suggested as
effects. These recent introductions include at least two valuable agents in treating AD, because they also appear
based on natural products. It should be stressed that to inhibit fibrillary tangle and amyloid production, suc-
AChE inhibitors only alleviate some of the cognitive cess has been limited as far as clinical studies are con-
symptoms of the disease for a time and ultimately do not cerned, although results in animals were initially promis-
arrest the cognitive decline of the patient. ing [13]. Two major alkaloidal natural products are
Another approach which has been proposed is the known to have this effect, arecoline 5 and pilocarpine 6
employment of cholinergic and, to some extent, nicotinic (fig. 4). It can be seen that these molecules are small and
agonists, but this has not proved to be as useful therapeu- that they incorporate at least two of the criteria noted
tically as the inhibition of cholinesterase. above for binding to the receptor.
More recently prevention of glutamate-mediated neu- Arecoline is the major alkaloid present in areca or betel
rotoxicity has been a therapeutic target in AD. The N- nut, the fruit of the palm tree Areca catechu L. (Areca-
methyl-D-aspartate (NMDA) receptor antagonist, mem- ceae), which is extensively used as a masticatory through-
antine, has been introduced in some countries for clinical out the Indian subcontinent and other parts of southeast
use in AD patients. It should be noted that other types of Asia. It is estimated that 500 million people regularly
activity, besides increase of neurotransmitter levels, are chew betel nut (often referred to as ‘pan’ in India) in a

8 Neurosignals 2005;14:6–22 Houghton/Howes


form which is usually shredded, mixed with lime and
wrapped in a leaf of Piper betel L. Excessive salivation
occurs, which is a direct result of its cholinergic activity,
CH3
which also gives the mild CNS stimulation for which the Aspartic acid
product is principally used. There was some interest in -
COO CH3
arecoline as a treatment for AD since it showed improve- O
O CH3
N
ment in memory tests in rats [14]. Arecoline has been Hydrophobic sites
CH3
shown to bind to the M2 muscarinic receptors but not the H H
Hydrophobic site
O N
nicotinic receptors [15]. A small clinical study showed that,
when arecoline was given continually by intravenous infu-
sion in AD patients, it enhanced verbal memory [16]. Asparagine
Derivatives of arecoline have been synthesized in order to
improve selectivity for cortical muscarinic ACh receptors Fig. 3. Binding of acetylcholine to nicotinic receptor.
and two examples are Lu 25-109 and talsaclidine 8 which
are M1 receptor agonists. Although Lu 25-109 showed
encouraging results in vitro [17], it failed to improve cogni-
tion when tested clinically in patients with mild to moder-
O
ate AD [18]. Talsaclidine has been shown to increase choli- CH3 H
nomimetic central activation in animals and humans with- OCH3 N
O
out some of the side effects seen with AChE inhibitor ther- N N
H
apy, but cognitive function was not significantly improved CH3 CH3 O

[19]. Tests on rhesus monkeys did however show some


5 Arecoline 6 Pilocarpine
improvement in memory-related tasks but at doses which
gave unacceptable side effects [20].
Pilocarpine 6 is one of a series of related alkaloids N N

found in species of the South American plant genus Pilo- N


N CH3
O
carpus, known commonly as Jaborandi leaf, which was N
C
CH
N
used in traditional medicine since it induced sweating and CH3
urination, features which were perceived as being useful 7 LU 25-109 8 Talsaclidine
for eliminating toxins from the body. The molecular
structure of pilocarpine 6 bears similarities to ACh since
the positively charged N atom and the lactone binding to Fig. 4. Cholinergic compounds.
the serine are about the same distance apart. Chewing the
leaf causes copious salivation as well as other typical fea-
tures of cholinergic stimulation such as contraction of the
pupils. Pilocarpine has been shown to enhance memory A similar enzyme, butylcholinesterase BuChE, also occurs.
performance in aged rats [21], but no studies on its appli- If the cholinesterase is inhibited, the ACh does not hydro-
cation in humans for the treatment of AD have been lyse so quickly, and levels of ACh rise. The ‘classic’ cho-
reported and it appears to have been discarded as a poten- linesterase inhibitor is the alkaloid physostigmine 9, also
tial therapeutic lead. This is possibly due to its poor phar- called eserine. This was isolated from the Calabar Bean, the
macokinetic profile as it cannot pass through the blood- seeds of Physostigma venenosum Balf., in the nineteenth
brain barrier, as well as its undesirable side effects. century in studies stimulated by the use of the seeds as an
ordeal poison in what is now southeastern Nigeria.
The toxic effects of calabar bean extract were found to
AD: The Use of Cholinesterase Inhibitors – be due to excessive cholinergic stimulation resulting in
Alkaloids increased salivation, nausea, bradycardia, muscle cramps
and respiratory failure, as well as CNS effects such as agi-
To avoid the undesirable effects of excess cholinergic tation. The cholinergic excess was found to be caused by
stimulation, ACh is rapidly hydrolysed after release at the inhibition of the rapid breakdown of acetylcholine by
synapse by an enzyme named acetylcholinesterase AChE. physostigmine.

Natural Products and Neurotransmission Neurosignals 2005;14:6–22 9


H CH3 CH3
CH3
Aspartate N O CH3
OH CH3 N O N
N CH3 CH3
Serine CH3
O H O
O O N Histidine N
CH3
N
H3C O
9 Physostigmine 10 Neostigmine
N
H3C O CH3

CH3 H CH3
CH3
N CH3
CH3 N O
Tyrosine CH3
O

11 Rivastigmine

OH
H OH
O OH
CH3O O
Fig. 5. Interaction between acetylcholine and the acetylcholinester-
ase active site. N
N O
CH3

12 Galantamine 13 Hamayne
CH3
In recent years, the structure of AChE has been deter-
mined and the mode of binding for AChE inhibitors has
also been elucidated. A variety of AChEs exist according H
CH3 N
to the source species, but they vary only in small details
NH2
and all contain the active site at the base of a deep cleft in O

the enzyme.
Figure 5 illustrates the particular amino acid residues 14 Huperzine A

which are considered to be the most important in the


binding process and from this knowledge, structure-activ-
Fig. 6. Acetylcholinesterase inhibitors.
ity relationships of AChE inhibitors can be understood at
the intermolecular level. The important regions of an
inhibitor appear to be a positively charged nitrogen,
which binds to an aspartate residue, and a region, sepa- doses. This has been exploited in the area of insecticides,
rated by a lipophilic area from the positive charge, which and the insecticide carbaryl was developed by making
can form a hydrogen bond with a tyrosine or serine resi- synthetic analogues of physostigmine. In therapeutics,
due. A positively charged nitrogen is common in many cholinesterase inhibition had, until recently, a somewhat
alkaloids at body pH and it is not surprising that many of limited application in only ophthalmology and the treat-
the most powerful AChE inhibitors are alkaloids. In ment of myasthenia gravis. However, the realisation that
recent years, however, a number of natural products, early symptoms of AD could be improved by the use of
mentioned below, which do not contain any nitrogen, cholinesterase inhibitors awakened renewed interest and,
have been found to be AChE inhibitors. This raises ques- since physostigmine was known to cross the blood-brain
tions about their binding characteristics to AChE since barrier, several in vivo studies were conducted which
the key role played by a positively-charged moiety of the showed that it reduced symptoms of ACh deficiency in
molecule bonding with the anionic aspartate residue, can the CNS. Physostigmine was reported to protect mice
no longer account for the activity. The interactions be- against cognitive impairment caused by oxygen deficit
tween the enzyme and such non-alkaloidal compounds and it improved learning in rats [22]. Clinical studies
have generally not yet been investigated. showed significant cognitive benefits in both normal and
The cholinesterase inhibitors (fig. 6) cause overstimu- AD patients [23], but it had a short half-life, which has
lation of a number of functions in many animal species prevented its application clinically in AD patients, since
and exert a considerable toxic effect even at quite low this would require multiple daily dosing.

10 Neurosignals 2005;14:6–22 Houghton/Howes


As well as inhibiting AChE, physostigmine also inhib- have therapeutic advantages over other AChE inhibitors
its butylcholinesterase (BuChE), another enzyme found in and its value in vascular dementia as well as AD is recog-
the CNS, so adverse effects associated with BuChE inhibi- nised in recent studies [36]. Several other alkaloids with
tion, such as gastrointestinal disturbance, may also occur AChE inhibitory activity have been recently reported
with physostigmine. However, BuChE has recently been from members of the Amaryllidaceae, but they have not
implicated in the aetiology and progression of AD [24], so been subjected to vigorous pharmacological and clinical
inhibition of BuChE may therefore prove to be beneficial testing. Several other alkaloids isolated from Iberian Nar-
in treating AD and physostigmine and other BuChE cissus species have been tested for cholinesterase activity
inhibitors such as rivastigmine may have clinical efficacy [37]. A study of two Crinum species used in Nigerian tra-
superior to AChE-selective inhibitors. ditional medicine to help ailing memory resulted in the
To improve its pharmacokinetic profile, there is a con- isolation of four alkaloids of which the most active was
siderable history of the synthesis of analogues of physo- hamayne 13, although its IC50 value of 250 ÌM was three
stigmine. These have been applied to the treatment of orders of magnitude weaker than physostigmine, so it is
myasthenia gravis, neostigmine 10 being the most widely unlikely that it would be present in sufficient quantity to
used drug for this disease. Neostigmine is a quaternary have a strong therapeutic effect [38].
amine and this feature severely impairs its ability to cross Whilst physostigmine analogues and galantamine have
the blood-brain barrier and so be of value in treating AD. entered the pharmacopoeia in the Western world, another
Rivastigmine 11 was produced with the express purpose natural cholinesterase inhibitor, huperzine A 14 has been
of engineering a better pharmacokinetic profile for useful- introduced in China for treating AD. Huperzine A is one
ness in AD and it inhibits the G1 form of AChE in the of the alkaloids found in the clubmoss Huperzia serrata
cortex and hippocampus, brain areas involved in cogni- Thunb. (Lycopodiaceae) which is used in various formu-
tion [25], and it has been shown to improve cognition in lae in traditional Chinese medicine (TCM) to alleviate
AD patients [26, 27]. Clinical studies have borne out the problems of memory loss, promote circulation and for
usefulness of rivastigmine (Exelon®) in mild to moderate fever and inflammation [39]. Huperzine A is related to
AD and it has been licensed as a treatment for symptom- the quinolizidine alkaloids and it reversibly inhibits
atic relief of AD since 2000. AChE in vitro and in vivo [40, 41].
Galantamine 12 (sometimes referred to as galanth- Huperzine A has been shown to improve memory in
amine) is found in members of the Amaryllidaceae, e.g. cognitively impaired rats [42] and in gerbils following
the Chinese medicinal herb Lycoris radiata Herb. and the ischaemia [43]. These observations suggest that huperzine
European Galanthus nivalis L. and Narcissus spp. [28]. A has clinical potential in cerebrovascular disorders, as
The ethnopharmacological uses of plants containing this well as in AD. Indeed, since it has also been shown to be
compound are not very clear, but a full report of the histo- neuroprotective, AChE inhibition may not be the only
ry of the development of galantamine from Galanthus explanation for the clinical effects observed. Huperzine A
nivalis has been recently published [29]. Its cholinesterase has been shown to be neuroprotective against ß-amyloid
inhibitory properties were first exploited in Bulgaria in peptide fragment 25–53 and free radical-induced cytotox-
the mid-twentieth century for the treatment of polio vic- icity [44] and to attenuate apoptosis by inhibiting the
tims, but it only came into prominence as a treatment for mitochondria-caspase pathway [45]. In a multi-centre,
AD in the last decade of the twentieth century. double-blind trial, huperzine A significantly improved
Galantamine has been licensed in Europe for AD treat- memory and behaviour in AD patients, and was reported
ment since 2001. Multi-centre randomised clinical trials to be more selective for AChE than BuChE and was less
showed that it was well tolerated and significantly im- toxic than the synthetic AChE inhibitors donepezil and
proved cognitive function when administered to AD pa- tacrine [36]. A randomised, placebo-controlled study on
tients [30–32]. The cognitive benefits appear to be sus- over 200 patients, 100 of whom were given 400 Ìg huper-
tained for at least 3 years, a much longer time than for zine A daily for 12 weeks and showed significantly higher
other drugs of this type [33]. Galantamine is well ab- scores for improvement compared with the placebo group
sorbed when given orally and is also more selective for in the various scorings used [46]. Recent progress on
AChE than BuChE [34]. It also stimulates nicotinic recep- many aspects of huperzine A has been reviewed [47].
tors indirectly by its allosteric potentiation of ACh [35], Other alkaloids have shown AChE activity and most of
and so may also enhance cholinergic function and memo- these have been isolated from plants used in traditional
ry (see below). This effect suggests that galantamine may medicine (fig. 7). Coptis chinensis Franch. (Ranunculaceae)

Natural Products and Neurotransmission Neurosignals 2005;14:6–22 11


inhibited AChE in vitro, and reversed scopolamine-
O
induced memory impairment in rats [54]. It also in-
N+
O creased cerebral blood flow in vivo in cats, a property
OCH3 which would supplement its usefulness in AD [55]. Sola-
nidine 20 and related compounds and their glycosides are
OCH3
steroidal alkaloids produced in green parts of the genus
15 Berberine
Solanum, which includes the potato. The toxic properties
CH3O CH3O of these alkaloids have been known for a long time and
N+ N+ have been shown to be due to AChE inhibition, with char-
CH3O CH3O
acteristic signs of cholinergic excess such as sweating, pal-
O OCH3
pitations and CNS disturbances including hallucinations
O OCH3 [56]. Reports of Solanum species being used to treat AD
16 Coptisi ne 17 Palmatine or related conditions in traditional medicine do not exist
and the alkaloids have not been investigated for any use-
O fulness clinically, presumably because of their toxicity.
O
N N Although it is comparatively easy to explain the fact
that some alkaloids inhibit AChE because of their molec-
N N N
N
CH3
ular features, it is less easy to correlate chemical structure
and activity for some other phytochemical types for which
18 Rutaecarpine 19 Dehydroevodiamine AChE inhibition has recently been reported.
CH3 H
CH3
N AD: The Use of Cholinesterase Inhibitors –
CH3 H H CH3
H Terpenoids and Other Types of Compound
H H
HO
Terpenoids comprise a very large group of natural
20 Solanidine products and comprise two or more branched 5 carbon
units, formed from a common precursor named meva-
lonic acid. Skeletons consisting of multiplets of 2, 3, 4 or 6
Fig. 7. Minor alkaloidal cholinesterase inhibitors.
of these linked together in many different ways are found
in a variety of mostly cyclic compounds named monoter-
penes (10 carbons in the skeleton), sesquiterpenes (15 car-
has been used in TCM for several conditions including age- bons), diterpenes (20 carbons) and triterpenoids (30 car-
related cognitive and memory decline. Some alkaloids bons), respectively (fig. 8). These compounds tend to be
found in this species, such as berberine 15, coptisine 16 and lipophilic, so they are able to cross the blood-brain bar-
palmatine 17, are reported to also be anti-ChE [48, 49]. rier, and the monoterpenes, and some of the sesquiter-
Coptis chinensis extract improved a scopolamine-induced penes, are volatile, and so effects could occur through
learning and memory deficit in rats [50] and this is likely to inhalation. These compounds are responsible for the
be due to the alkaloids present raising ACh levels. strong odours and flavours of many herbs, spices and tra-
Berberine 15 has been shown to be selectively active ditional medicines. Many such molecules are increasingly
against AChE compared with BuChE [51] and it has been recognized as having a variety of roles in living organisms,
shown to improve scopolamine-induced amnesia in rats including signalling between members of the same spe-
[52]. Rutaecarpine 18 is the major alkaloid found in Evo- cies, thus comprising part of the group of compounds
dia rutaecarpa (Juss.) Benth. (Rutaceae), the unripe fruit known as pheromones, and protective or attractant roles
of which is used in TCM for cardiotonic, restorative and in flowering plants against herbivores and pollinators
analgesic effects. Pharmacological activities relevant to respectively. An effect on CNS activity by the volatile
AD have been identified with the extract and with rutae- substances in perfumes and other odoriferous materials
carpine. Rutaecarpine inhibited COX-2 activity in vitro, has attracted interest in recent years and one of the first
and was anti-inflammatory in vivo [53]. Dehydroevo- findings that monoterpenes had AChE inhibitory effects
diamine 19, another alkaloid found in the same plant, was made only in the mid 1990s in studies investigating

12 Neurosignals 2005;14:6–22 Houghton/Howes


historical records that monoterpene-containing plants
were ‘good for the memory’ [57]. CH3 CH3
One such group of plants was the various European CH3

species of Salvia, commonly known as sage. An ethanolic CHO


H3C
extract and oil of S. officinalis L. (Labiatae) and S. lavan- O CH3

dulaefolia Vahl. (Labiatae) were investigated for anti-ChE H3C CH3 H3C CH3

activity and it was found that all gave inhibition of AChE


21 1,8-Cineole 22 α-Pinene 23 Citral
at quite low concentrations [57].
The cholinesterase inhibition shown by the S. lavandu-
O O
laefolia oil was shown to be partly due to the cyclic mono- CH3 CH3
O O
terpenes 1,8-cineole 21 and ·-pinene 22, which were
shown to inhibit AChE in vitro, with some contribution O O
from other constituents, perhaps by acting synergistically
[58, 59]. However, the monoterpenes were considerably CH3 H3C CH3
less active, by a factor of at least 103, than the alkaloidal 24 Dihydrotanshinone 25 Cryptotanshinone
AChE inhibitors such as physostigmine 9 [58].
CH3
Since the effects of the oil were better than those of
H3C
individual monoterpenes, further in vivo and clinical
studies, described below, were carried out on the essential
CH3 CH3 H COOH
oils, which consist of a mixture of monoterpenes, rather
than isolated compounds. Oral administration of S. la- H CH3
vandulaefolia essential oil to rats decreased striatal AChE HO H
activity in both the striatum and the hippocampus com- CH3 CH3

pared to the control rats. Thus, it appeared that constitu- 26 Ursolic acid

ents of the S. lavandulaefolia oil, or their metabolites,


reach the brain and inhibit AChE in select brain areas,
Fig. 8. Terpenoidal cholinesterase inhibitors.
consistent with evidence of inhibition of the brain enzyme
in vitro [60].
Clinical studies on human volunteers and even pa-
tients with AD have been reported in recent years. The crossover study showed an improvement in cognitive per-
effect of sage in twenty participants using a placebo-con- formance and mood in 20 healthy young participants, fol-
trolled, double-blind, balanced, cross-over design showed lowing treatment with dried leaf of M. officinalis shown
significant effects on cognition associated with the lowest by previous in vitro tests to be cholinergically active [65].
dose of Salvia, including improvements in both imme- In another study, an extract of M. officinalis was adminis-
diate and delayed word recall scores [61]. A small trial tered to patients with mild to moderate AD for 4 months
with 11 patients showing mild to moderate symptoms of and gave a significantly better outcome on cognitive func-
AD showed that oral administration of the essential oil of tion than placebo [66]. Although the constituents present
S. lavandulaefolia significantly improved cognitive func- have not been investigated, numerous monoterpenes
tion in one of the three different methods of assessment have been identified in the essential oil of M. officinalis,
used [62]. Another, more thorough, trial in Iran on 48 including citral 23 (a mixture of the isomers geranial and
patients with similar symptoms of AD showed that those neral) and it is known that these are weak inhibitors of
treated with an extract of S. officinalis gave significantly AChE [67].
better values in measurements of cognitive function [63]. The root of another species of Salvia, S. miltiorrhiza
Melissa officinalis L. (Labiatae) leaf is another species Bung. (Labiatae) is extensively used in TCM to stabilise
that contains monoterpenes in its essential oil. It has been the heart and calm nerves [48]. S. miltiorrhiza has been
used as a medicinal plant for more than 2000 years and the subject of thorough investigation, and consequently
has a reputation of for promoting long life and for re- numerous pharmacological activities that may be relevant
storing memory [64]. Recent studies have focused on in CNS disorders, including AD, have been identified. S.
the reputed cognitive effects of M. officinalis. A recent miltiorrhiza has been employed for the treatment of cere-
randomised, placebo-controlled, double-blind, balanced bral vascular disease, and there are several studies to

Natural Products and Neurotransmission Neurosignals 2005;14:6–22 13


The withanolides are a group of compounds related to
CH3
the steroids which are found in some genera of the Solana-
CH2OH ceae, notably Withania somnifera (L.) Dun. The root of
O O
H3C H OH this plant is one of the most highly regarded herbs in
CH3 OH
O O OH Ayurvedic medicine where it is known as ‘ashwagandha’
O and has a history of use for almost 4,000 years. It is
CH3
classed among the rejuvenative tonics known as ‘Rasaya-
H
nas’ and several groups have described the cognitive
OH
O enhancing potential of extracts of the roots in experimen-
27 Sitoindoside IX tal animals. Some individual compounds have also been
CH3
CH2OH
investigated and the sitoindosides IX 27 and X (fig. 9)
H3C H
have been shown to augment learning acquisition and
CH3 O O memory in both young and old rats [71]. The mechanisms
H
O
CH3
to explain this effect are unclear, but may involve modu-
lation of cholinergic neurotransmission. An extract con-
H taining the sitoindosides VII–X and withaferin A 28 was
O administered to mice and effects on the neurotransmitter
OH
systems in the brain were observed. The results from this
study showed that the extract enhanced AChE activity in
28 Withaferin A the lateral septum and globus pallidus areas of the brain
and also enhanced muscarinic M1 receptor binding in cor-
tical regions, but it did not affect Á-aminobutyric acid
Fig. 9. Steroidal derivatives with anticholinesterase inhibitory activ- (GABA)A, benzodiazepine receptor binding, nor NMDA
ity. or amino-3-hydroxy-5-methyl-4-isoxazole propionic acid
(AMPA) glutamate receptor subtypes [72].
The extract containing the sitoindosides VII–X and
investigate possible mechanisms for the protective effect withaferin A also reversed the reduction in cholinergic
of S. miltiorrhiza against cerebral ischaemia. markers (e.g. ACh, choline acetyltransferase; ChAT) in
There is evidence that S. miltiorrhiza root extract may rats [73]. These activities could explain the reputed cogni-
protect neurons from ischaemia and attenuate dysfunc- tion enhancing effects of W. somnifera root because of
tion of neuropeptides of importance in neurodegenerative preferential action on cholinergic neurotransmission in
disease [68]. An inhibitory effect on AChE has been the cortical and basal forebrain, brain areas involved in
recently demonstrated and has been shown to be due to cognitive function. Based on this information, it could be
the diterpenes present known as tanshinones [69]. Dihy- speculated that the sitoindosides and withaferin A could
drotanshinone 24 was shown to be the most active (IC50 = have potential in AD therapy, but more studies need to be
1.0 ÌM) with cryptotanshinone 25 (IC50 = 7.0 ÌM ) also carried out before they can be used with any degree of
showing activity. A feature which appears to be necessary confidence in being able to achieve this or whether the
for activity is the saturated bond in the furan ring of the crude extract might produce a better effect.
molecules. In addition to their cholinergic activity, the glycowi-
Screening of 139 different Indian medicinal plants and thanolides showed anxiolytic and anti-depressant activi-
spices for AChE inhibitory activity led to Origanum ties in rats [74], which may be applicable in the symptom-
majorana L. (Labiatae) showing the highest activity. The atic treatment of AD.
active component was identified as the triterpene ursolic W. somnifera root and some constituents are also
acid 26, a fairly common compound, which exhibited an reported to have anti-oxidant properties which may also
IC50 value of 7.5 nM, less than an order of magnitude be relevant in AD therapy [75]. Anti-inflammatory effects
weaker than that of the positive control tacrine [70]. This of the extract of roots have been demonstrated in rats [76]
result is of interest in view of the widespread occurrence and the extract has also been shown to reduce levels of the
of ursolic acid and may account for the traditional use of pro-inflammatory interleukins IL-1 and TNF-·, which
several plant species for memory improvement and AD- are considered to be involved in senile plaque formation
related conditions. and neurodegeneration [77].

14 Neurosignals 2005;14:6–22 Houghton/Howes


There are many types of phenolic compounds, but the OH R
only ones which have been shown to possess AChE inhibi-
tory activity are neolignans from Magnolia officinalis. OH

The root and stem bark of Magnolia officinalis Rehd. Et


Wils. (Magnoliaceae) have been used in TCM to treat anx- H2C CH2

iety and nervous disturbances. M. officinalis contains the


29 R = H Honokiol
biphenolic lignans, honokiol 29 and magnolol 30 (fig. 10). 30 R = OH Magnolol
Both lignans increased ChAT activity and inhibited
AChE activity in vitro, and increased hippocampal ACh
Fig. 10. Neolignans with anticholinesterase
release in vivo [78]. Honokiol and magnolol show anxio- inhibitory activity.
lytic effects [79] and these appear to be due to their ability
to potentiate GABAergic neurotransmission [80]. These
two compounds also appear to have antioxidant anti-
O OH
inflammatory and neuroprotective properties and such
N
polyvalency in activity is of interest in their potential use
CH3
in treating AD [81, 82].
31 Lobeline

AD: Use of Nicotinic Compounds O


O

N N
A link between smokers and a lower incidence of AD
H
has been noted and this is thought to be associated with
increased nicotine intake, although some recent reports H H
N N
present a contrary view in linking smoking with an H
increased incidence of AD [83, 84]. Nicotine 3 is reported 32 Sophoramine 33 Cytisine

to have cognition-enhancing effects and these may be due


to nicotinic receptor stimulation but also protection
Fig. 11. Nicotinergic alkaloids.
against AD by other mechanisms such as inhibition of ß-
amyloid formation [85], inhibition of the neurotoxic
effects of excitatory amino acids (e.g. glutamate) and
enhancement of the effects of nerve growth factor (NGF) patients, although an estimated 5% of sufferers are under
[86]. There are several other alkaloids which are nicotinic 40 years old [89]. Its characteristic feature is an increasing
agonists at the cholinergic receptor [87] (fig. 11). Lobeline tremor in resting limbs and a rigidity, known as dyskine-
31 from Lobelia inflata interacts with the nicotinic recep- sia, particularly exhibited as a shuffling gait, but it is also
tor [88] and could also be exploited to influence choliner- associated with the degeneration of cognitive function
gic function in AD. Other alkaloids such as sophoramine and memory.
32 and cytisine 33, found in members of the Legumino- It is thought that oxidative stress in the substantia
sae, have nicotinic actions. Cytisine is used as a pharma- nigra plays a significant role in the loss of neurons which
cological tool because of its strong binding affinity to nico- produce DA [90]. This results in the characteristic defi-
tinic receptors, but it does not appear to have been devel- ciency of DA in the substantia nigra seen in the brain of
oped for any pharmaceutical purposes, probably because PD patients post mortem, the other characteristic histo-
of its toxicity. pathological observation being the presence of deposits
called Lewy bodies in the surviving neurons.
The major therapeutic approach to PD has been to ele-
Parkinson’s Disease vate the levels of DA by either inhibition of monoamine
oxidase (MAO), which metabolises DA to less active com-
Parkinson’s disease is named after the English surgeon pounds, or by increasing the concentration of the precur-
who first described a syndrome which he called the ‘shak- sor of DA by administering L-hydroxyphenylalanine (L-
ing palsy’. It affects a large number of people (about DOPA) 34 (fig. 12). Another approach involves the use of
20,000 in the UK) and is most commonly seen in older compounds which are agonists on the DA receptors.

Natural Products and Neurotransmission Neurosignals 2005;14:6–22 15


Ph alanine Ph alanine
COOH COOH 617 307
CH3
Aspartate
NH2 311
NHNH2 Serine
505
+
HO NH3 Ph alanine
616
HO HO
OH OH HO
Serine Tryptamine
508 613
34 L-DOPA 35 Carbidopa Ph alanine
509

NHR
HO Fig. 13. Interaction between dopamine and amino acid residues in
the dopamine receptor.

HO
OH
36 R = CH 3 Adrenaline
37 R=H Noradrenaline Tryptamine Tryptamine
617 307
CH3 CH3 Aspartate
HO O Serine 311
OH +
NHCH3 NH2 505
HO NH2 Ph alanine
616
CH3
HO
Serine Tryptamine
508 613
38 Ephedrine 39 Cathinone
Ph alanine
509

Fig. 12. Dopamine and related compounds. Fig. 14. Interaction between adrenaline (epinephrine) and amino
acid residues in the adrenergic receptor.

PD: Use of Dopaminergic Agonists is now administered routinely to PD patients. It is found


in commercially viable amounts in various species of
The structure-activity relationships of compounds bean, notably Mucuna spp., and this has been used as a
which are agonists at the DA receptor are generally commercial source, although the drug is now mainly
accepted to be the two ortho OH groups on the aromatic obtained by synthesis. L-DOPA is often given together
ring which bind with serine residues 505 and 508, the aro- with another analogue of DA, carbidopa 35, which is not
matic ring enabling hydrophobic interactions with a phe- dopaminergic but which inhibits dopa-decarboxylase and
nylalanine at 617 and, in the terminal position of the ethyl so, by maintaining levels of L-DOPA in the blood, pro-
amino group attached to the aromatic ring, a nitrogen longs its activity. When L-DOPA is given over long peri-
with a positive charge which interacts electrostatically ods of time, it is common for a sudden decline in sensitiv-
with an aspartate residue (fig. 13). It is noteworthy that ity to occur from time to time and this is called the ‘on-off’
the adrenergic receptor is very similar in some respects effect. This is probably due to a number of factors includ-
(fig. 14) and so it is possible that compounds with a struc- ing depletion in the ability of the substantia nigra cells to
ture favouring binding to one of these receptors, also have store DA and desensitization of the receptors. Other
some effect on the other. dopaminergic agents, some of which are mentioned be-
DA itself is quite unstable and cannot cross the blood- low, are often used as adjuvants to reverse the ‘off’ effect.
brain barrier. However, it can be formed within the brain It is interesting that the powdered seeds of Mucuna
by conversion of its precursor L-DOPA. This compound pruriens L. have been used in Ayurvedic medicine for dis-

16 Neurosignals 2005;14:6–22 Houghton/Howes


eases of the nervous system [91] and this product has
shown to reduce adverse effects, such as the ‘on-off’ effect,
HO N HO N
in patients [92]. A commercial extract of M. pruriens HP- H
H3C
O H O
O H3C
200 was shown to be twice as effective as the equivalent CH3
H N H N CH3
N N
dose of L-DOPA in rats [93]. This may be due to the pres- O O
CH3 CH3 CH3
ence of other, more active, compounds. However, a later O N O N
H H
study showed that when the same preparation was given
to rats over a 52-week period, it elevated DA levels in the
cortex but not in the striatum nigrum, this calling into Br
question whether the observed improvements in parkin- N N
H H
sonian symptoms were due to the hypothesis originally 40 Ergotamine 41 Bromocriptine
proposed, i.e. that the L-DOPA in the Mucuna extract
was converted to DA and reached the parts of the brain NHC 2H5
CH3 O N(C 2H5)2
where a deficiency is associated with PD [93]. SCH3
O N
N
C 3H7 CH3
The transmitter dopamine DA is one of a group of N CH2 H
N
N
CH3
H O N H
compounds known as phenylpropylamines, which in- H
cludes some important neurotransmitters such as adrena-
line (epinephrine) 36 and noradrenaline (norepinephrine)
N
37. The phenylpropylamines also include several natural- H N
N
H
ly-occurring compounds known as protoalkaloids, since H

their N atom is not part of a heterocyclic ring. Most pro- 42 Pergolide 43 Cabergoline 44 Lisuride

toalkaloids appear to have a greater adrenergic than dopa-


minergic effect, although it appears that they can stimu-
Fig. 15. Ergot alkaloid derivatives used to treat parkinsonism.
late both types of receptor. The major naturally-occurring
compound of this type used therapeutically is ephedrine
38, obtained from some species of Ephedra (Ephedra-
ceae), and these plants which have been used for many ergot indole alkaloids (fig. 15). Ergot, Claviceps purpurea
centuries in TCM. Ephedrine is used principally for its Tulasne (Clavipitaceae), is a fungus which infects the ears
adrenergic sympathomimetic properties in drying up se- of cereal crops, notably rye Secale cereale L. Ergot has a
cretions, but it is well-known for its CNS-stimulant side long history of poisoning animals and humans who eat the
effects, which may be due also to dopaminergic proper- cereal flour contaminated with ergot, but also has been
ties. In contrast, the synthetic phenylpropylamines known exploited in traditional medicine in some parts of Europe
as amphetamines are primarily employed as CNS stimu- to aid childbirth, since an extract causes contraction of the
lants and there appears to be some association between uterus towards the end of pregnancy and also constricts
their use and alleviation of the dyskinesia often associated blood vessels, thus reducing bleeding which often occurs
with PD. A natural compound very similar in chemistry at birth. The effects of ergot on the CNS has also been
and pharmacology to the amphetamines is cathinone 39, well-documented and outbreaks of ‘madness’ involving
a major active constituent of Catha edulis Forsk. (Celas- hallucinations have been shown to correlate with times of
traceae). The fresh young leaves of this plant are known as heavy contamination of rye flour in the communities
‘khat’ and are used as a stimulant masticatory in Ethiopia, affected. The activity of ergot has been shown to be due to
Yemen and surrounding countries and by the diaspora of the alkaloids present such as ergotamine 40. All of the
those communities in Europe [94]. Cathinone has been alkaloids contain the indole ring structure known as lyser-
shown to be present mainly in the young leaves only and, gic acid and similarities with the three neurotransmitters
since there have been anecdotal reports of reduction in noradrenaline (norepinephrine), DA and serotonin can be
PD-like tremors induced by neuroleptic drugs in some seen in this (fig. 16). This probably explains the wide spec-
regular chewers of khat, it might be that this is due to a trum of activity of these alkaloids and so it has been found
similar effect of cathinone as that observed for amphet- necessary to alter the chemical structure to produce com-
amines [95]. pounds which more specifically bind to only one of the
Probably the second most important group of com- receptors. A large amount of derivatives of ergot alkaloids
pounds used for the treatment of PD are derivatives of the have been synthesized for differing therapeutic effects

Natural Products and Neurotransmission Neurosignals 2005;14:6–22 17


NHR
NHR
NH2 NH2
NCH3 NH2
O HO NCH3
H O
H
HO

OH OH N
N
H OH H N
OH H

Ergot alkaloid Noradrenaline Dopamine Serotonin (5-HT) Ergot alkaloid

Fig. 16. Ergot alkaloid skeleton showing


parts similar to neurotransmitters.

and one of these has been dopaminergic receptor stimula-


tion for use in treating PD. Bromocriptine 41, pergolide
HO 42, cabergoline 43 and lisuride 44 are examples of com-
HO
pounds which have been developed in this way and are
HO
H
CH3
N now used clinically. The pharmacological differences be-
HO H
N
CH3
tween the compounds are not very great. All are D2 dopa-
mine receptor agonists, although pergolide also has ago-
45 Apomorphine 46 Salsolinol
nist effects at D1 and D3 receptors. All four drugs are well-
established in treatment and numerous reviews exist on
H
N
CH3 their clinical efficacy and application [96, 97].
CH3O
Bromocriptine and lisuride were the first of these drugs
N H to be introduced and have comparatively short half-lives.
H Pergolide has a half-life of 8 h [98] and cabergolide of
47 Ibogaine 68 h, making the latter especially useful for administra-
tion only once a day [99]. The ergot-derived drugs were
CH3
originally used in advanced cases of PD, but there is
N
increasing interest in their use in the first instance since,
unlike L-DOPA, they do not need to be metabolized to
O the active compound and so are not affected by any
degeneration in the dopaminergic terminals [100].
Another alkaloid with agonist activity is apomorphine
45, derived from the opium alkaloids (fig. 17). It acts on the
48 Benzatropine
D1 and D2 receptors, but, because it is a powerful emetic
when given orally, its use is restricted to parenteral admin-
N N istration. It is most commonly given when patients are not
CH3 CH3
exhibiting adverse effects to L-DOPA and as a diagnostic
CH3O N CH3O N
H H
agent for dopaminergic responsiveness. The isoquinolin-
49 Harmine 50 Harmaline
oid salsolinol 46 has been investigated extensively over the
last 10 years. This compound is found in the cocoa bean,
the seeds of Theobroma cacao L. (Sterculiaceae), and also
in chocolate derived from this plant. The addictive proper-
Fig. 17. Other alkaloids with dopaminergic and monoamine oxidase ties of chocolate have been ascribed to the dopaminergic
inhibitory activity. activity of this compound [101], but it also occurs as an
endogenous catechol in the brain. Salsolinol has been
shown to be dopaminergic at the D2 receptors and also to
have a protective effect on neurodegeneration [102]. The
N-methyl derivative, which can be synthesized in the

18 Neurosignals 2005;14:6–22 Houghton/Howes


brain, has a role in the pathogenesis of PD [103], and this
complicates the usefulness of giving salsolinol as a protec- R

tive substance to prevent or alleviate PD. OH

Mention should be made of another indole alkaloid, HO O


ibogaine 47, obtained from the African plant Tabernanthe
iboga Baill. (Apocynaceae), the roots of which were used OH
as a stimulant and, in larger doses, as a hallucinogen in OH O
central Africa. This compound has enjoyed much atten- 51 R = H Kaempferol
52 R = OH Quercetin
tion recently because of its claimed usefulness in combat-
ing addiction, particularly to cocaine, although the basis
of its activity is not clear. Ibogaine was shown to release OCH3
OCH3
DA in isolated striatal tissue in mice [104], but it has also
CH3O O
been shown to inhibit the release of catecholamines by
blocking the nicotinic receptor [105] and to block NMDA CH3O
receptors. These activities preclude its usefulness in treat- OCH3 O
ing neurodegenerative disease, but much remains to be
done in the study of its antiaddictive potential. 53 Tangeretin

The tropane alkaloids, especially hyoscine (scopol-


amine) 4, have been used to treat PD since they antago- OH
OH
nise cholinergic activity at the muscarinic receptors in the
striatum and so increase DA activity. The naturally- HO O
OH
occurring alkaloids, found in various genera of the Solana-
O
ceae such as Atropa, Hyoscyamus, Datura and Duboisia,
OH OH
are not much used for this purpose, but synthetic deriva- O
tives and analogues such as benzatropine 48 are used. OH
These also inhibit DA reuptake, so increasing the levels of OH
DA and compensating for the DA deficiency associated
54 Epigallocatechin -3-gallate
with PD.

Fig. 18. Flavonoids with monoamine oxi-


PD: Monoamine Oxidase Inhibitors dase inhibitory activity.

Monoamine oxidase (MAO) is a major enzyme respon-


sible for the fast breakdown of DA and related com- stimulated the release of DA from striatal cells [106].
pounds at the synapse. Inhibitors of this enzyme, known These compounds would therefore have a double effect in
as MAOIs, cause a net increase in DA levels and, although helping improve DA levels, and this may underlie the
their major therapeutic use has been as antidepressants, reputed improvements in PD patients when the extract
they have potential use in PD. This has not generally been was taken.
realised because of the side effects associated with eleva- The role of flavonoids in the diet as important antioxi-
tion of peripheral DA levels. The ß-carboline alkaloids dant contributors has received much attention and their
harmane 49 and harmaline 50 are found in several tradi- neuroprotective properties because of this effect has been
tional medicine plant species, including Banisteriopsis demonstrated by several workers. However, they have
caapi (Spruce ex Griseb.) Morton (Malpighiaceae), a liana also been demonstrated to have MAOI activity and this
used in Brazil as an ingredient in the hallucinogenic drink has been proposed as part of the explanation of the use of
‘ayahuasca’. It is thought that the MAOI properties of the the common herb St John’s Wort, as an antidepressant
B. caapi constituents prevent metabolism of amines from [107]. This dual role has now been proposed for a variety
other plants used to make ayahuasca. Following reports of of flavonoids such as kaempferol 51 from the leaves of
the successful use of B. caapi root extracts for treating PD Ginkgo biloba, a widely used herbal product which has
patients in Ecuador, it was shown that, in addition to the been suggested as a preventative against neurodegenera-
MAOI properties, the alkaloids harmane and harmaline tion [108] (fig. 18). Quercetin 52 has also shown to inhibit

Natural Products and Neurotransmission Neurosignals 2005;14:6–22 19


MAO-B [109] and reverse the effects of induced catalep- cules can be subjected to ‘fine-tuning’ by chemical deriva-
sy, which mimics the bradykinesia often seen in PD [110]. tisation and synthesis of analogues to bind more closely
Tangeretin 53 has been shown to inhibit MAO-B and to and more exclusively to one type of receptor. However,
cross the blood-brain barrier in a rat model and conse- investigation of traditional plants has revealed classes of
quently reduce DA depletion, so it may have therapeutic molecules that have appreciable activity which is unlikely
potential [111]. A compound related to the flavonoids, the would have been predicted only from receptor studies.
polyphenol (–)-epigallocatechin-3-gallate 54, found in Regarding the treatment of the major diseases in-
green tea, has a similar polyvalent activity which may be volved, it can be said that chemotherapy of AD with cho-
sufficient to have a protective effect in PD and other con- linesterase inhibitors is unlikely to develop very much fur-
ditions [112]. Since most of these flavonoids occur in rea- ther since effective agents such as galantamine have been
sonably large amounts in common fruits and vegetables, introduced. Future trends could involve the use of a poly-
the question is raised as to whether the apparent increase valent ‘cocktail’ of drugs which act in different ways by
in incidence of neurodegenerative disease is related to mechanisms such as antioxidant and anti-inflammatory
some extent with the decline in consumption in the diet of activity and the inhibition of the formation of fibrillary
such foods in some sectors of the industrialized world. tangles and ß-amyloid plaques. Although the introduction
of L-DOPA and other dopaminergic compounds over the
last three decades has improved the condition of many
Conclusions sufferers of PD, the side effects and their unpredictability
of occurrence highlight the fact that more work is needed.
Several natural products useful compounds them- In this context also, the exploitation of compounds de-
selves, or have provided lead compounds, for present and rived from plants with other mechanisms, such as mono-
potential treatment of Alzheimer’s and Parkinson’s dis- amine oxidase inhibition, may provide a better treatment
eases. Some compounds have reached widespread clinical experience in due course. This may occur in extracts in
use, whilst the interest others at present lies more in their any case, and clinical trials should be carried out to follow
providing explanation for traditional uses of plants. Ra- up preliminary results, such as those observed with Mucu-
tional drug design according to the characteristics of the na bean extract, which indicate an advantage in using an
receptors involved is now a possibility, and natural mole- extract over a pure substance.

References

1 http://www.alzheimers.org.uk/Facts_about_de- 8 McGuffey EC: Alzheimer’s disease: An over- 14 Bratt AM, Kelly ME, Domeney AM, et al:
mentia/index.htm July 2004. view for the pharmacist. JAMA 1997;NS37: Acute and chronic arecoline: Effects of a sco-
2 http://www.who.int/mediacentre/factsheets/ 347–352. polamine-induced deficit in complex maze
fs218/en/ August 2004. 9 Perry E, Tomlinson E, Blessed G, et al: Correla- learning. Pharm Pharmacol Behav 1996;53:
3 Howes M-JR, Houghton PJ: Plants used in tion of cholinergic abnormalities with senile 713–721.
Chinese and Indian traditional medicine for plaques and mental test scores in senile demen- 15 Yang YR, Chang KC, Chen CL, et al: Arecoline
improvement of memory and cognitive func- tia. BMJ 1978;2:1457–1459. excites rat locus coerulus neurons by activating
tion. Pharm Biochem Behavior 2003;75:513– 10 Giacobini E: The cholinergic system in Alz- the M-2-muscarinic receptor. Chin J Physiol
527. heimer disease. Prog Brain Res 1990;84:321– 2000;43:23–28.
4 Howes M-JR, Perry NSL, Houghton PJ: Plants 332. 16 Soncrant TT, Raffaele KC, Asthana S, et al:
with traditional uses and activities, relevant to 11 Balfour DJK, Fagerström KO: Pharmacology Memory improvement without toxicity during
the management of Alzheimer’s disease and of nicotine and its therapeutic use in smoking chronic, low-dose intravenous arecoline in Alz-
other cognitive disorders. Phytother Res 2003; cessation and neurodegenerative disorders. heimers disease. Psychopharmacology 1993;
17:1–18. Pharmacol Ther 1996;72:51–81. 112:421–427.
5 Francis PT, Palmer AM, Snape M, et al: The 12 Whitehouse PJ, Kalaria RN: Nicotinic recep- 17 Meier E, Frederiksen K, Nielsen M, et al: Phar-
cholinergic hypothesis of Alzheimer’s disease: tors and neurodegenerative dementing dis- macological in vitro characterization of the
A review of progress. J Neurol Neurosurg Psy- eases: Basic research and clinical implications. arecoline bioisostere Lu 25-109-T, a muscarin-
chiatry 1999;66:137–147. Alzheimer Dis Assoc Disord 1995;9(suppl ic compound with M-1-agonistic and M-2/M-3
6 Desgranges B, Baron J-C, de la Sayette V, et al: 2):3–5. antagonistic properties. Drug Dev Res 1997;
The neural substrates of memory systems im- 13 Avery EE, Baker LD, Asthana S: Potential role 40:1–16.
pairment in Alzheimer’s disease. Brain 1998; of muscarinic agonists in Alzheimers disease. 18 Tlal LJ, Forreest M, Loft H, et al: Lu-25-109, a
121:611–631. Drugs Aging 1997;11:450–459. muscarinic agent, fails to improve cognition in
7 Förstl H, Hentschel F, Sattel H, et al: Age-asso- Alzheimer’s disease. Neurology 2000;54:421–
ciated memory impairment and early Alzhei- 426.
mer’ disease. Drug Res 1995;45:394–397.

20 Neurosignals 2005;14:6–22 Houghton/Howes


19 Wienrich M, Meier D, Ensinger HA, et al: 36 Small GW, Rabins RV, Barry PP, et al: Diag- 52 Peng WH, Hsieh MT, Wu CR: Effect of long-
Pharmacodynamic profile of the M-1 agonist nosis and treatment of Alzheimer’s disease and term administration of berberine on scopol-
talsclidine in animals and man. Life Sci 2001; related disorders. JAMA 1997;278:1363– amine-induced amnesia in rats. Jpn J Pharma-
22:2593–2600. 1371. col 1997;74:261–266.
20 Terry AV, Buccafusco JJ, Borsini F, et al: 37 Lopez S, Bastida J, Viladomat F, et al: Acetyl- 53 Moon TC, Murakami M, Kudo I, et al: A new
Memory-related performance by aged rhesus cholinesterase inhibitory activity of some Ama- class of COX-2 inhibitor, rutaecarpine from
monkeys administered the muscarinic M-1- ryllidaceae alkaloids and Narcissus extracts. Evodia rutaecarpa. Inflamm Res 1999;48:621–
preferring agonist, talsaclidine. Psychopharma- Life Sciences 2002;71:2521–2529. 625.
cology 2002;162:292–300. 38 Houghton PJ, Agbedahunsi JM, Adegbulugbe 54 Park CH, Kim S, Choi W, et al: Novel anticho-
21 Levin ED, Torry D: Acute and chronic nicotine A: Choline esterase inhibitory properties of linesterase and antiamnesic activities of dehy-
effects on working memory in aged rats. Psy- alkaloids from two Nigerian Crinum species. droevodiamine, a constituent of Evodia rurae-
chopharmacology 1996;123:88–97. Phytochemistry 2004;65:2893–2896. carpa. Planta Med 1996;62:405–409.
22 McCaleb R: Nature’s medicine for memory 39 Skolnick AA: Old herbal Chinese medicine 55 Haji A, Momose Y, Takeda R, et al: Increased
loss. HerbalGram 1990;23:15. used for fever yields possible new Alzheimer feline cerebral blood flow induced by dehy-
23 Sitaram N, Weingartner H, Gillin JC: Physo- disease therapy. JAMA 1997;277:776. droevodiamine hydrochloride from Evodia ru-
stigmine: Improvement of long-term memory 40 Wang Y-E, Yue D-X, Tang X-C: Anti-cholines- taecarpa. J Nat Prod 1994;57:387–389.
processes in normal humans. Science 1978; terase activity of huperzine A. Acta Pharm Si- 56 Krasowski MD, McGehee DS, Moss J: Natural
201:272–276. nica 1986;7:110–113. inhibitors of cholinesterases: implications for
24 Greig NH, Utsuki T, Yu QS, et al: A new thera- 41 Bai DL, Tang XC, He XC: Huperzine A, a adverse drug reactions. Can J Anaesth 1997;44:
peutic target in Alzheimer’s disease treatment: potential therapeutic agent for treatment of 525–534.
Attention to butyrylcholinesterase. Curr Med Alzheimers disease. Curr Med Chem 2000;7: 57 Perry N, Court G, Bidet N, et al: European
Res Opin 2001;17:159–165. 355–374. herbs with cholinergic activities: Potential in
25 Polinsky RJ: Clinical pharmacology of rivastig- 42 Lu W-H, Shou J, Tang X-C: Improving effect dementia therapy. Int J Geriatr Psychiatry
mine: A new-generation acetylcholinesterase of huperzine A in aged rats and adult rats with 1996;11:1063–1069.
inhibitor for the treatment of Alzheimer’s dis- experimental cognitive impairment. Acta 58 Perry NSL, Houghton PJ, Theobald A, et al: In
ease. Clin Ther 1998;20:634–647. Pharm Sinica 1988;9:11–15. vitro inhibition of human erythrocyte acetyl-
26 Grossberg G, Desai A: Review of rivastigmine 43 Zhou J, Zhang HY, Tang XC: Huperzine A cholinesterase by Salvia lavandulaefolia essen-
and its clinical applications in Alzheimer’s dis- attenuates cognitive deficits and hippocampal tial oil and constituent terpenes. J Pharm Phar-
ease and related disorders. Expert Opin Phar- neuronal damage after transient global isch- macol 2000a;52:895–902.
macother 2001;2:653–666. emia in gerbils. Neurosci Lett 2001;313:137– 59 Savelev S, Okello E, Perry NSL, et al: Synergis-
27 Spencer CM, Noble S: Rivastigmine – a review 140. tic and antagonistic interactions of anticholin-
of its use in Alzheimer’s disease. Drugs Aging 44 Xiao XQ, Zhang HY, Tang XC: Huperzine A esterase terpenoids in Salvia lavandulaefolia
1998;13:391–411. attenuates amyloid ß-peptide fragment 25–35- essential oil. Pharmacol Biochem Behav 2003;
28 Bores GM, Huger FP, Petko W, et al: Pharma- induced apoptosis in rat cortical neurons via 75:661–668.
cological evaluation of novel Alzheimer’s dis- inhibiting reactive oxygen species formation 60 Perry NSL, Houghton PJ, Jenner P, et al: Sal-
ease therapeutics: Acetylcholinesterase inhibi- and caspase-3 activation. J Neurosci Res 2002; via lavandulaefolia essential oil inhibits cholin-
tors related to galanthamine. Am Soc Pharma- 67:30–36. esterase in vivo. Phytomedicine 2002;9:48–51.
col Exp Ther 1996;277:728–738. 45 Zhou J, Tang XC: Huperzine A attenuates 61 Tildesley NTJ, Kennedy DO, Perry EK, et al:
29 Heinrich M, Teoh HL: Galanthamine from apoptosis and mitochondria-dependent cas- Salvia lavandulaefolia (Spanish sage) enhances
snowdrop – the development of a modern drug pase-3 in rat cortical neurons. FEBS Lett 2002; memory in healthy young volunteers. Pharma-
against Alzheimer’s disease from local Cauca- 526:21–25. col Biochem Behav 2003;75:669–674.
sian knowledge. J Ethnopharmacol 2004;92: 46 Zhang Z, Wang X, Chen Q, et al: Clinical effi- 62 Perry NSL, Bollen C, Perry EK, et al: Salvia for
147–162. cacy and safety of huperzine alpha in treatment dementia therapy: Review of pharmacological
30 Wilcock GK, Lilienfeld S, Gaens E, on behalf of mild to moderate Alzheimer disease, a place- activity and pilot tolerability clinical trial.
of the Galantamine International-1 Study bo-controlled, double-blind, randomised trial. Pharmacol Biochem Behav 2003;75:651–660.
Group: Efficacy and safety of galantamine in Chin Med J 2002;82:941–944. 63 Akhondzadeh S, Noroozian M, Mohammadi
patients with mild to moderate Alzheimer’s 47 Jiang H, Luo X, Bai D: Progress in clinical, M, et al: Salvia officinalis extract in the treat-
disease: Multicentre randomised controlled pharmacological, chemical and structural bio- ment of patients with mild to moderate Alz-
trial. BMJ 2000;321:1445–1449. logical studies of huperzine A: A drug of tradi- heimer’s disease: A double-blind, randomised
31 Wilkinson D, Murray J: Galantamine: A ran- tional Chinese medicine origin, in the treat- and placebo-controlled trial. J Clin Pharm
domised, double-blind, dose comparison in pa- ment of Alzheimer’s disease. Curr Med Chem Ther 2003;28:53–59.
tients with Alzheimer’s disease. Int J Geriatr 2003;10:2231–2252. 64 Yarnell E: Lemonbalm. Altern Complement
Psychiatry 2001;16:852–857. 48 Huang KC: The Pharmacology of Chinese Ther 1998;4:417–419.
32 Marcusson J, Bullock R, Gauthier S, et al: Gal- Herbs. Boca Raton, CRC Press, 1993. 65 Kennedy DO, Wake G, Savelev S, et al: Modu-
antamine demonstrates efficacy and safety in 49 Shigeta K, Ootaki K, Tatemoto H, et al: Poten- lation of mood and cognitive performance fol-
elderly patients with Alzheimer’s disease. Alz- tiation of nerve growth factor-induced neurite lowing acute administration of single doses of
heimer Dis Assoc Disord 2003;17S3:S86–S91. outgrowth in PC12 cells by a Coptidis rhizoma Melissa officinalis (Lemon balm) with human
33 Raskind MA, Peskind ER, Truyen L, et al: The extract and protoberberine alkaloids. Biosci CNS nicotinic and muscarinic receptor-bind-
cognitive benefits of galantamine are sustained Biotechnol Biochem 2002;66:2491–2494. ing properties. Neuropsychopharmacology
for at least 36 months: A long term extension 50 Hsieh MT, Peng WH, Wu CR, et al: The ame- 2003;28:1871–1881.
trial. Arch Neurol 2004;61:252–256. liorating effects of the cognitive-enhancing 66 Akhondzadeh S, Noroozian M, Mohammadi S,
34 Fulton B, Benfield P: Galantamine. Drugs Chinese herbs on scopolamine-induced amne- et al: Melissa officinalis extract in the treatment
Aging 1996;1:60–65. sia in rats. Phytother Res 2000;14:375–377. of patients with mild to moderate Alzheimer’s
35 Woodruff-Pak DS, Vogel RW, Wenk GL: Gal- 51 Kuznetsova LP, Nikol’skaya EB, Sochilina EE, disease: A double blind, randomised, placebo
antamine: Effect on nicotinic receptor binding, et al: Inhibition of human blood acetylcholines- controlled trial. J Neurol Neurosurg Psychiatry
acetylcholinesterase inhibition, and learning. terase and butyrylcholinesterase by some alka- 2003;74:863–866.
Proc Natl Acad Sci 2001;98:2089–2094. loids. J Evol Biochem Physiol 2002;38:35–39.

Natural Products and Neurotransmission Neurosignals 2005;14:6–22 21


67 Ryan MF, Byrne O: Plant-insect coevolution 81 Lo YC, Teng CM, Chen CF, et al: Magnolol 100 Stocchi F: Dopamine agonists in Parkinson’s
and inhibition of acetylcholinesterase. J Chem and honokiol from Magnolia officinalis protect disease –What is their role in early treatment?
Ecol 1988;14:1965–1975. rat heart mitochondria against lipid peroxida- CNS Drugs 1998;10:159–170.
68 Kuang P, Tao Y, Tian Y: Effect of radix Salviae tion. Biochem Pharmacol 1994;47:549–553. 101 Melzig MF, Putscher I, Henklein P, et al: In
miltiorrhizae on nitric oxide in cerebral isch- 82 Wang LM, Mineshita S: Preventive effects of vitro pharmacological activity of the tetrahy-
emic-reperfusion injury. J Tradit Chin Med Unsei-in and Oren-gedoku-to, Chinese tradi- droisoquinoline salsinol present in products
1996a;16:224–227. tional medicines, against rat paw oedema and from Theobroma cacao L. like cocoa and
69 Ren Y, Houghton PJ, Hider RC, et al: Novel abdominal constriction in mice. J Pharm Phar- chocolate. J Ethnopharmacol 2000;73:153–
diterpenoid acetylcholinesterase inhibitors macol 1996;48:327–331. 159.
from Salvia miltiorrhiza. Planta Med 2004;70: 83 Merchant C, Tang MX, Albert S, et al: The 102 Antkiewicz-Michaluk L, Wardas J, Michaluk
201–204. influence of smoking on the risk of Alzheimer’s J, et al: Protective effect of 1-methyl-1,2,3,4-
70 Chung YK, Heo HJ, Kim EK, et al: Inhibitory disease. Neurology 1999;52:1408–1412. tetrahydroisoquinoline against dopaminergic
effect of ursolic acid purified from Origanum 84 Ott A, Slooter AJC, Hofman A, et al: Smoking neurodegeneration in the extrapyramidal
majorana L. on the acetylcholinesterase. Mol and risk of dementia and Alzheimer’s disease structures produced by intracerebral injection
Cells 2001;11:137–143. in a population-based cohort study: The Rot- of rotenone. Int J Neuropsychopharmacol
71 Ghosal S, Lal R, Srivastava SK, et al: Immuno- terdam Study. Lancet 1998;351:1840–1843. 2004;7:153–163.
modulatory and CNS effects of sitoindosides 85 Salomon AR, Marcinowski KJ, Friedland RP, 103 Naoi M, Maruyama W, Nagy GM: Dopa-
IX and X, two new glycowithanolides from et al: Nicotine inhibits amyloid formation by mine-derived salsolinol derivatives as endog-
Withania somnifera. Phytother Res 1989;3: the ß-peptide. Biochemistry 1996;35:13568– enous monoamineoxidase inhibitors: Occur-
201–206. 13578. rence, metabolism and function in human
72 Schliebs R, Liebmann A, Bhattacharya SK, et 86 Whitehouse PJ, Kalaria RN: Nicotinic recep- brains. Neurotoxicology 2004;25:193–204.
al: administration of defined extracts from tors and neurodegenerative dementing dis- 104 Harsing LG, Sershen H, Lajtha A: Evidence
Withania somnifera (Indian ginseng) and shila- eases: Basic research and clinical implications. that ibogaine releases dopamine from the cy-
jit differentially affects cholinergic but not glu- Alzheimer Dis Assoc Disord 1995;9(suppl toplasmic pool in isolated mouse striatum. J
tamatergic markers in rat brain. Neurochem 2):3–5. Neural Transm 1994;96:215–225.
Int 1997;30:181–190. 87 Polya G: Biochemical Targets of Plant Bioac- 105 Mah SJ, Tang YM, Liauw PE, et al: Ibogaine
73 Bhattacharya SK, Kumar A, Ghosal S: Effects tive Compounds. London, Taylor & Francis, acts at the nicotinic acetylcholine receptor to
of glycowithanolides from Withania somnifera 2003. inhibit catecholamine release. Brain Res
on an animal model of Alzheimer’s disease and 88 Decker MW, Majchrzak MJ, Arneric SP: Ef- 1998;797:173–180.
perturbed central cholinergic markers of cogni- fects of lobeline, a nicotinic receptor agonist, 106 Schwarz MJ, Houghton PJ, Rose S, et al:
tion in rats. Phytother Res 1995;9:110–113. on learning and memory. Pharmacol Biochem Activities of extract and constituents of Ba-
74 Bhattacharya A, Ramanathan M, Ghosal S, et Behav 1993;45:571–576. nisteriopsis caapi relevant to parkinsonism.
al: Effect of Withania somnifera glycowithano- 89 http://www.parkinsons.org.uk/Templates/] Pharmacol Biochem Behav 2003;75:627–
lides on iron-induced hepatotoxicity in rats. 90 Kidd PM: Parkinson’s disease as multifactorial 633.
Phytother Res 2000;14:568–570. oxidative neurodegeneration: Implications for 107 Wagner H, Bladt S: MAO inhibition by frac-
75 Parihar MS, Hemnani T: Phenolic antioxi- integrative management. Altern Med Rev tions and constituents of Hypericum extract.
dants attenuate hippocampal neuronal cell 2000;5:502–529. Nervenheilkunde 1993;12:349–352.
damage against kainic acid induced excitotox- 91 Hussain G, Manyam BV: Mucuna pruriens 108 Sloley BD, Urichuk LJ, Morley P, et al: Iden-
icity. J Biosci 2003;28:121–128. proves more effective than L-DOPA in parkin- tification of kaempferol as a monoamineoxi-
76 Begum V, Sadique J: Long term effect of herbal son’s disease animal model. Phytother Res dase inhibitor and potential neuroprotectant
drug Withania somnifera on adjuvant induced 1997;11:419–423. in extracts of Ginkgo biloba leaves. J Pharm
arthritis in rats. Ind J Exp Biol 1998;26:887– 92 Vaidya RA, Sheth AR, Alookar SD, et al: The Pharmacol 2000;52:451–459.
882. inhibitory effect of the Cowhage plant – Mucu- 109 Singh A, Naidu PS, Kulkarni SK: Quercetin
77 Dhuley JN: Effect of some Indian herbs on na pruriens – and L-DOPA on chlorpromazine- potentiates L-Dopa reversal of drug-induced
macrophage functions in ochratoxin A treated induced hyperprolactinaemia in man. Neurol catalepsy in rats: Possible COMT/MAO inhi-
mice. J Ethnopharmacol 1997;58:15–20. India 1978;26:171–176. bition. Pharmacology 2003;68:81–88.
78 Hou YC, Chao PD, Chen SY: Honokiol and 93 Manyam BV, Dhanasekaran M, Hare TA: Ef- 110 Naidu PS, Kulkarni SK: Quercetin, a biofla-
magnolol increased hippocampal acetylcholine fect of antiparkinson drug HP-200 (Mucuna vonoid, reverses haloperidol-induced catalep-
release in freely-moving rats. Am J Chin Med pruriens) on the central monoaminergic neuro- sy. Meth Findings Exp Clin Pharmacol 2004;
2000;28:379–384. transmitters. Phytother Res 2004;18:97–101. 26:323–326.
79 Kuribara H, Kishi E, Hattori N, et al: The 94 Al-Motarreb A, Baker K, Broadley KJ: Khat: 111 Datla KP, Christidou MA, Widmer WW, et
anxiolytic effect of two oriental herbal drugs in Pharmacological and medical aspects and its al: Citrus flavonoid tangeretin accumulates in
Japan attributed to honokiol from Magnolia social use in Yemen. Phytother Res 2002;16: the brain and pre-treatment protects against
bark. J Pharm Pharmacol 2000;52:1425– 403–413. dopaminergic neuronal loss in a rat model of
1429. 95 Ismail M: Pers commun, 2004. Parkinson’s disease. Br J Pharmacol 2002;
80 Squires RF, Ai J, Witt MR, et al: Honokiol and 96 Bonucelli U: Comparing dopamine agonists in 135:350P.
magnolol increase the number of [3H]musci- Parkinson’s disease. Curr Opin Neurology 112 Mandel S, Weinreb O, Amit T, et al: Cell sig-
mol binding sites three-fold in rat forebrain 2003;16:S13–S19. naling pathways in the neuroprotective ac-
membranes in vitro using a filtration assay, by 97 Sit SY: Dopamine agonists in the treatment of tions of the green tea polyphenol (–)-epigallo-
allosterically increasing the affinities of low- Parkinson’s disease. Past, present and future. catechin-3-gallate: Implications for neurode-
affinity sites. Neurochem Res 1999;24:1593– Curr Pharm Design 2000;6:1211–1248. generative diseases. J Neurochem 2004;88:
1602. 98 Poewe W: Pergolide – a review of its clinical 1555–1569.
potential. Akt Neurol 1995;22:71–74.
99 Pastor P, Tolosa E: Cabergoline in the treat-
ment of Parkinson’s disease. Neurologia 2003;
18:202–209.

22 Neurosignals 2005;14:6–22 Houghton/Howes


Review

Neurosignals 2005;14:23–33 Received: July 28, 2004


Accepted after revision: September 20, 2004
DOI: 10.1159/000085383

Regulation of Neuroinflammation by
Herbal Medicine and Its Implications for
Neurodegenerative Diseases
A Focus on Traditional Medicines and Flavonoids

Kyoungho Suk
Department of Pharmacology, Pain and Neural Injury Research Center, School of Medicine, Kyungpook
National University, Daegu, Korea

Key Words Introduction


Inflammation  Neuroglia  Neurodegenerative disease 
Flavonoid  Neuroprotection  Microglia  Astrocyte  In neuroinflammation, microglia and astrocytes play
Central nervous system a critical role. Microglial cells are ubiquitously distrib-
uted in the central nervous system (CNS) and comprise
up to 20% of the total glial cell population in brain [1, 2].
Abstract Although the ontogeny of microglial cells has long been
Herbal medicine has long been used to treat neural debated, recent works using monoclonal antibodies spe-
symptoms. Although the precise mechanisms of action cific for microglial cells indicated that these cells are close-
of herbal drugs have yet to be determined, some of them ly related to monocytes and macrophages [3]. As the pri-
have been shown to exert anti-inflammatory and/or anti- mary immune effector cells in the CNS, microglial cells
oxidant effects in a variety of peripheral systems. Now, migrate to the site of tissue injury or inflammation, where
as increasing evidence indicates that neuroglia-derived they respond to invading pathogens or other inflamma-
chronic inflammatory responses play a pathological role tory signals [4, 5]. Like monocytes/macrophages, they
in the central nervous system, anti-inflammatory herbal also secrete inflammatory cytokines and toxic mediators
medicine and its constituents are being proved to be a which may amplify the neuroinflammatory responses [6,
potent neuroprotector against various brain patholo- 7]. Astrocytes form an intimately connected network with
gies. Structural diversity of medicinal herbs makes them neurons in the CNS, and they provide mechanical and
valuable source of novel lead compounds against thera- metabolic support for neurons [8]. The critical role of
peutic targets that are newly discovered by genomics, these cells in ion buffering and clearance of neurotrans-
proteomics, and high-throughput screening. mitters is also well established [9, 10]. Upon inflamma-
Copyright © 2005 S. Karger AG, Basel tory stimulation, astrocytes proliferate and produce di-
verse intercellular mediators such as nitric oxide (NO)
and tumor necrosis factor (TNF)- [11–13]. There is
growing evidence that inflammatory mediators produced
by activated astrocytes may be involved in the pathogen-
esis of various neurodegenerative diseases [10, 14]. Thus,

© 2005 S. Karger AG, Basel Kyoungho Suk


1424–862X/05/0142–0023$22.00/0 Department of Pharmacology, Pain and Neural Injury Research Center
Fax +41 61 306 12 34 Kyungpook National University School of Medicine
E-Mail karger@karger.ch Accessible online at: 101 Dong-In, Joong-Gu, Daegu, 700-422 (Korea)
www.karger.com www.karger.com/nsg Tel. +82 53 420 4835, Fax +82 53 256 1566, E-Mail ksuk@knu.ac.kr
Chemicals and drugs

Physical factors Infection Autoimmunity

Genetic disturbance
Nutritional
imbalance

Hypoxia
Tissue damage

Reversible Irreversible
damage damage
Fig. 1. Relationship between inflammation
and tissue injury. Tissue damage can be
triggered by a number of genetic or environ-
mental factors. While reversible tissue dam- Tissue repair,
functional Inflammation Diseases
ages could be repaired for a functional re- recovery
covery, irreversible damages associated Regulation No regulation,
with inappropriately controlled inflamma- chronic inflammation
tion (chronic inflammation) could lead to
diseases.

the activation of astrocytes and ensuing production of serves to contain and destroy the infection or damaging
toxic inflammatory mediators may need to be tightly reg- agents, and to prevent continued tissue damage and initi-
ulated. Activation of inflammatory cells in CNS (micro- ate repair processes to restore normal function. This rap-
glia or astrocytes) may be intended to protect neurons at id response is known as acute inflammation [20]. The
first. More frequently, however, activation of these neu- toxic reactions, which are employed to destroy infectious
roglial cells and inflammatory products derived from organisms or protect host, also paradoxically have the
them have been implicated in neuronal destruction com- capacity to injure host tissues. If these toxic responses are
monly observed in various neurodegenerative diseases not tightly regulated, tissue injury may predominate over
[7]. Thus, our understanding of pathogenesis of neurode- tissue protection and repair, thereby leading to inflamma-
generative diseases may be enhanced by elucidation of tory diseases (fig. 1). The characteristics of the inflamma-
the molecular mechanism underlying the regulation of tory response include localized changes within the dam-
neuroglial activation. Among many endogenous or exog- aged tissue such as the followings: (1) the release of pre-
enous factors that regulate neuroglial activation and re- formed inflammatory mediators from intracellular stores;
sulting neuroinflammation [15], herbal medicine has re- (2) the initiation of reaction cascade through the activa-
cently drawn much attention due to its potent inhibitory tion of soluble plasma components; (3) the new synthesis
effects on inflammatory responses and neuroprotective of inflammatory mediators such as eicosanoids and cyto-
activity [16, 17]. A central role of microglia and astrocytes kines, and (4) resolution of the inflammatory response.
in neuroinflammation (and potentially neurodegenera- The acute inflammatory response is beneficial to the or-
tion) and a regulatory effect of herbal medicine on the ganism in that it helps to deal with potentially dangerous
inflammatory activation of the neuroglia will be discussed microorganisms. However, inflammation does cause
in this review. some degree of damage to surrounding tissues. Reactive
oxygen species (ROS), reactive nitrogen species (RNS),
prostanoids, leukotrienes, and hydrolytic enzymes pro-
Inflammation and Tissue Injury duced by neutrophils, macrophages, and monocytes may
all play a role in mediating inflammation. Persistence of
Injury, trauma or infection induce a series of complex infection or defective resolution of inflammatory reaction
and interconnected reaction sequences, initiated at the results in chronic inflammation where severe tissue dam-
site of tissue damage [18, 19]. This sequence of reaction age may occur. Although inflammation is normally a self-

24 Neurosignals 2005;14:23–33 Suk


Physiological Neuro-
Intrinsic neuroprotection regeneration
Fig. 2. Neuroprotective versus neurotoxic ac- or
tivities of neuroglia. Neuroglial activation is extrinsic
factors Apoptosis (self-regulation)
induced by a variety of intrinsic or extrinsic
factors. Physiological neuroglial activation is Neuroglial
beneficial and designed to ameliorate damage activation
and stress. In contrast, pathological activation
of neuroglia leads to neurotoxicity. Apoptotic Pathological Neuro-
neurotoxicity degeneration
elimination of overactivated neuroglia repre-
sents a self-regulatory mechanism. When the
auto-regulation is at work, physiological neuro- Apoptosis (self-regulation)
protection may allow a neuroregeneration. On
the contrary, a defective regulation may cause
neurodegeneration.

limiting event and its benefit outweighs the minor tissue roglia will either nurse the injured neurons into
damage it causes, abnormal activation of the immune or regeneration or kill them if they are not viable. These
inflammatory system has the potential to provoke a dev- types of neuroglial responses are considered to represent
astating response [21]. In gout, for example, elevated con- normal physiological and neuroprotective responses. In
centration of uric acid in the blood leads to precipitation contrast, some processes that are chronic in nature per-
of sodium urate crystal within joints which triggers in- sistently activate neuroglia eventually causing a failure in
flammation by a variety of mechanisms. Another striking their physiological ability to maintain homeostasis. This
consequence of abnormal inflammatory response is auto- could have detrimental consequences and may lead to
immune diseases such as systemic lupus erythematosus, bystander damage due to neuroglial dysfunction. In this
rheumatoid arthritis, autoimmune vasculitis, dermato- scenario, neuroglia exert neurotoxic effects through the
myositis, chronic autoimmune gastritis, and myasthenia secretion of a variety of toxic inflammatory mediators.
gravis. Tissue-damaging chronic inflammatory response Thus, although activation of neuroglial cells may be in-
may also occur in CNS, where main inflammatory cells tended to protect neurons, inflammatory products de-
are microglia and astrocytes instead of monocytes/mac- rived from activated neuroglia may also be implicated in
rophages or neutrophils in periphery [22–24]. neuronal injury, potentially leading to neurodegenerative
diseases [7]. These deleterious effects of neuroglial activa-
tion may be exacerbated by the failure of auto-regulatory
Neuroglia (Microglia, Astrocytes), mechanisms of neuroglia. Recently, activated macro-
Neuroinflammation, and Neurodegeneration phages, whose functions are closely related to microglia,
have been shown to undergo apoptosis [26–28]. It has
Microglia and astrocytes are essential for ensuring been suggested that the apoptosis of activated macro-
proper functioning of neurons. They are quick to inter- phages is one mechanism whereby an organism may reg-
vene when neurons become injured or stressed. As they ulate immune and inflammatory responses involving
are sentinels of neuron well-being, pathological impair- macrophages [28]. It has been recently demonstrated that
ment of microglia or astrocytes could have devastating a similar regulatory mechanism exists for microglial cells
consequences for brain function. Nevertheless, there is [29, 30] and astrocytes [31] as well. Microglial cells and
still a debate over neuroprotective and neurotoxic func- astrocytes underwent apoptosis upon inflammatory acti-
tions of these neuroglial cells [22, 25] (fig. 2). It is assumed vation in a manner similar to activation-induced cell
that neuroglial activation is largely determined by neuro- death (AICD) of lymphocytes [30, 31]. AICD is an active
nal signals. Acute injury causes neurons to generate sig- process. T and B lymphocytes undergo AICD as an auto-
nals that inform neuroglia about the neuronal status. De- regulatory mechanism for the body to remove unwanted
pending on how severe a degree of neuronal injury, neu- activated cells after making appropriate use of them [32,

Herbal Medicine and Neuroinflammation Neurosignals 2005;14:23–33 25


33]. Compared to lymphocytes, neuroglial cells in CNS diators produced by activated neuroglia. Recently, up-
are not well studied in this respect. Now, as results in this regulated Bcl-xL expression has been detected in reactive
and other laboratories indicated that neuroglial cells microglia of patients with neurodegenerative diseases
might be under the control of a similar regulatory mech- [41]. Authors proposed that high level of Bcl-xL protein
anism [29–31, 34–37], further investigation is warranted might render microglia more resistant to cytotoxic envi-
to better understand the molecular mechanism(s) of neu- ronment such as areas of neurodegeneration. Expression
roglial AICD and its physiological significance. Neverthe- of anti-apoptotic Bcl-2 protein has been also associated
less, it has been shown that, in contrast to AICD of T with aged brain and neurodegenerative diseases [42]. An
lymphocytes where Fas-FasL interaction plays a central importance of the physiological regulation of neuroglial
role, neither Fas-FasL interaction nor TNF- is impor- activation by AICD is supported by these previous re-
tant in AICD of microglial cells [30]. Instead, NO pro- ports.
duced by activated neuroglial cells themselves was the Recent studies focused on the possible role of neuro-
major cytotoxic mediator [30, 31]. However, the presence glia in causing neurodegeneration. Convincing evidence
of NO-independent cytotoxic mechanism has been also from in vitro studies pointed to the neurotoxic role of
suggested [38, 39]. neuroglia during traumatic or ischemic brain injury [4]
Elimination of activated neuroglial cells by apoptosis and AD pathogenesis [43]. Supernatants obtained from
could be an important mechanism whereby undesirable neuroglial cell cultures kill cultured neurons. Such super-
effects of long-term neuroglial activation can be mini- natants contain various neurotoxic substances which in-
mized. Inflammatory mediators that are produced by ac- clude glutamate, NO, ROS, inflammatory cytokines, as
tivated neuroglia in CNS may have harmful effects on well as yet unidentified neurotoxins [44, 45]. Production
neurons or other neuroglial cells that they originally in- of these neurotoxins by neuroglia is enhanced by treat-
tended to protect [6, 7]. Thus, in various neurodegenera- ment with inflammatory stimuli such as lipopolysaccha-
tive diseases involving chronic neuroglial activation, neu- ride (LPS) and/or interferon (IFN)-. Paradoxically, oth-
roglial functions seem to play a more significant role in er investigators have shown that neuroglia-conditioned
mediating diseases than in the protection of neurons. Ac- media promote neuronal survival [46]. Thus, the balance
cording to the model of activation-induced apoptosis of of neurotoxic and neurotrophic effects of neuroglia ap-
neuroglial cells, inflammatory signals that activate neu- pears to depend on the nature of the experimental para-
roglia may also initiate internal death program [38, 40]. digm used. In traumatic brain injury where neuronal re-
One interesting question that can be raised then is how generation may occur, neuroglial secretory products
neuroglial cells could survive the inflammatory activa- might help to promote regenerative efforts by injured, but
tion. It should be kept in mind that neuroglial cells in vivo surviving, neurons. However, a situation may be differ-
are heterogeneous and interact with other neuroglial cells ent in neurodegenerative diseases such as Alzheimer’s
as well as neurons. There is also growing evidence that disease (AD) or human immunodeficiency virus (HIV)-
activated neuroglial cells proliferate in vivo as one way of associated dementia, where functionally compromised
replenishment [1]. Thus, not all neuroglial cells may re- neuroglia may produce neurotoxins, thereby resulting in
spond to the inflammatory signals in the same fashion. neuronal damage. There is considerable evidence from
Upon inflammatory activation, individual neuroglial postmortem examinations of AD brains that auto-de-
cells in heterogeneous population may either undergo structive mechanisms are at work, which could in part be
AICD or return to the resting state via other regulatory responsible for the neurodegeneration [47, 48].
mechanisms depending on the specific microenviron-
ment under which they react to the signals. Although
many of activated neuroglial cells may be eliminated, Neuroglia as a Target of Pharmacological
some would survive to be deactivated. Whatever the Intervention
mechanism of down-regulation is, this may be an excel-
lent auto-regulatory system for the neuroglial activation. Considering neuroglial activation as a common fea-
One can easily imagine pathological situations where this ture in many neuropathologies, and keeping in mind that
type of auto-regulatory mechanism goes wrong. Failure overactivation of neuroglia can have neurotoxic out-
of the auto-regulation of ‘over-activated’ neuroglial cells comes, it is reasonable to assume that manipulation
may result in pathological destruction of bystander cells of neuroglial activation could serve future clinical ap-
(neurons and other neuroglial cells) exposed to toxic me- proaches [49]. Although treatment of the primary events

26 Neurosignals 2005;14:23–33 Suk


in neurodegenerative diseases would still be the preferred organic constituents. Similarly, a host of environmental
intervention, this may not always be possible. Brain or factors, including soil, altitude, and seasonal variation in
spinal cord injury is a sudden event that is followed by weather, may affect the levels of components in any given
secondary cascades of destruction. Invading macrophages batch of an herb. Because of these multiple factors that
and intrinsic neuroglia in brain may carry a significant affect concentration of active ingredients in the final
portion of these cascades of reaction. Now, there is grow- herbal product, standardization is inevitable, in which
ing evidence that toxic mediators produced by activated certain unique chemical components of the herbs known
neuroglial cells might be involved in the pathogenesis of as markers are identified and the production process is
various neurodegenerative diseases such as Parkinson’s altered to achieve a consistent level of these markers in
disease (PD), AD, and HIV-associated dementia [6, 7, every final batch of the herbal product. The ten most com-
47]. Thus, it is of great interest to find a means to modu- monly used herbs in United States are echinacea, garlic,
late neuroglial activation and CNS inflammatory re- Ginkgo biloba, saw palmetto, ginseng, grape seed extract,
sponses for the therapeutic interventions against these green tea, St. John’s wort, bilberry, and aloe [50]. The
neurodegenerative diseases. Based on understanding of medical use of herbs in their natural and unprocessed
intracellular signaling pathways that are specific for acti- form began when mankind first noticed that certain plants
vated neuroglia, a temporary inhibition of signaling mol- altered particular body functions. Now, much informa-
ecules or protein-protein interaction associated with sig- tion exists about the historical use and effectiveness of
naling pathways would probably allow for a rather selec- botanical products. Unfortunately, however, the quality
tive effect on activated neuroglia, while respective of this information is extremely variable. Necessary is
functions in other cell types are unaffected. Elucidation evidence-based approach to the pharmacology and clini-
of the intracellular key events that drive neuroglial acti- cal efficacy of the herbal medicine.
vation could provide new routes for drug development
[49]. Alternatively, potentially harmful products of neu-
roglia could be neutralized to limit undesired conse- Herbal Medicine against CNS Disorders
quences for CNS cells and tissues. Whether it is a direct
inhibition of neuroglial activation or indirect suppression In traditional practices of Chinese medicine, numer-
of neuroglia-derived toxic inflammatory mediators, a bet- ous plants have been used to treat stroke and cognitive
ter understanding of neuroglial biology and selective ma- disorders, including neurodegenerative diseases such as
nipulation of neuroglial activation processes represent a AD [52, 53]. Neurodegenerative diseases and brain inju-
promising goal for developing novel neuroprotective ries resulting from stroke are the major and increasing
strategies. public health problem in both developed and developing
countries worldwide [54]. It is believed that traditional
Chinese medicines (TCM) are effective, with few or no
Medical Use of Herbs side-effects. There are more than 120 traditional medi-
cines in use for the therapy of CNS disorders in Asian
Herbs are generally defined as any form of plants or countries. Some of their therapeutic effects have been
plant products, including leaves, stems, roots, and seeds confirmed by recent clinical studies. An ethnopharmaco-
[50, 51]. Herbal products may contain a single herb or logical approach has provided a potentially rich source
combinations of several different herbs that are thought for drug discovery and development [55]. Many drugs
to have complementary effects. Herbal products are usu- currently available in Western medicine were originally
ally extracts of the plants, which are obtained by boiling isolated from plants. Although a large number of com-
or percolating the herbs in water, alcohol, or other or- pounds have been isolated from TCMs, most of these
ganic solvents to release biologically active ingredients of resources have not yet been fully characterized for phar-
the plants. Herbal products contain complicated mix- macological purposes. Some of the TCMs used in stroke
tures of organic chemicals, which may include fatty acids, therapy include Ledebouriella divaricata, Scutellaria
sterols, alkaloids, flavonoids, glycosides, saponins, tan- baicalensis, Angelica pubescens, Morus alba, Salvia milti-
nins, terpenes and so forth. It is often difficult to deter- orrhiza, Uncaria rhynchophylla, and Ligusticum chuan-
mine which component(s) of the herb has biological activ- xiong [52]. Among these, S. baicalensis and U. rhyncho-
ity. In addition, the processing of herbs, such as heating phylla have been found to confer neuroprotection against
or boiling, may alter the pharmacological activity of the transient global ischemia [56, 57]. S. baicalensis is one of

Herbal Medicine and Neuroinflammation Neurosignals 2005;14:23–33 27


Inflammatory stimuli

Fig. 3. Herbal medicine as a neuroprotector


that targets neuroglial inflammation. Rest-
ing neuroglia can be activated by inflamma-
tory stimuli such as LPS, IFN- and TNF-. Neurotoxic
Resting Activated
Activated neuroglia secrete a variety of in- inflammatory
neuroglia neuroglia
mediators
flammatory mediators including ROS, ni-
tric oxide, TNF-, and IL-1, which cause
neuronal injury (thereby resulting in neuro-
degeneration). Herbal medicine may be Herbal medicine Neurodegeneration
neuroprotective by blocking the inflamma-
tory activation of neuroglia.

the most widely used herbal medicines against bacterial genase-2 in hippocampus at 24 h after ischemia was sig-
infections of the respiratory and gastrointestinal tract, nificantly inhibited at both mRNA and protein levels.
and various inflammatory diseases. The herb has anti- Furthermore, U. rhynchophylla extract inhibited TNF-
pyretic, antibacterial, and antihypertensive properties. and NO production in BV-2 mouse microglial cells in vi-
The main components of S. baicalensis – baicalin, baica- tro in a manner similar to what has been observed with
lein, and wogonin – have been previously shown to exert S. baicalensis extract. These anti-inflammatory actions of
anti-inflammatory effects [58–60]. Based on the use of S. U. rhynchophylla extract (and other herbal medicines)
baicalensis for the treatment of stroke in traditional ori- may contribute to its neuroprotective effects (fig. 3).
ental medicine, neuroprotective effects of S. baicalensis Ginkgo leaf extracts have been primarily used for the
have been evaluated after transient global ischemia using treatment of dementia and neurosensory problems [51,
rat 4-vessel occlusion model [56]. Methanol extracts from 53, 61]. They contain terpenoids (ginkgolides and bi-
the dried roots of S. baicalensis (0.1–10 mg/kg) adminis- lobalides) and flavonoids. Administration of ginkgo ex-
tered intraperitoneally significantly protected CA1 neu- tracts (EGb 761) has shown biological activities relevant
rons against 10 min transient forebrain ischemia as dem- to the treatment of CNS disorders [61]. Favorable effects
onstrated by measuring the density of neuronal cells have been observed on cerebral circulation and neuronal
stained with cresyl violet. The neuroprotective effects of cell metabolism. The extract was also neuroprotective
S. baicalensis observed in vivo was explained in part by against -amyloid- and NO-induced toxicity [62, 63].
its inhibitory effects on microglial TNF- and NO pro- These effects have been attributed, in part, to platelet-ac-
duction as well as protection of nerve growth factor tivating factor antagonism of the ginkgolides [64] and the
(NGF)-differentiated PC12 cells from hydrogen peroxide free radical scavenging and anti-oxidant properties of the
toxicity in vitro. U. rhynchophylla also exerted neuropro- flavonoids [65]. In addition to Ginkgo biloba, Huperzia
tective effects against transient global ischemia [57]. In serrata, Lycoris radiata, Magnolia officinalis, and Poly-
traditional Oriental medicine, U. rhynchophylla has been gala tenuifolia have been used for improvement of mem-
used to lower blood pressure and to relieve various neu- ory and cognitive function.
rological symptoms. However, scientific evidence related
to its effectiveness or precise modes of action has not been
available. Methanol extract of U. rhynchophylla admin- Plant Flavonoids as a Neuroprotector:
istered intraperitoneally (100–1,000 mg/kg at 0 and Inhibition of Neuroinflammation
90 min after reperfusion) significantly reduced the death
of hippocampal CA1 neurons following the transient Flavonoids are a group of low molecular weight poly-
forebrain ischemia. Measurement of neuronal cell den- phenolic compounds of plant origin, many of which alter
sity in CA1 region at 7 days after ischemia by Nissl stain- metabolic processes and have a positive impact on health
ing revealed more than 70% protection in U. rhyncho- [66]. They exhibit a variety of biological activities such
phylla-treated rats compared to saline-treated animals. In as anti-inflammatory, anti-oxidant, anti-viral, and anti-
U. rhynchophylla-treated animals, induction of cyclooxy- tumor actions [67, 68]. Wogonin (5,7-dihydroxy-8-me-

28 Neurosignals 2005;14:23–33 Suk


Sham Ischemia Ischemia Wogonin

Ischemia

Ischemia
Wog.
No ischemia

Sham
300 Ischemia
(cell number/mm2)
Neuronal density

200 iNOS

1.0 4.5 1.2


100

TNF
0
Sham Saline 0.5 1 10
g Wogonin (mg/kg) h 1.0 3.3 1.8

Fig. 4. Neuroprotective effects of wogonin against experimental staining and cell counting. Asterisks indicate statistically significant
brain injury. a–f Treatment of experimental animals with wogonin differences from saline-treated ischemic group (p ! 0.05). Sham =
(10 mg/kg i.p., 0 and 90 min right after 10 min ischemia and reper- Sham-operated animals (n = 7); saline = saline-treated animals fol-
fusion) conferred neuroprotection by markedly reducing the num- lowing ischemia (n = 7); wogonin = wogonin-treated animals fol-
ber of damaged pyramidal cells in the CA1 subfield. Representative lowing ischemia (n = 3). h Wogonin inhibited expression of inflam-
photomicrographs of cresyl violet-stained hippocampal regions of matory mediators following the ischemic brain injury. At 4 days
sham-operated animals (a, d) or animals that had been subjected after forebrain ischemia, the expression of iNOS and TNF- was
to 10 min ischemia followed by treatment with either saline (b, e) assessed by RT-PCR analysis of hippocampal tissue followed by
or 10 mg/kg of wogonin (c, f). Boxed regions in a, b, and c (!40) Southern blot analysis using sequence-specific oligonucleotide
are shown in d, e, and f (!200), respectively. Scale bar is 100 m. probes. Wogonin (10 mg/kg) markedly reduced the ischemic induc-
g The neuroprotective effect of wogonin was dose dependent. Ei- tion of iNOS and TNF- . Results are representative of three inde-
ther saline or wogonin (0.5, 1 and 10 mg/kg) was intraperitoneally pendent experiments. The numbers indicate a fold induction of the
administered into animals following 10 min ischemia. Seven days gene expression normalized to GAPDH as determined by densito-
later, neuronal cell density in CA1 region was measured by Nissl metric analysis of Southern blot of RT-PCR products.

Herbal Medicine and Neuroinflammation Neurosignals 2005;14:23–33 29


Table 1. Effect of flavonoid wogonin on the histological changes in hibited inflammatory activation of cultured brain micro-
hippocampus after kainate injection glia by diminishing LPS-induced TNF-, IL-1, and NO
production. Wogonin inhibited NO production by sup-
Experimental animal groups Histology scores1
pressing iNOS induction and NF-B activation in mi-
Saline (n = 3) 0 croglia. Inhibition of inflammatory activation of micro-
Kainate (n = 5) 3.1180.452 glia by wogonin led to the reduction in microglial cyto-
Kainate + wogonin, 1 mg/kg (n = 5) 2.6780.37 toxicity toward co-cultured PC12 cells, supporting a
Kainate + wogonin, 10 mg/kg (n = 6) 2.2580.322 neuroprotective role for wogonin in vitro. The neuropro-
To examine the neuroprotective effect of wogonin in vivo, ex- tective effect of wogonin was further demonstrated in
citotoxic neuronal injury was induced by systemic administration vivo using two experimental brain injury models; tran-
of kainate (30 mg/kg i.p.). Injection of kainate induced a severe sient global ischemia by 4-vessel occlusion (fig. 4) and
neuronal cell death in CA1 and CA3 of hippocampus. Pretreatment excitotoxic injury by systemic kainate injection (table 1).
with wogonin (10 mg/kg i.p., 60 min prior to kainate injection) sig-
nificantly attenuated the hippocampal cell death both in CA1 and In both animal models, wogonin conferred neuroprotec-
CA3 as determined by histological scoring. tion by attenuating the death of hippocampal neurons,
Values represent mean 8 SEM. and the neuroprotective effect was associated with inhibi-
1
Damage or loss of hippocampal neurons was assessed by Nissl tion of the inflammatory activation of microglia. Hippo-
staining at 2 days after kainate administration with or without campal induction of inflammatory mediators such as
wogonin pretreatment. Histological damage was scored as follows:
0 = no damage; 1 = occasional injured neurons in CA1 or CA3; iNOS and TNF- was reduced by wogonin in the global
2 = small area (!10%) with neuronal damage or loss in CA1 or CA3; ischemia model (fig. 4), and microglial activation was
3 = greater area (10–50%) with neuronal damage of loss in CA1 or markedly down-regulated by wogonin in the kainate in-
CA3; 4 = extended (150%) neuronal damage of loss in both in CA1 jection model as judged by microglia-specific isolectin B4
and CA3.
2 staining. A similar neuroprotective activity has been
Statistically significant differences among each other (p <
0.05). demonstrated with baicalein in rats [Kim et al., unpub-
lished results] as well as in gerbils [79]. Baicalein attenu-
ated NO production and apoptosis of LPS-activated, but
not IFN--activated, BV-2 mouse microglial cells as well
as rat primary microglia cultures [80]. The inhibition of
thoxyflavone) and baicalein (5,6,7-trihydroxyflavone) are NO production by baicalein was due to the suppression
flavonoids derived from the root of S. baicalensis. These of iNOS induction. Moreover, baicalein inhibited LPS-
flavonoids have been shown to exert various anti-inflam- induced NF-B activity in BV-2 cells without affecting
matory activities in vitro as well as in vivo. Wogonin in- caspase-11 activation, interferon regulatory factor (IRF)-1
hibited LPS-induced production of NO [60, 69] and pros- induction, or signal transducer and activator of tran-
taglandin E2 [70] in macrophages. Wogonin inhibited scription (STAT)-1 phosphorylation. IRF-1 and STAT-1
monocyte chemotactic protein-1 gene expression in hu- are central components of IFN- signaling [81]. Taken
man endothelial cells [71]. It also inhibited TPA-induced together, flavonoids such as wogonin and baicalein seem
cyclooxygenase-2 expression and skin inflammation in to exert their neuroprotective effects by inhibiting mi-
mice [72]. Moreover, wogonin showed free radical scav- croglial activation, which is a critical component of patho-
enging and anti-oxidant activities [73–75]. In the CNS, genic inflammatory responses in neurodegenerative dis-
however, little information is available about its effects eases. These findings emphasize the importance of herb-
on glial cells and neurons. Gao et al. [73, 76] demonstrat- al medicines and their constituents as an invaluable
ed neuroprotective effects of four flavonoids from S. bai- source for the development of novel neuroprotective
calensis, including wogonin, in cultured human neuro- drugs. In neurodegenerative diseases, a pathogenic role
blastoma cells. Recently, it has been demonstrated that of uncontrolled microglial activation is widely accepted
wogonin inhibits NO production and inducible NO syn- at present [7]. In search of neuroprotective agents, now it
thase (iNOS) induction in cultured rat astrocytes [77], is time to focus on killer cells (microglia and astrocytes)
suggesting that the flavonoid may act as an anti-inflam- instead of killed cells (neurons); eliminating or at least
matory agent in CNS as well. This was confirmed by a suppressing killer microglial activation will provide a bet-
recent study where wogonin has been shown to be neuro- ter chance for neuroprotection compared to just salvaging
protective against experimental brain injury by inhibiting dying neurons. Identification of a potent neuroprotector
inflammatory activation of microglia [78]. Wogonin in- from natural source that inhibits the killer cell activity

30 Neurosignals 2005;14:23–33 Suk


will certainly instigate further investigations in the re- which in turn leads to further inflammation. The benefi-
lated areas, which will ultimately lead to the successful cial versus detrimental effect of ROS and RNS is tightly
development of novel neuroprotective drugs based on fla- regulated by antioxidant defense systems, whereas neu-
vonoids or other constituents of the medicinal herbs. roinflammation is controlled by various endogenous me-
diators as well as by auto-regulatory apoptosis of inflam-
matory cells [30, 31]. Inflammation and generation of
Other Components of Herbal Medicine and ROS or RNS seem to be interconnected physiological re-
Other Mechanisms of Neuroprotection sponses, which may also have pathological implications
if left uncontrolled. This is supported by the findings that
As mentioned above, herbal extracts contain compli- many herbal extracts and their components with neuro-
cated mixtures of organic chemicals, including fatty ac- protective activities exert both anti-inflammatory and an-
ids, sterols, alkaloids, flavonoids, glycosides, saponins, tioxidant effects at the same time [56, 65, 95, 96].
tannins, and terpenes. Flavonoids are not the only com-
ponent possessing neuroprotective effects [82]. Anti-de-
mentia effects of galanthamine, an alkaloid widely occur- Conclusions
ring in Amaryllidaceous plants, have been well demon-
strated in a variety of animal models [83–85]. Acute and Activation of microglia and astrocytes plays a pivotal
chronic treatment with galanthamine significantly im- role in the initiation and progression of various neurode-
proved the impairment of learning, short-term and spa- generative diseases. Inhibition of the neuroglial activa-
tial memory. The p-hydroxybenzyl alcohol and gastrodin tion may provide an effective therapeutic intervention
are the active ingredients isolated from Gastrodia elata that alleviates the progression of the neurodegenerative
roots, and they have been shown to possess anti-amnesic diseases. Herbal medicine, especially their flavonoid con-
activities in experimental animals [86–88]. Huperzine A, stituents, may be a useful candidate for such a therapeu-
a sesquiterpene alkaloid purified from the Chinese me- tic approach. Continual investigation of the mechanisms
dicinal herb Huperia serrata, exhibits a broad range of underlying neuroglial activation, regulation of neuroin-
neuroprotective actions [89]. Huperzine A ameliorated flammation, modulatory role of herbal medicine in these
learning and memory impairments and improved spatial processes would not only lead to the discovery of novel
working memory. Ginsenosides (ginseng saponins), as neuroprotective agents based on medicinal herbs, but also
the major active constituents of ginseng, are another ex- help to understand complex pathophysiology of neurode-
ample of herb components with potent neuroprotective generative diseases.
effects [90–92]. The cellular and molecular mechanisms
underlying the neuroprotective effects of various compo-
nents of herbal extracts may be as diverse as the plants Acknowledgement
which these components were isolated from. Although
This work was supported by the Korea Research Foundation
this review has been focused on the role of neuroinflam-
Grant (KRF-2004-000-E00058). This work was also supported by
mation in neurodegenerative diseases and its inhibition the Neurobiology Research Program from the Korea Ministry of
by neuroprotective herbs, the antioxidant activity of Science and Technology (2004-01323).
herbal extracts is certainly another important aspect of
neuroprotection [93, 94]. A variety of herbal extracts and
their components have been demonstrated to exert neu-
roprotective effects associated with antioxidant activi-
ties, either by directly stimulating antioxidant response
genes or by potentiating the bodies’ own natural antioxi-
dant defense systems. Modulation of neuroinflammation
and the antioxidant activity are not mutually exclusive
mechanisms of action. ROS and RNS can be generated
during inflammatory responses. These compounds func-
tion as important signal-transducing messengers or as an
effector to kill invading microorganisms. High concentra-
tions of ROS or RNS, however, may cause tissue injury,

Herbal Medicine and Neuroinflammation Neurosignals 2005;14:23–33 31


References

1 Gehrmann J, Matsumoto Y, Kreutzberg GW: 20 Rankin JA: Biological mediators of acute in- 36 Yang MS, Park EJ, Sohn S, Kwon HJ, Shin
Microglia: Intrinsic immuneffector cell of the flammation. AACN Clin Issues 2004; 15: 3– WH, Pyo HK, Jin B, Choi KS, Jou I, Joe EH:
brain. Brain Res Brain Res Rev 1995;20:269– 17. Interleukin-13 and -4 induce death of activated
287. 21 Halliwell B: Oxygen radicals, nitric oxide and microglia. Glia 2002;38:273–280.
2 Gremo F, Sogos V, Ennas MG, Meloni A, Per- human inflammatory joint disease. Ann 37 Takano K, Nakamura Y, Yoneda Y: Micro-
sichini T, Colasanti M, Lauro GM: Features Rheum Dis 1995;54:505–510. glial cell death induced by a low concentration
and functions of human microglia cells. Adv 22 Nakajima K, Kohsaka S: Microglia: Activation of polyamines. Neuroscience 2003; 120: 961–
Exp Med Biol 1997;429:79–97. and their significance in the central nervous 967.
3 Stoll G, Jander S: The role of microglia and system. J Biochem (Tokyo) 2001; 130: 169– 38 Lee J, Hur J, Lee P, Kim JY, Cho N, Lee MS,
macrophages in the pathophysiology of the 175. Kim SY, Kim H, Suk K: Dual role of inflam-
CNS. Prog Neurobiol 1999;58:233–247. 23 Liu B, Hong JS: Role of microglia in inflamma- matory stimuli in activation-induced cell death
4 Streit WJ, Walter SA, Pennell NA: Reactive tion-mediated neurodegenerative diseases: of mouse microglial cells: initiation of two sep-
microgliosis. Prog Neurobiol 1999; 57: 563– Mechanisms and strategies for therapeutic in- arate apoptotic pathways via induction of in-
581. tervention. J Pharmacol Exp Ther 2003; 304: terferon regulatory factor-1 and caspase-11. J
5 Kreutzberg GW: Microglia: A sensor for path- 1–7. Biol Chem 2001;276:32956–32965.
ological events in the CNS. Trends Neurosci 24 Chavarria A, Alcocer-Varela J: Is damage in 39 Lee H, Cha S, Lee MS, Cho GJ, Choi WS, Suk
1996;19:312–318. central nervous system due to inflammation? K: Role of antiproliferative B cell translocation
6 Minghetti L, Levi G: Microglia as effector cells Autoimmun Rev 2004;3:251–260. gene-1 as an apoptotic sensitizer in activation-
in brain damage and repair: Focus on pros- 25 Popovich PG, Jones TB: Manipulating neuro- induced cell death of brain microglia. J Immu-
tanoids and nitric oxide. Prog Neurobiol 1998; inflammatory reactions in the injured spinal nol 2003;171:5802–5811.
54:99–125. cord: Back to basics. Trends Pharmacol Sci 40 Suk K, Kim SY, Kim H: Essential role of cas-
7 Gonzalez-Scarano F, Baltuch G: Microglia as 2003;24:13–17. pase-11 in activation-induced cell death of rat
mediators of inflammatory and degenerative 26 von Knethen A, Lotero A, Brune B: Etoposide astrocytes. J Neurochem 2002;80:230–238.
diseases. Annu Rev Neurosci 1999; 22: 219– and cisplatin induced apoptosis in activated 41 Drache B, Diehl GE, Beyreuther K, Perlmutter
240. RAW 264.7 macrophages is attenuated by LS, Konig G: Bcl-xl-specific antibody labels ac-
8 Araque A, Perea G: Glial modulation of syn- cAMP-induced gene expression. Oncogene tivated microglia associated with Alzheimer’s
aptic transmission in culture. Glia 2004; 47: 1998;17:387–394. disease and other pathological states. J Neuro-
241–248. 27 Albina JE, Cui S, Mateo RB, Reichner JS: Ni- sci Res 1997;47:98–108.
9 Vesce S, Bezzi P, Volterra A: The active role of tric oxide-mediated apoptosis in murine peri- 42 Migheli A, Cavalla P, Piva R, Giordana MT,
astrocytes in synaptic transmission. Cell Mol toneal macrophages. J Immunol 1993; 150: Schiffer D: bcl-2 protein expression in aged
Life Sci 1999;56:991–1000. 5080–5085. brain and neurodegenerative diseases. Neuro-
10 Aschner M: Astrocytes as mediators of im- 28 Adler B, Adler H, Jungi TW, Peterhans E: In- report 1994;5:1906–1908.
mune and inflammatory responses in the CNS. terferon-alpha primes macrophages for lipo- 43 Streit WJ: Microglia and Alzheimer’s disease
Neurotoxicology 1998;19:269–281. polysaccharide-induced apoptosis. Biochem pathogenesis. J Neurosci Res 2004;77:1–8.
11 Galea E, Feinstein DL, Reis DJ: Induction of Biophys Res Commun 1995;215:921–927. 44 Giulian D: Ameboid microglia as effectors of
calcium-independent nitric oxide synthase ac- 29 Liu B, Wang K, Gao HM, Mandavilli B, Wang inflammation in the central nervous system. J
tivity in primary rat glial cultures. Proc Natl JY, Hong JS: Molecular consequences of acti- Neurosci Res 1987;18:155–171, 132–153.
Acad Sci USA 1992;89:10945–10949. vated microglia in the brain: Overactivation 45 Giulian D, Haverkamp LJ, Li J, Karshin WL,
12 Sawada M, Kondo N, Suzumura A, Marunou- induces apoptosis. J Neurochem 2001;77:182– Yu J, Tom D, Li X, Kirkpatrick JB: Senile
chi T: Production of tumor necrosis factor-al- 189. plaques stimulate microglia to release a neuro-
pha by microglia and astrocytes in culture. 30 Lee P, Lee J, Kim S, Yagita H, Lee MS, Kim toxin found in Alzheimer brain. Neurochem
Brain Res 1989;491:394–397. SY, Kim H, Suk K: NO as an autocrine me- Int 1995;27:119–137.
13 Simmons ML, Murphy S: Induction of nitric diator in the apoptosis of activated microglial 46 Nagata K, Takei N, Nakajima K, Saito H, Koh-
oxide synthase in glial cells. J Neurochem cells: Correlation between activation and saka S: Microglial conditioned medium pro-
1992;59:897–905. apoptosis of microglial cells. Brain Res 2001; motes survival and development of cultured
14 Becher B, Prat A, Antel JP: Brain-immune con- 892:380–385. mesencephalic neurons from embryonic rat
nection: Immunoregulatory properties of CNS- 31 Suk K, Lee J, Hur J, Kim YS, Lee MS, Cha SH, brain. J Neurosci Res 1993;34:357–363.
resident cells. Glia 2000;29:293–304. Kim SY, Kim H: Activation-induced cell death 47 McGeer PL, McGeer EG: The inflammatory
15 Nakamura Y: Regulating factors for microglial of rat astrocytes. Brain Res 2001; 900: 342– response system of brain: Implications for ther-
activation. Biol Pharm Bull 2002; 25: 945– 347. apy of Alzheimer and other neurodegenerative
953. 32 Donjerkovic D, Scott DW: Activation-induced diseases. Brain Res Brain Res Rev 1995; 21:
16 Ho LJ, Lai JH: Chinese herbs as immunomod- cell death in B lymphocytes [In Process Cita- 195–218.
ulators and potential disease-modifying anti- tion]. Cell Res 2000;10:179–192. 48 McGeer PL, Kawamata T, Walker DG, Aki-
rheumatic drugs in autoimmune disorders. 33 Crispe IN: Death and destruction of activated yama H, Tooyama I, McGeer EG: Microglia in
Curr Drug Metab 2004;5:181–192. T lymphocytes. Immunol Res 1999; 19: 143– degenerative neurological disease. Glia 1993;
17 Li FQ, Lu XZ, Liang XB, Zhou HF, Xue B, Liu 157. 7:84–92.
XY, Niu DB, Han JS, Wang XM: Triptolide, 34 Kingham PJ, Cuzner ML, Pocock JM: Apop- 49 Hanisch UK: Microglia as a source and target
a Chinese herbal extract, protects dopaminer- totic pathways mobilized in microglia and neu- of cytokines. Glia 2002;40:140–155.
gic neurons from inflammation-mediated rones as a consequence of chromogranin A-in- 50 Bent S, Ko R: Commonly used herbal medi-
damage through inhibition of microglial acti- duced microglial activation. J Neurochem cines in the United States: A review. Am J Med
vation. J Neuroimmunol 2004;148:24–31. 1999;73:538–547. 2004;116:478–485.
18 Ward PA, Warren JS, Johnson KJ: Oxygen 35 Kingham PJ, Pocock JM: Microglial apoptosis 51 De Smet PA: Herbal remedies. N Engl J Med
radicals, inflammation, and tissue injury. Free induced by chromogranin A is mediated by mi- 2002;347:2046–2056.
Radic Biol Med 1988;5:403–408. tochondrial depolarisation and the permeabil- 52 Gong X, Sucher NJ: Stroke therapy in tradi-
19 Henson PM, Johnston RB Jr: Tissue injury in ity transition but not by cytochrome c release. tional Chinese medicine (TCM): Prospects for
inflammation: Oxidants, proteinases, and cat- J Neurochem 2000;74:1452–1462. drug discovery and development. Trends
ionic proteins. J Clin Invest 1987; 79: 669– Pharmacol Sci 1999;20:191–196.
674.

32 Neurosignals 2005;14:23–33 Suk


53 Howes MJ, Houghton PJ: Plants used in Chi- 68 Middleton E Jr, Kandaswami C: Effects of fla- 83 Chopin P, Briley M: Effects of four non-cho-
nese and Indian traditional medicine for im- vonoids on immune and inflammatory cell linergic cognitive enhancers in comparison
provement of memory and cognitive function. functions. Biochem Pharmacol 1992;43:1167– with tacrine and galanthamine on scopol-
Pharmacol Biochem Behav 2003; 75: 513– 1179. amine-induced amnesia in rats. Psychophar-
527. 69 Kim HK, Cheon BS, Kim YH, Kim SY, Kim macology (Berl) 1992;106:26–30.
54 Chalela JA, Merino JG, Warach S: Update on HP: Effects of naturally occurring flavonoids 84 Woodruff-Pak DS, Vogel RW 3rd, Wenk GL:
stroke. Curr Opin Neurol 2004;17:447–451. on nitric oxide production in the macrophage Galantamine: Effect on nicotinic receptor
55 Harvey AL: Medicines from nature: Are natu- cell line RAW 264.7 and their structure-activ- binding, acetylcholinesterase inhibition, and
ral products still relevant to drug discovery? ity relationships. Biochem Pharmacol 1999; learning. Proc Natl Acad Sci USA 2001; 98:
Trends Pharmacol Sci 1999;20:196–198. 58:759–765. 2089–2094.
56 Kim YO, Leem K, Park J, Lee P, Ahn DK, Lee 70 Wakabayashi I, Yasui K: Wogonin inhibits in- 85 Sweeney JE, Bachman ES, Coyle JT: Effects of
BC, Park HK, Suk K, Kim SY, Kim H: Cyto- ducible prostaglandin E(2) production in mac- different doses of galanthamine, a long-acting
protective effect of Scutellaria baicalensis in rophages [In Process Citation]. Eur J Pharma- acetylcholinesterase inhibitor, on memory in
CA1 hippocampal neurons of rats after global col 2000;406:477–481. mice. Psychopharmacology (Berl) 1990; 102:
cerebral ischemia. J Ethnopharmacol 2001;77: 71 Chang YL, Shen JJ, Wung BS, Cheng JJ, Wang 191–200.
183–188. DL: Chinese herbal remedy wogonin inhibits 86 Ha JH, Shin SM, Lee SK, Kim JS, Shin US,
57 Suk K, Kim SY, Leem K, Kim YO, Park SY, monocyte chemotactic protein-1 gene expres- Huh K, Kim JA, Yong CS, Lee NJ, Lee DU: In
Hur J, Baek J, Lee KJ, Zheng HZ, Kim H: Neu- sion in human endothelial cells. Mol Pharma- vitro effects of hydroxybenzaldehydes from
roprotection by methanol extract of Uncaria col 2001;60:507–513. Gastrodia elata and their analogues on
rhynchophylla against global cerebral ischemia 72 Park BK, Heo MY, Park H, Kim HP: Inhibi- GABAergic neurotransmission, and a struc-
in rats. Life Sci 2002;70:2467–2480. tion of TPA-induced cyclooxygenase-2 expres- ture-activity correlation. Planta Med 2001;67:
58 Kubo M, Matsuda H, Tanaka M, Kimura Y, sion and skin inflammation in mice by wogo- 877–880.
Okuda H, Higashino M, Tani T, Namba K, nin, a plant flavone from Scutellaria radix. Eur 87 Kim HJ, Moon KD, Lee DS, Lee SH: Ethyl
Arichi S: Studies on Scutellariae radix. VII. J Pharmacol 2001;425:153–157. ether fraction of Gastrodia elata Blume pro-
Anti-arthritic and anti-inflammatory actions 73 Gao Z, Huang K, Yang X, Xu H: Free radical tects amyloid beta peptide-induced cell death.
of methanolic extract and flavonoid compo- scavenging and antioxidant activities of flavo- J Ethnopharmacol 2003;84:95–98.
nents from Scutellariae radix. Chem Pharm noids extracted from the radix of Scutellaria 88 Kim HJ, Lee SR, Moon KD: Ether fraction of
Bull (Tokyo) 1984;32:2724–2729. baicalensis Georgi. Biochim Biophys Acta methanol extracts of Gastrodia elata, medici-
59 Sekiya K, Okuda H: Selective inhibition of 1999;1472:643–650. nal herb protects against neuronal cell damage
platelet lipoxygenase by baicalein. Biochem 74 Shieh DE, Liu LT, Lin CC: Antioxidant and after transient global ischemia in gerbils. Phy-
Biophys Res Commun 1982;105:1090–1095. free radical scavenging effects of baicalein, ba- tother Res 2003;17:909–912.
60 Wakabayashi I: Inhibitory effects of baicalein icalin and wogonin [In Process Citation]. An- 89 Zangara A: The psychopharmacology of huper-
and wogonin on lipopolysaccharide-induced ticancer Res 2000;20:2861–2865. zine A: An alkaloid with cognitive enhancing
nitric oxide production in macrophages. Phar- 75 Kandaswami C, Middleton E Jr: Free radical and neuroprotective properties of interest in
macol Toxicol 1999;84:288–291. scavenging and antioxidant activity of plant the treatment of Alzheimer’s disease. Pharma-
61 Gertz HJ, Kiefer M: Review about Ginkgo bi- flavonoids. Adv Exp Med Biol 1994;366:351– col Biochem Behav 2003;75:675–686.
loba special extract EGb 761 (Ginkgo). Curr 376. 90 Wen TC, Yoshimura H, Matsuda S, Lim JH,
Pharm Des 2004;10:261–264. 76 Gao Z, Huang K, Xu H: Protective effects of Sakanaka M: Ginseng root prevents learning
62 Bastianetto S, Ramassamy C, Dore S, Christen flavonoids in the roots of Scutellaria baicalen- disability and neuronal loss in gerbils with 5-
Y, Poirier J, Quirion R: The Ginkgo biloba ex- sis Georgi against hydrogen peroxide-induced minute forebrain ischemia. Acta Neuropathol
tract (EGb 761) protects hippocampal neurons oxidative stress in HS-SY5Y cells. Pharmacol (Berl) 1996;91:15–22.
against cell death induced by beta-amyloid. Res 2001;43:173–178. 91 Lim JH, Wen TC, Matsuda S, Tanaka J, Mae-
Eur J Neurosci 2000;12:1882–1890. 77 Kim H, Kim YS, Kim SY, Suk K: The plant da N, Peng H, Aburaya J, Ishihara K, Sakana-
63 Bastianetto S, Zheng WH, Quirion R: The flavonoid wogonin suppresses death of activat- ka M: Protection of ischemic hippocampal
Ginkgo biloba extract (EGb 761) protects and ed C6 rat glial cells by inhibiting nitric oxide neurons by ginsenoside Rb1, a main ingredient
rescues hippocampal cells against nitric oxide- production. Neurosci Lett 2001;309:67–71. of ginseng root. Neurosci Res 1997; 28: 191–
induced toxicity: Involvement of its flavonoid 78 Lee H, Kim YO, Kim SY, Kim H, Noh HS, 200.
constituents and protein kinase C. J Neuro- Kang SS, Cho GJ, Choi WS, Suk K: Flavonoid 92 Kennedy DO, Scholey AB: Ginseng: Potential
chem 2000;74:2268–2277. wogonin from medicinal herb is neuroprotec- for the enhancement of cognitive performance
64 Braquet P, Hosford D: Ethnopharmacology tive by inhibiting inflammatory activation of and mood. Pharmacol Biochem Behav 2003;
and the development of natural PAF antago- microglia. FASEB J 2003;17:1943–1944. 75:687–700.
nists as therapeutic agents. J Ethnopharmacol 79 Hamada H, Hiramatsu M, Edamatsu R, Mori 93 Fahn S, Cohen G: The oxidant stress hypoth-
1991;32:135–139. A: Free radical scavenging action of baicalein. esis in Parkinson’s disease: Evidence support-
65 Oyama Y, Chikahisa L, Ueha T, Kanemaru K, Arch Biochem Biophys 1993;306:261–266. ing it. Ann Neurol 1992;32:804–812.
Noda K: Ginkgo biloba extract protects brain 80 Suk K, Lee H, Kang SS, Cho GJ, Choi WS: 94 Metodiewa D, Koska C: Reactive oxygen spe-
neurons against oxidative stress induced by hy- Flavonoid baicalein attenuates activation-in- cies and reactive nitrogen species: Relevance
drogen peroxide. Brain Res 1996; 712: 349– duced cell death of brain microglia. J Pharma- to cyto(neuro)toxic events and neurologic dis-
352. col Exp Ther 2003;305:638–645. orders: An overview. Neurotox Res 2000; 1:
66 Beecher GR: Overview of dietary flavonoids: 81 Boehm U, Klamp T, Groot M, Howard JC: 197–233.
Nomenclature, occurrence and intake. J Nutr Cellular responses to interferon-gamma. Annu 95 Ahlemeyer B, Krieglstein J: Neuroprotective
2003;133:3248S–3254S. Rev Immunol 1997;15:749–795. effects of Ginkgo biloba extract. Cell Mol Life
67 Middleton E, Jr., Kandaswami C, Theoharides 82 Zhang ZJ: Therapeutic effects of herbal ex- Sci 2003;60:1779–1792.
TC: The effects of plant flavonoids on mam- tracts and constituents in animal models of 96 Rahman K: Garlic and aging: New insights
malian cells: implications for inflammation, psychiatric disorders. Life Sci 2004; 75: 1659– into an old remedy. Ageing Res Rev 2003; 2:
heart disease, and cancer. Pharmacol Rev 1699. 39–56.
2000;52:673–751.

Herbal Medicine and Neuroinflammation Neurosignals 2005;14:23–33 33


Review

Received: October 11, 2004


Neurosignals 2005;14:34–45
Accepted after revision: November 16, 2004
DOI: 10.1159/000085384

Search for Natural Products Related to


Regeneration of the Neuronal Network
Chihiro Tohda a Tomoharu Kuboyama a, b Katsuko Komatsu a, b
a Research
Center for Ethnomedicines, Institute of Natural Medicine, and b 21st Century COE Program,
Toyama Medical and Pharmaceutical University, Toyama, Japan

Key Words Introduction


Neuritic atrophy W Synaptic loss W Dendrite W Axon W
Alzheimer’s disease W Amyloid-beta W Ginseng W Withania Despite the great number of ongoing investigations,
somnifera W Ashwagandha W Coffee bean neurodegenerative diseases remain incurable. The drugs
currently available for dementia, such as donepezil, an
acetylcholinesterase inhibitor, are efficacious in the tem-
Abstract porary treatment of memory dysfunction, but do not pre-
The reconstruction of neuronal networks in the damaged vent or reverse the underlying neurodegeneration [1]. In
brain is necessary for the therapeutic treatment of neu- patients with Alzheimer’s disease, neuritic atrophy and
rodegenerative diseases. We have screened the neurite synaptic loss are considered the major causes of cognitive
outgrowth activity of herbal drugs, and identified several impairment, based on the results of neuropathological
active constituents. In each compound, neurite out- postmortem studies of the brain [2–4]. In the brains of
growth activity was investigated under amyloid-ß-in- patients suffering from other neurodegenerative diseases,
duced neuritic atrophy. Most of the compounds with such as Parkinson’s disease, Huntington’s disease, and
neurite regenerative activity also demonstrated memory Creutzfeldt-Jakob disease, neurite atrophy has also been
improvement activity in Alzheimer’s disease-model observed [5–7]. Such atrophy leads to the destruction of
mice. Protopanaxadiol-type saponins in Ginseng drugs neuronal networks, and subsequently to the fatal dysfunc-
and their metabolite, M1 (20-O-ß-D-glucopyranosyl- tion of brain systems in these patients. The exclusion of,
(20S)-protopanaxadiol), showed potent regeneration ac- or at least a decrease in the magnitude of, the causes of
tivity for axons and synapses, and amelioration of mem- each disease may prevent the progression of symptoms,
ory impairment. Withanolide derivatives (withanolide A, but such inhibition is not associated with the repair of
withanoside IV, and withanoside VI) isolated from the already severely damaged brain function. We hypothe-
Indian herbal drug Ashwagandha, also showed neurite sized that the reconstruction of neuronal networks in the
extension in normal and damaged cortical neurons. Tri- injured brain would be the most necessary step in the fun-
gonelline, a constituent of coffee beans, demonstrated damental recovery of brain function, requiring neuritic
the regeneration of dendrites and axons, in addition to regeneration and synaptic reconstruction.
memory improvement.
Copyright © 2005 S. Karger AG, Basel

© 2005 S. Karger AG, Basel Katsuko Komatsu, PhD


ABC 1424–862X/05/0142–0034$22.00/0 Research Center for Ethnomedicines, Institute of Natural Medicine
Fax + 41 61 306 12 34 Toyama Medical and Pharmaceutical University, 2630 Sugitani
E-Mail karger@karger.ch Accessible online at: Toyama 930-0194 (Japan)
www.karger.com www.karger.com/nsg Tel. +81 76 434 7645, Fax +81 76 434 5064, E-Mail katsukok@ms.toyama-mpu.ac.jp
Table 1. Natural medicine-oriented compounds which enhance neurite outgrowth

Compound Main botanical source Cell used Effective dose Function Reference

Ginsenoside Rb1 Panax ginseng rat cortical 0.1–100 ÌM axon extension 14, 21
Panax notoginseng neuron synaptogenesis
memory improvement
Metabolite 1* (protopanaxadiol-type rat cortical 0.01–1 ÌM axon extension 21
saponins) neuron synaptogenesis
memory improvement
Withanolide A Withania somnifera rat cortical 1 ÌM axon extension 36
neuron dendrite extension 37
synaptogenesis
memory improvement
Withanoside IV Withania somnifera rat cortical 1 ÌM axon extension 36
neuron dendrite extension
synaptogenesis
memory improvement
Withanoside VI Withania somnifera rat cortical 1 ÌM axon extension 36
neuron dendrite extension
synaptogenesis
memory improvement
Trigonelline coffee bean rat cortical 30–100 ÌM axon extension 41
neuron dendrite extension
memory improvement
Honokiol Magnolia obovata rat cortical 0.1–10 ÌM neurite outgrowth 43
Magnolia officinalis neuron
(–)-3,5-Dicaffeoyl- Aster scaber PC12 1–10 ÌM neurite outgrowth 44
muco-quinic acid
Catalpol Rehmannia glutinosa PC12h 0.1–1 Ìg/ml neurite outgrowth 45
Geniposide Gardenia jasminoides PC12h 0.1–10 Ìg/ml neurite outgrowth 45
Gardenoside Gardenia jasminoides PC12h 0.1–10 Ìg/ml neurite outgrowth 45
Picroside I Picrorhiza PC12D 10–100 ÌM potentiating 46
scrophulariiflora NGF-induced
neurite outgrowth
Picroside II Picrorhiza PC12D 0.1–100 ÌM potentiating 46
scrophulariiflora NGF-induced
neurite outgrowth
Nardosinone Nardostachys chinensis PC12D 0.1–100 ÌM potentiating 47
NGF-induced
neurite outgrowth

* 20-O-ß-D-Glucopyranosyl-(20S)-protopanaxadiol.

Natural Products Enhancing Neurite Outgrowth extracts, several constituents have been identified as ac-
tive compounds (table 1). It is critical that extended neu-
Neurite outgrowth is the first step in the construction rites have specific functions, such as axons and dendrites,
of the neuronal network, and neurite outgrowth activity and can make circuits by synaptic connections. However,
has been investigated in many crude drugs. Of these the identification of axons and dendrites and the mea-

Natural Products Related to Regeneration Neurosignals 2005;14:34–45 35


of the Neuronal Network
surement of synaptogenesis have not been undertaken in gests that ppd-type saponins are active compounds. Gin-
studies of natural products, apart from in our research. seng, Red Ginseng, Notoginseng and Ye-Sanchi, which
Ginseng drugs, Ashwagandha and coffee beans contain showed neurite outgrowth activity, have been demon-
interesting compounds with potent neurite regeneration, strated to contain comparatively rich ppd-type saponins
synaptic reconstruction and memory improvement ac- in our quantitative study [18]. This suggested that the
tivities. effects of these drugs could be mainly attributed to ppd-
type saponins. However, Zhuzishen (dried rhizome of P.
japonicus var. major from Yunnan province) and a rhi-
Ginseng Drugs zome of P. stipuleanatus, which inhibited cell viability,
may contain some cytotoxic compounds.
Neurite Outgrowth Using Methanol Extracts and
Isolated Saponins in SK-N-SH Cells Effect of M1, a Metabolite of Protopanaxadiol-Type
Ginseng, the root of Panax ginseng, is widely used as a Saponins, on Aß(25–35)-Induced Memory Impairment,
tonic throughout the world, and is efficacious in the treat- Axonal Atrophy and Synaptic Loss in Mice
ment of amnesia. In addition, significant improvement in When taken orally, ppd-type saponins are mostly me-
learning and memory has been observed in brain-dam- tabolized by intestinal bacteria to ppd monoglucoside, 20-
aged [8, 9] and aged rats [9] after the oral administration O-ß-D-glucopyranosyl-(20S)-protopanaxadiol (M1) [19,
of Ginseng powder, and the major Ginseng saponins, gin- 20] (fig. 1). As Ginseng is generally taken orally, a metabo-
senoside Rb1 and Rg1, are known to improve spatial lite of ppd-type saponins, M1, should be investigated to
learning in normal mice [10]. Regarding the effects on determine the active constituent of Ginseng responsible
neuronal cells, it has been shown that neurite outgrowth for its major effects. We therefore conducted experiments
of cultured rat cerebral cortical neurons is enhanced by to determine whether treatment with ginsenoside Rb1, as
crude Ginseng saponins [11], and that ginsenoside Rb1 a representative of ppd-type saponins, and its metabolite,
potentiates the nerve growth factor (NGF)-mediated neu- M1, can induce recovery from memory disorder, axonal
rite outgrowth of chick dorsal root ganglia [12, 13]. atrophy, and synaptic loss induced by the active fragment
We tested the neurite outgrowth activity of methanol of the amyloid-ß peptide (Aß(25–35)) [21].
extracts of 6 types of Ginseng drugs and P. stipuleanatus Male ddY mice (6 weeks old) were prepared to create a
plant material in SK-N-SH cells [14]. The methanol mouse model of Alzheimer’s disease (AD). Seven days
extracts of Ginseng (dried root of P. ginseng), Red Gin- after an i.c.v. injection of Aß(25–35), ginsenoside Rb1
seng (steamed and dried root of P. ginseng), Notoginseng (10 Ìmol/kg), M1 (10 Ìmol/kg), donepezil hydrochloride
(dried root of P. notoginseng) and Ye-Sanchi (dried rhi- (DNP, 0.5 mg/kg), or the vehicle (tap water) was adminis-
zome and root of P. vietnamensis var. fuscidiscus) in- tered orally once daily for 14 days. Mice were trained in
creased neurite outgrowth, with the effects of Red Gin- the water maze for 7 days starting 14 days after the i.c.v.
seng and Ye-Sanchi being particularly significant. administration of Aß(25–35) (fig. 2a). The escape latency
Thirty saponins were isolated from Ye-Sanchi and to find the platform in the Aß(25–35)-injected group sig-
structurally elucidated [15, 16]. Oleanolic acid-type sa- nificantly increased compared with the saline-injected
ponins were also isolated from Kouzichi (dried rhizome group, whereas the escape latencies of the groups adminis-
of P. japonicus var. major from Hubei province) [17], and tered ginsenoside Rb1 and M1 p.o. significantly decreased
19 saponins (ginsenosides Rb1, Rb3, Rg1 and Re, notogin- as compared with the vehicle-administered group. The
senosides R4, Fa and R1, Yesanchinoside J, 20-O-glc-gin- donepezil-administered group showed no significant
senoside Rf, majonoside R2, (24S)-pseudoginsenoside shortening of the escape latency.
RT4 and F11, vina-ginsenoside R1, R2 and R6 from Ye- In the retention test (fig. 2b), the number of crossings
Sanchi, notoginsenoside R2, ginsenoside Rg2 and Ro, chi- over a previous platform position was significantly de-
kusetsusaponin IVa from Kouzichi) were tested. Proto- creased in the Aß(25–35)-injected group compared with
panaxadiol (ppd)-type saponins, ginsenosides Rb1 and the saline-injected group. The number of crossings recov-
Rb3, and notoginsenosides R4 and Fa significantly ex- ered after treatment with ginsenoside Rb1 and M1. Treat-
tended the neurites in SK-N-SH cells at a concentration of ment with donepezil showed the smallest effect in the
100 ÌM, and their activity increased dose-dependently. retention test. All mice showed normal swimming perfor-
On the other hand, protopanaxatriol, ocotillol and olean- mance and a constant increase in body weight. Locomotor
olic acid-type saponins showed no effect [14]. This sug- activity did not differ among groups.

36 Neurosignals 2005;14:34–45 Tohda/Kuboyama/Komatsu


Glc1-O

OH

COO⫺

N⫹
HO
CH3
␤-D-glucopyranosyl-(20S)-protopanaxadiol
20-O-␤ Trigonelline
(M1)

CH3
CH2R2
OH
R1
O O
H O O
H
O H
H OH H
H

H H
H H

OH O Glc1-6Glc1–O

Withanolide A R1 R2
(WL-A)
Withanoside IV (WS-IV) H OH
Withanoside VI (WS-VI) OH H

Fig. 1. Chemical structures of compounds that regenerate the neuronal network.

After the retention test, the expression levels of phos- the temporal cortex and CA1. In all areas, the synapto-
phorylated NF-H (axonal marker), synaptophysin (synap- physin levels were almost equal to or higher than control
tic marker) and MAP2 (dendritic marker) were measured levels in ginsenoside Rb1- and M1-treated mice. Donepe-
in mouse brains. We observed two cortical areas (parietal zil treatment had no effect on the synaptophysin levels.
cortex and temporal cortex) and three hippocampal areas The MAP2 levels were also reduced in the cerebral cortex
(CA1, CA3, and the dentate gyrus), as it is known that and CA1 of the brain in Aß(25–35)-injected compared
synaptic loss occurs primarily in the cerebral cortex and with saline-injected mice (fig. 3c). Significant decreases
hippocampus in AD patients [22, 23] and in AD model were seen in the temporal cortex; however, these de-
mice [24]. The phosphorylated NF-H levels were remark- creases in the expression levels of MAP2 were not clearly
ably reduced in these five areas of the brain in Aß(25– recovered by ginsenoside Rb1, M1 or donepezil. Although
35)-injected compared with saline-injected mice (fig. 3a). treatment with M1 tended to increase the MAP2 level in
Significant decreases were seen in the parietal cortex, CA1 the temporal cortex, the effect was weak. No differences
and CA3; however, the expression levels of phosphorylat- in neuronal density were observed among the groups in
ed NF-H were nearly equal to those of the control in ginse- any brain areas. Treatment with M1, a metabolite of gin-
noside Rb1- and M1-treated mice. Donepezil treatment senoside Rb1, results in the recovery of impaired learning
had no effect on the levels of phosphorylated NF-H. The and memory in Aß(25–35)-injected mice with degener-
synaptophysin levels were also reduced in these five areas ated axons and synapses. The maintained retention of
of the brain in Aß(25–35)-injected compared with saline- spatial memory was also seen after the discontinuation of
injected mice (fig. 3b). Significant decreases were seen in ginsenoside Rb1 and M1 administration. These results

Natural Products Related to Regeneration Neurosignals 2005;14:34–45 37


of the Neuronal Network
suggest that ginsenoside Rb1 and M1 may induce the
60
structural repair of neuronal connections.
]
Sal/Veh In the rat large intestine, ginsenoside Rb1 is completely
A␤/Veh metabolized to M1 3 h after administration [25]. In mice,
50 ] A␤/GRb1 only M1 is continuously detected in the blood from 30
] A␤/M1
min to 16 h after oral administration of ginsenoside Rb1
[26]. In humans, M1 is detected in plasma from 7 h after
40 A␤/DNP
Escape latencies (s)

the ingestion of Ginseng, and in urine from 12 h after


intake, and aglycone is not detected in either plasma or
30 urine [20]. These results suggest that M1 is the final
metabolite of ppd-type saponins. The recovery potency in
Aß(25–35)-injected mice by p.o.-administered ginseno-
20
side Rb1 and M1 was almost identical, indicating that the
majority of orally administered ginsenoside Rb1 was me-
10 tabolized into M1. Considering that most ppd-type sapo-
nins are metabolized to M1, which is the active principal,
the total content of ppd-type saponins is possibly an
0
1 2 3 4 5 6 7
important index of the anti-AD activity of Ginseng.
a Days of acquisition test
Effect of M1 on Aß(25–35)-Induced Axonal Atrophy
8
in Rat Cortical Neurons
]
Number of crossings over a platform position for 60 s

In in vitro experiments, M1 demonstrated an axonal


7 regeneration effect. To investigate the Aß(25–35)-induced
damage to the neuronal network and the reconstructive
6 activity of drugs, 10 ÌM Aß(25–35) was added to the cor-
tical neurons on day 7, and after 3 days the medium was
5 replaced by fresh medium, including drugs. Although the
cortical neurons connected with each other during the 7-
4 day culture, some of the connections were lost 3 days after
Aß(25–35) treatment. At 4 days, both phosphorylated
3
NF-H-positive (fig. 4a) and MAP2-positive (fig. 4b) neu-
rites were significantly shortened by Aß(25–35) treat-
ment. Treatment with 0.01 ÌM M1 (to 78.5% of the con-
2
trol) significantly increased the recovery of the length of
phosphorylated NF-H-positive neurites (fig. 4a), while
1
MAP2-positive neurites were not extended (fig. 4b). NGF
significantly enhanced the lengths of phosphorylated NF-
Veh Veh GRb 1 M1 DNP
b Saline A␤(25–35)

Fig. 2. Effects of ginsenoside Rb1 and M1 on the impairment of spa- been measured over 60 s, 6 days after the last acquisition test. This
tial memory induced by Aß(25–35) injection. a Escape latencies per was also 6 days after the discontinuance of drug treatment. Vehicle
group in four trials were tested in a Morris water maze over 7 days. was administered p.o. to saline-i.c.v.-injected mice. To Aß(25–35)-
Vehicle was administered p.o. to saline-i.c.v.-injected mice. To i.c.v.-injected mice, vehicle (Veh), ginsenoside Rb1 (GRb1), M1, or
Aß(25–35)-i.c.v.-injected mice (5 nmol), the vehicle, ginsenoside Rb1 donepezil (DNP) was administered p.o. Values represent the means
(10 Ìmol/kg), M1 (10 Ìmol/kg), or donepezil (0.5 mg/kg) was admin- and SEM of 9 mice. * p ! 0.05 when compared with the Aß(25–35)
istered p.o. for 14 days. Values represent the means and SEM of 9 plus vehicle-treated group. One-way analysis of variance was carried
mice. * p ! 0.05 when compared with the Aß(25–35) plus vehicle- out, followed by Dunnett’s post hoc test.
treated group. Two-way repeated measure analysis of variance was
carried out, followed by Dunnett’s post hoc test. b The number of
crossings over the previous position of a platform had previously

38 Neurosignals 2005;14:34–45 Tohda/Kuboyama/Komatsu


Fig. 3. Effects of ginsenoside Rb1 and M1 on
axonal atrophy and synaptic loss induced by
Aß(25–35) injection. Expression levels of
phosphorylated NF-H (a), synaptophysin
(b) and MAP2 (c) in brain slices were quan-
tified. Vehicle was administered p.o. to
saline-i.c.v.-injected mice. To Aß(25–35)-
i.c.v.-injected mice, vehicle, ginsenoside Rb1
(10 Ìmol/kg), M1 (10 Ìmol/kg), or donepezil
(0.5 mg/kg) was administered p.o. for 14
days. The parietal cortex (PC), temporal cor-
tex (TC), hippocampal CA1 and CA3, and
dentate gyrus (DG) were observed. The fluo-
rescence intensities of six areas in each slice
were measured. Values represent the means
and SEM of three mice. * p ! 0.05 when
compared with the Aß(25–35) plus vehicle-
treated group. One-way analysis of variance
was carried out, followed by Dunnett’s post
hoc test.

Natural Products Related to Regeneration Neurosignals 2005;14:34–45 39


of the Neuronal Network
H-positive (to 82.0% of the control) and MAP2-positive
500 (to 95.3% of the control) neurites. In addition, M1
]
increased in pre-synaptic density to the control level after
]
Aß(25–35)-induced synaptic loss occurred [our unpub-
Length of NF-H-positive neurites

400 ] lished data].


Neuritic atrophy by Aß(1–40) and Aß(25–35) has been
reported in chick sympathetic neurons [27] and rat corti-
per cell (␮m)

300
cal neurons [28]. As neurite atrophy is thought to be due
to unusual cell adhesion [27, 29], M1 may be capable of
200 normalizing the adhesive mechanism. Although Aß is
known to cause neuronal death through increased [Ca2+]
neurons [30], increased peroxynitrites in microglias [31],
100
and mitochondrial dysfunction in neurons [32], the death
pathway has been shown to be mediated by separate
0 molecular mechanisms of a neuritic dystrophy event [27–
Veh Veh M1 NGF
29]. Since ginsenoside Rb1 did not inhibit neuronal death
a A␤(25–35)
induced by Aß(25–35), the mechanism of rescuing axonal
atrophy may not be identical to that for recovery from
500
Aß-induced neuronal death.

] ]
Length of MAP2-positive neurites

400
Ashwagandha
per cell (␮m)

300 Neurite Outgrowth with Methanol Extract and Isolated


Withanolides
Ashwagandha (root of Withania somnifera Dunal) is
200
the most popular herbal drug in Ayurvedic medicine, and
has been used traditionally and commonly as a tonic and
100 nootropic agent. It has also been reported as associated
with improvements in scopolamine-induced memory def-
icits in mice [33]. Treatment with a methanol extract of
0 Ashwagandha induced neurite extension [34]. We further
Veh Veh M1 NGF
identified 6 withanolide derivatives from methanol ex-
A␤(25–35) tract (withanolide A, withanoside IV, withanoside VI,
Culture Immunostaining
etc.; fig. 1), which induced neurite outgrowth in human
neuroblastoma SH-SY5Y cells [35]. In normal cortical
neurons, the predominant dendritic outgrowth was in-
7d 3d 4d
duced by treatment with withanoside IV or withanoside
b A␤ Drugs VI, whereas predominant axonal outgrowth was observed
in treatment with withanolide A in normal cortical neu-
rons [36].
Fig. 4. Effect of post-treatment with M1 on Aß(25–35)-induced
axonal and dendritic atrophy. Aß(25–35) (10 ÌM ) was added to rat
cortical neurons at 7 days in vitro. Three days later, the medium was Effect of Withanolides on Aß(25–35)-Induced Neuritic
replaced by new medium containing M1 (0.01 ÌM ), NGF (100 ng/ Atrophy and Synaptic Loss
ml) or the vehicle (Veh, DMSO). Four days later, the cells were fixed In Aß(25–35)-induced damaged cortical neurons,
and immunostained for phosphorylated NF-H (a) and MAP2 (b). withanolide A, withanoside IV, and withanoside VI
The lengths of neurites positive for phosphorylated NF-H or MAP2
showed neuritic regeneration and synaptic reconstruc-
per cell were measured. Values represent the means and SEM of 30
cells. * p ! 0.05 when compared with the Aß(25–35) plus vehicle- tion. 24 h after culture initiation, 10 ÌM Aß(25–35) was
treated group. One-way analysis of variance was carried out, followed added to the culture medium simultaneously with the
by Dunnett’s post hoc test. drugs. Four days later, Aß(25–35) treatment significantly

40 Neurosignals 2005;14:34–45 Tohda/Kuboyama/Komatsu


]

Length of MAP2-positive neurites


150
]
]
]

per cell (␮m)


100

0
Cont Veh WL-A WS-IV WS-VI NGF
a A␤(25–35)

]
]

150
Length of NF-H-positive neurites
per cell (␮m)

Fig. 5. Effects of withanolide A, withanoside


IV, and withanoside VI on the prevention of
Aß(25–35)-induced dendritic and axonal
atrophy. Cortical neurons were cultured for
24 h, and then the cells were treated simulta-
neously with 10 ÌM Aß(25–35), and with- 100
anolide A (WL-A), withanoside IV (WS-IV),
or withanoside VI (WS-VI) at a concentra-
tion of 1 ÌM; or NGF or BDNF at a concen- 0
Cont Veh WL-A WS-IV WS-VI NGF
tration of 100 ng/ml; or vehicle (Veh); or
A␤(25–35)
with vehicle alone (Cont). Four days after
treatment, the cells were fixed and immu- Immunostaining
Culture A␤
nostained for MAP2 or phosphorylated NF-
H. Lengths of MAP2-positive neurites (a)
and phosphorylated NF-H-positive neurites
(b) were measured in each treatment. The
values represent the means and SEM of 30 1d 4d
cells. * p ! 0.05 when compared with the
Aß(25–35) plus vehicle-treated group. One- b Drug
way analysis of variance was carried out, fol-
lowed by Dunnett’s post hoc test.

inhibited the outgrowth of both MAP2-positive neurites was completely prevented by treatment with withanolide
and phosphorylated NF-H-positive neurites, showing that A (97.0% of the control), withanoside IV (106.3% of the
Aß(25–35) induced both dendritic and axonal atrophy in control), or withanoside VI (117.4% of the control)
rat cortical neurons. Simultaneous treatment with Aß(25– (fig. 5a). In particular, treatment with withanosides IV
35) and withanolide A, withanoside IV, or withanoside VI and VI tended to induce the growth of longer dendrites
at a concentration of 1 ÌM prevented both dendritic and than treatment with withanolide A.
axonal atrophy induced by Aß(25–35). Dendritic atrophy

Natural Products Related to Regeneration Neurosignals 2005;14:34–45 41


of the Neuronal Network
Axonal atrophy was partially prevented by treatment
with withanoside IV (88.0% of the control) and withano-
side VI (90.0% of the control), whereas treatment with
withanolide A (98.6% of the control) completely prevent-
ed axonal atrophy (fig. 5b).
To determine whether regenerated neurites are able to
reconstruct synapses, the expressions of synaptic markers
were investigated. Rat cortical neurons were cultured for
21 days to construct mature synapses in vitro, and after
the culture period, Aß(25–35) was added to the samples.
Four days later, the cells were immunostained with an
antibody for post-synaptic density, (PSD)-95 (post-synap-
tic marker), or with synaptophysin (pre-synaptic marker).
PSD-95- and synaptophysin-positive puncta were signifi-
cantly decreased by treatment with Aß(25–35) [37]. With-
anolide A, withanoside IV, withanoside VI, or NGF was
added to the culture medium after 4 days of treatment
with Aß(25–35) after synaptic loss had occurred. Seven
days after the addition of the drug, the cells were fixed and
immunostained for PSD-95 or synaptophysin. Treatment
with withanolide A, withanoside IV, or withanoside VI
significantly induced both PSD-95 and synaptophysin
expression, as compared with treatment with the vehicle.
These results indicate that withanolide A, withanoside IV,
and withanoside VI facilitated the reconstruction of both
post-synaptic and pre-synaptic regions in neurons in
which severe synaptic loss had already occurred. This
increase in post-synaptic structures tended to be signifi-
cant following treatment with withanoside IV (86.0% of
the control) and withanoside VI (83.6% of the control), as
compared with withanolide A treatment (68.0% of the
control). However, reconstruction of the pre-synaptic re-
gion was induced significantly and markedly by treatment
with withanolide A (108.1% of the control), as compared
with withanoside IV (81.3% of the control) and withano-
side VI (75.8% of the control) treatments. Treatment with
NGF did not lead to an increase in the development of
either the post-synapses (57.7% of the control) or the pre-
synapses (54.4% of the control).

Fig. 6. The effect of trigonelline on the prevention of Aß(25–35)- phosphorylated NF-H. Lengths of MAP2-positive neurites (a) and
induced dendritic and axonal atrophy. Cortical neurons were cul- phosphorylated NF-H-positive neurites (b) were measured in each
tured for 3 days, and then the cells were treated simultaneously with treatment. The values represent the means and SEM of 12–20 cells
10 ÌM Aß(25–35), and trigonelline at a concentration of 30 or (a) or 14–22 cells (b). * p ! 0.05 when compared with the Aß(25–35)
100 ÌM, or vehicle (Veh), or with the vehicle alone (Cont). Five days plus vehicle-treated group. One-way analysis of variance was carried
after treatment, the cells were fixed and immunostained for MAP2 or out, followed by Dunnett’s post hoc test.

42 Neurosignals 2005;14:34–45 Tohda/Kuboyama/Komatsu


Although NGF extended both axons and dendrites
(fig. 4, 5), it has no effect on synaptogenesis. Since NGF
itself is not able to pass through the blood-brain barrier,
low-molecular-weight substances that mimic NGF action
have been developed as anti-dementia drugs. However,
such NGF-like drugs are not expected to cure dementia
because of a lack of synaptogenesis activity.

Coffee Beans

Neurite Outgrowth with Trigonelline


Coffee is consumed as a drink, and is known to stimu-
late the central nervous system as well as the heart and
circulation [38]. It is thought that these effects are mainly
caused by caffeine [39] but the effects of other coffee con-
stituents on the central nervous system have hardly been
reported. Coffee beans are crude drugs, used in the tradi-
Fig. 7. Effect of trigonelline on the impairment of spatial memory
tional system of Unani medicine [40]. induced by Aß(25–35) injection. The number of crossings over the
Among the extracts of raw and roasted coffee beans, a previous position of a platform was measured over 60 s, 6 days after
methanol-soluble fraction of the ethanol extract (1 Ìg/ml) the last acquisition test in a Morris water maze. This was also 6 days
of raw beans significantly increased the percentage of cells after the discontinuance of drug treatment. Vehicle was administered
p.o. to saline-i.c.v.-injected mice. To Aß(25–35)-i.c.v.-injected mice
with neurites in human neuroblastoma SK-N-SH cells
(4.7 nmol), the vehicle (Veh), 500 mg/kg trigonelline (TGN), or
[41]. It was demonstrated that the neurite outgrowth 0.5 mg/kg donepezil (DNP) was administered p.o. Values represent
activity of the methanol fraction decreased depending on the means and SEM of 9 mice. * p ! 0.05 when compared with the
the extent of roasting. Among subfractions of this metha- Aß(25–35) plus vehicle-treated group. One-way analysis of variance
nol fraction, the basic fraction had significant neurite out- was carried out, followed by Dunnett’s post hoc test.
growth activity. In this basic fraction, trigonelline was
identified as an active constituent (fig. 3). It is known that
a decrease in trigonelline is related to the degree of roast-
ing [42].
In rat cortical neurons, trigonelline showed dendritic jected group. The number of crossings was recovered by
and axonal regeneration. Three days after initiation of the treatment with trigonelline, suggesting that memory re-
culture, Aß(25–35) was added to the culture medium with tention is improved by trigonelline.
trigonelline. Trigonelline (30 and 100 ÌM) treatment
dose-dependently prevented both dendritic (fig. 6a) and
axonal (fig. 6b) atrophy induced by Aß(25–35). Conclusions

Effect of Trigonelline on Aß(25–35)-Induced Memory The ppd-type saponins of Ginseng drugs and M1 (a
Impairment metabolite of ppd-type saponins by intestinal bacteria)
Fourteen days after the i.c.v. injection of Aß(25–35) in induced significant recovery from memory impairment,
male ddY mice (6 weeks old), trigonelline (500 mg/kg), axonal atrophy and synaptic loss in mice. The effect of
donepezil hydrochloride (0.5 mg/kg), or the vehicle (tap M1 on axonal reconstruction was further confirmed in
water) was administered orally once daily for 15 days. cultured cortical neurons. These results suggest that orally
Mice were trained in the water maze for 5 days, starting administered ppd-type saponins potentially ameliorate
21 days after the i.c.v. administration of Aß(25–35). Six dementia by reconstructing the neuronal network. With-
days after the last acquisition test, the retention test was anolide A, withanoside IV, and withanoside VI, which
performed (fig. 7). The number of crossings over a pre- were isolated from Ashwagandha, facilitated the regenera-
vious platform position was significantly decreased in the tion of dendrites and axons, and led to the dramatic con-
Aß(25–35)-injected group compared with the saline-in- struction of synapses, although the neuron damage was

Natural Products Related to Regeneration Neurosignals 2005;14:34–45 43


of the Neuronal Network
profound and severe. Trigonelline also had dendritic and icine may offer a treasury of new medicines to treat intrac-
axonal regeneration activity, and improved memory re- table diseases with the use of novel study concepts and the
tention. These compounds, sourced from natural prod- application of objective scientific analyses.
ucts, and used with treatments preventing pathogenesis
and neuronal death, are expected to play an important
role as new categorized drugs in curing neurodegenerative Acknowledgments
diseases in the near future. Although we have shown the
We thank Prof. M. Hattori, Dr. K. Zou, Mr. N. Matsumoto, Dr.
high potential of neuronal regeneration from compounds
M. R. Meselhy, Dr. N. Nakamura and Dr. J. Zhao of the Institute of
isolated from Ginseng drugs, Ashwagandha and coffee Natural Medicine, Toyama Medical and Pharmaceutical University
beans, it is dangerous to simply imply that these herbal for their extensive contribution to this study. This work was support-
drugs are expected to be excellent anti-dementia drugs. ed by Kampou Science Foundation, Uehara Memorial Foundation,
When taking herbal drugs, the risk of side effects brought Grants-in-Aid for Scientific Research (B), No. 11695086 in 1999–
2001 and No. 14406030 in 2002–2004 from the Japan Society for the
by other constituents, and sufficient efficacy compared
Promotion of Science, and the 21st Century COE Program of the
with isolated compounds should be investigated and care- Ministry of Education, Culture, Sports, Science and Technology,
fully considered. However, drugs used in traditional med- Japan.

References

1 Ogura H, Kosasa T, Kuriya Y, Yamanishi Y: 10 Mook-Jung I, Hong HS, Boo JH, Lee KH, Yun 18 Zhu S, Zou K, Fushimi H, Cai SQ, Komatsu K:
Donepezil, a centrally acting acetylcholinester- SH, Cheong MY, Joo I, Huh K, Jung MW: Gin- Comparative study on triterpene saponins of
ase inhibitor, alleviates learning deficits in hy- senoside Rb1 and Rg1 improve spatial learning Ginseng drugs. Planta Medica 2004;70:666–
pocholinergic models in rats. Methods and and increase hippocampal synaptophysin level 677.
Findings in Exp Clin Pharmacol 2000;22:89– in mice. J Neurosci Res 2001;63:509–515. 19 Odani T, Tanizawa H, Takino Y: Studies on
95. 11 Sugaya A, Yuzurihara M, Tsuda T, Yasuda K, the absorption, distribution, excretion and me-
2 DeKosky S, Scheff SW: Synapse loss in frontal Kajiwara K, Sugaya E: Proliferative effect of tabolism of Ginseng saponins. III. The absorp-
cortex biopsies in Alzheimer’s disease: Correla- Ginseng saponin on neurite extension of pri- tion, distribution and excretion of ginsenoside
tion with cognitive severity. Ann Neurol 1990; mary cultured neurons of the rat cerebral cor- Rb1 in the rat. Chem Pharm Bull 1983;31:
27:457–464. tex. J Ethnopharmacol 1988;22:173–181. 1059–1066.
3 Terry RD, Masliah E, Salmon DP, Butters N, 12 Saito H, Suda K, Schwab M, Thoenen H: 20 Tawab MA, Bahr U, Karas M, Wurglics M,
DeTeresa R, Hill R, et al: Physical basis of cog- Potentiation of the NGF-mediated nerve fiber Schubert-Zsilavecz M: Degeneration of ginse-
nitive alterations in Alzheimer’s disease: Syn- outgrowth by ginsenoside Rb1 in organ cultures nosides in humans after oral administration.
apse loss is the major correlate of cognitive of chicken dorsal root ganglia. Jpn J Pharmacol Drug Metab Dispos 2003;31:1065–1071.
impairment. Ann Neurol 1991;30:572–580. 1977;27:445–451. 21 Tohda C, Matsumoto N, Zou K, Meselhy RM,
4 Dickson TC, Vickers JC: The morphological 13 Nishiyama N, Cho SI, Kitagawa I, Saito H: Komatsu K: Aß(25–35)-induced memory im-
phenotype of ß-amyloid plaques and associated Malonylginsenoside Rb1 potentiated nerve pairment, axonal atrophy, and synaptic loss are
neuritic changes in Alzheimer’s disease. Neuro- growth factor (NGF)-induced neurite out- ameliorated by M1, a metabolite of protopan-
science 2001;105:99–107. growth of cultured chick embryonic dorsal root axadiol-type saponins. Neuropsychopharma-
5 Jackson M, Gentleman S, Lennox L, Ward L, ganglia. Biol Pharm Bull 1994;17:509–513. cology 2004;29:860–868.
Gray T, Randall K, et al: The cortical neuritic 14 Tohda C, Matsumoto N, Zou K, Meselhy RM, 22 DeKosky ST, Scheff SW: Synapse loss in fron-
pathology by Huntington’s disease. Neuropa- Komatsu K: Axonal and dendritic extension by tal cortex biopsies in Alzheimer’s disease: Cor-
thol Appl Neurobiol 1995;21:18–26. protopanaxadiol-type saponins from Ginseng relation with cognitive severity. Ann Neurol
6 Mattila PM, Rinne JO, Helenius H, Roytta M: drugs in SK-N-SH cells. Jpn J Pharmacol 2002; 1990;27:457–464.
Neuritic degeneration in the hippocampus and 90:254–262. 23 Heinonen O, Soininen H, Sorvari H, Kosunen
amygdala in Parkinson’s disease in relation to 15 Zou K, Zhu S, Tohda C, Cai SQ, Komatsu K: O, Paljarvi L, Koivisto E, Riekkinen PJ Sr:
Alzheimer pathology. Acta Neuropathol 1999; Dammarane-type triterpene saponins from Pa- Loss of synaptophysin-like immunoreactivity
98:157–164. nax japonicus. J Natural Products 2002;65: in the hippocampal formation is an early phe-
7 Liberski PP, Budka H: Neuroaxonal pathology 346–351. nomenon in Alzheimer’s disease. Neuroscience
in Creutzfeldt-Jakob disease. Acta Neuropa- 16 Zou K, Zhu S, Meselhy RM, Tohda C, Cai SQ, 1995;64:375–384.
thol 1999;97:329–334. Komatsu K: Dammarane-type saponins from 24 Games D, Adams D, Alessandrini R, Barbour
8 Zhao R, MaDaniel WF: Ginseng improves Panax japonicus and their neurite outgrowth R, Berthelette P, Blackwell C, Carr T, Clemens
strategic learning by normal and brain-dam- activity in SK-N-SH cells. J Natural Products J, Donaldson T, Gillespie F, Guido T, Hago-
aged rats. NeuroReport 1998;9:1618–1624. 2002;65:1288–1292. pian S, Johnson-Wood K, Khan K, Lee M, Lei-
9 Zhong YM, Nishijo H, Uwano T, Tamura R, 17 Zou K, Zhu S, Cai SQ, Komatsu K: In Ye San bowitz P, Lieberburg I, Little S, Masliah E,
Kawanishi K, Ono T: Red Ginseng amelio- Qi, Kou Zi Qi: Constituents from the under- McConlogue L, Montoya-Zavala M, Mucke L,
rated place navigation deficits in young rats ground part of Panax plants. Abstract 121th Paganini L, Penniman E, Power M, Schenk D,
with hippocampal lesions and aged rats. Physi- Ann Meet Pharmaceutical Soc Japan, 2001, Seubert P, Snyder B, Soriano F, Tan H, Vitale
ol Behav 2000;69:511–525. p 110. J, Wadsworth S, Wolozin B, Zhao J: Alzhei-
mer-type neuropathology in transgenic mice
overexpressing V717Fß-amyloid precursor
protein. Nature 1995;373:523–527.

44 Neurosignals 2005;14:34–45 Tohda/Kuboyama/Komatsu


25 Karikura M, Miyase T, Tanizawa H, Taniyama 32 Casley CS, Land JM, Sharpe MA, Clark JB, 41 Tohda C, Nakamura N, Komatsu K, Hattori
T, Takino Y: Studies on absorption, excretion Duchen MR, Canevari L: ß-Amyloid fragment M: Trigonelline-induced neurite outgrowth in
and metabolism of Ginseng saponons. VII. 25–35 causes mitochondrial dysfunction in pri- human neuroblastoma SK-N-SH cells. Biol
Comparison of the decomposition of ginseno- mary cortical neurons. Neurobiol Dis 2002;10: Pharm Bull 1999;22:679–682.
side-Rb1 and -Rb2 in the digestive tract of rats. 258–267. 42 Taguchi H: Biosynthesis and metabolism of tri-
Chem Pharm Bull 1991;39:2357–2361. 33 Dhuley JN: Nootropic-like effect of Ashwa- gonelline, and physiological action of the com-
26 Hasegawa H, Sung J-H, Benno Y: Role of gandha (Withania somnifera L.) in mice. Phy- pound (abstract in English). Vitamins 1988;62:
human intestinal Prevotella oris in hydrolyzing tother Res 2001;15:524–528. 549–557.
Ginseng saponins. Planta Medica 1997;63: 34 Tohda C, Kuboyama T, Komatsu K: Dendrite 43 Fukuyama Y, Nakade K, Minoshima Y, Yo-
436–440. extension by methanol extract of Ashwagandha koyama R, Zhai H, Mitsumoto Y: Neurotro-
27 Postuma RB. He W, Nunan J, Beyreuther K, (roots of Withania somnifera) in SK-N-SH phic activity of honokiol on the cultures of fetal
Masters CL, Barrow CJ, Small DH: Substrate- cells. NeuroReport 2000;11:1981–1985. rat cortical neurons. Bioorg Med Chem Lett
bound beta-amyloid peptides inhibit cell adhe- 35 Zhao J, Nakamura N, Hattori M, Kuboyama 2002;12:1163–1166.
sion and neurite outgrowth in primary neuro- T, Tohda C, Komatsu K: Withanolide deriva- 44 Hur JY, Lee P, Kim H, Kang I, Lee KR, Kim
nal cultures. J Neurochem 2000;74:1122– tives from the roots of Withania somnifera and SY: (–)-3,5-Dicaffeoyl-muco-quinic acid iso-
1130. their neurite outgrowth activities. Chem lated from Aster scaber contributes to the dif-
28 Grace EA, Rabiner CA, Busciglio: Character- Pharm Bull 2002;50:760–765. ferentiation of PC12 cells: Through tyrosine
ization of neuronal dystrophy induced by fi- 36 Kuboyama T, Tohda C, Zhao J, Nakamura N, kinase cascade signaling. Biochem Biophys Res
brillar amyloid beta: Implications for Alzhei- Hattori M, Komatsu K: Axon- or dendrite-pre- Commun 2004;313:948–953.
mer’s disease. J Neurosci 2002;114:265–273. dominant outgrowth induced by constituents 45 Yamazaki M, Chiba K, Mohri T: Neuritogene-
29 Grace EA, Busciglio J: Aberrant activation of from Ashwagandha. NeuroReport 2002;13: sis effect of natural iridoid compounds on
focal adhesion proteins mediates fibrillar amy- 1715–1720. PC12h cells and its possible relation to signal-
loid ß-induced neuronal dystrophy. J Neurosci 37 Kuboyama T, Tohda C, Komatsu K: Neuritic ing protein kinases. Biol Pharm Bull 1996;19:
2003;23:493–502. regeneration and synaptic reconstruction in- 791–795.
30 Lin H, Bhatia R, Lal R: Amyloid ß protein duced by withanolide A. Br J Pharmacol 46 Li P, Matsunaga K, Yamakuni T, Ohizumi Y:
forms ion channels: implications for Alzhei- 2005;144:961–971. Potentiation of nerve growth factor-action by
mer’s disease pathophysiology. FASEB J 2001; 38 Czok G: Coffee and health (abstract in En- picrosides I and II, natural iridoids, in PC12D
15:2433–2444. glish). Z Ernährungswiss 1977;16:248–255. cells. Eur J Pharmacol 2000;406:203–208.
31 Xie Z, Wei M, Morgan TE, Fabrizio P, Han D, 39 Riedel W, Hogervorst E, Leboux R, Verhey F, 47 Li P, Matsunaga K, Yamamoto K, Yoshikawa
Finch CE, Longo VD: Peroxynitrite mediates van Praag H, Jolles J: Caffeine attenuates sco- R, Kawashima K, Ohizumi Y: Nardosinone, a
neurotoxicity of amyloid beta-peptide1–42- polamine-induced memory impairment in hu- novel enhancer of nerve growth factor in neu-
and lipopolysaccharide-activated microglia. J mans. Psychopharmacology 1995;122:158– rite outgrowth from PC12D cells. Neurosci
Neurosci 2002;22:3484–3492. 168. Lett 1999;273:53–56.
40 Liu YM, Shawuti Y: Pharmacography of Ui-
ghur. Wulumuqi, Xinjiang People’s Publishing
House, 1985, p 267.

Natural Products Related to Regeneration Neurosignals 2005;14:34–45 45


of the Neuronal Network
Review

Neurosignals 2005;14:46–60 Received: January 28, 2005


Accepted after revision: March 1, 2005
DOI: 10.1159/000085385

Multifunctional Activities of Green Tea


Catechins in Neuroprotection
Modulation of Cell Survival Genes, Iron-Dependent Oxidative Stress and PKC
Signaling Pathway

Silvia A. Mandel Yael Avramovich-Tirosh Lydia Reznichenko Hailin Zheng


Orly Weinreb Tamar Amit Moussa B.H. Youdim
Eve Topf and USA National Parkinson Foundation Centers of Excellence for Neurodegenerative Diseases
Research, Technion-Faculty of Medicine, Haifa, Israel

Key Words tivities and to protect neuronal death in a wide array of


(–)-Epigallocatechin-3-gallate  Neurorescue  cellular and animal models of neurological diseases. Re-
Neurodegeneration  Neuroprotection  Parkinson’s cent studies have indicated that in addition to the known
disease  Neurite outgrowth  Green tea catechins  antioxidant activity of catechins, other mechanisms such
Iron chelation  Cell signaling  Protein kinase C as modulation of signal transduction pathways, cell sur-
vival/death genes and mitochondrial function, contrib-
ute significantly to the induction of cell viability. This
Abstract review will focus on the multifunctional properties of
Many lines of evidence suggest that oxidative stress re- green tea and its major component (–)-epigallocatechin-
sulting in reactive oxygen species (ROS) generation and 3-gallate (EGCG) and their ability to induce neuroprotec-
inflammation play a pivotal role in the age-associated tion and neurorescue in vitro and in vivo. In particular,
cognitive decline and neuronal loss in neurodegenera- their transitional metal (iron and copper) chelating prop-
tive diseases including Alzheimer’s (AD), Parkinson’s erty and inhibition of oxidative stress.
(PD) and Huntington’s diseases. One cardinal chemical Copyright © 2005 S. Karger AG, Basel

pathology observed in these disorders is the accumula-


tion of iron at sites where the neurons die. The buildup
of an iron gradient in conjunction with ROS (superoxide, Introduction
hydroxyl radical and nitric oxide) are thought to consti-
tute a major trigger in neuronal toxicity and demise in Polyphenols are natural substances present in bever-
all these diseases. Thus, promising future treatment of ages obtained from plants, fruits and vegetables such as
neurodegenerative diseases and aging depends on avail- olive oil, red wine and tea. Flavonoids are the largest
ability of effective brain permeable, iron-chelatable/rad- group of polyphenols, which include the subclasses of fla-
ical scavenger neuroprotective drugs that would prevent vones, isoflavones, flavanols, flavans and flavonols [1].
the progression of neurodegeneration. Tea flavonoids Several prototypes of these groups have been shown to
(catechins) have been reported to possess potent iron- promote a number of physiological benefits, especially in
chelating, radical-scavenging and anti-inflammatory ac- cognitive function and memory impairment. Fresh tea

© 2005 S. Karger AG, Basel Prof. M.B.H. Youdim


1424–862X/05/0142–0046$22.00/0 Eve Topf and USA National Parkinson Foundation Centers of Excellence for
Fax +41 61 306 12 34 Neurodegenerative Diseases Research, Technion-Faculty of Medicine
E-Mail karger@karger.ch Accessible online at: POB 9697, 31096 Haifa (Israel)
www.karger.com www.karger.com/nsg Tel. +972 4 8295290, Fax +972 4 8513145, E-Mail Youdim@Tx.Technion.ac.il
(Camellia sinensis) leaves contain a high amount of cat- resulting in ROS generation from H2O2 and inflamma-
echins, a group of flavonoids or flavanols, known to con- tory processes, trigger a cascade of events leading to
stitute 30–45% of the solid green tea extract [2, 3]. The apoptotic/necrotic cell death in neurodegenerative dis-
favorable properties ascribed to tea consumption are be- orders, such as Parkinson’s (PD), Alzheimer’s (AD) and
lieved to rely on its bioactive components, catechins and Huntington’s diseases and amyotrophic lateral sclerosis
their derivatives, demonstrated to act directly as radical (ALS) [17]. There is also evidence for increased expres-
scavengers and exert indirect antioxidant effects through sion of apoptotic proteins (for review see [18]), as well
activation of transcription factors and antioxidant en- as mitochondria (complex I) and ubiquitin-proteasome
zymes [for reviews, see 4, 5]. The most abundant poly- system (UPS) dysfunction, which may lead to break-
phenolic compound is EGCG, thought to contribute to down of energy metabolism and consecutive intraneu-
the beneficial effects attributed to green tea, such as its ronal calcium overload [19–22]. Thus, neurodegenera-
anticancer, cardiovascular function improvement and tion appears to be multifactorial, where a complex set
antioxidant anti-inflammatory properties. Indeed, a of reactions lead to the demise of neurons. This assump-
number of epidemiological studies have shown that phe- tion receives support from the familial (genetic) forms
nolic compounds reduce the risk of coronary heart dis- of neurodegenerative diseases identified in the last years,
ease, possibly via their anti-inflammatory effects, includ- where mutations in genes, such as -synuclein, parkin
ing inhibition of adhesion molecule and cytokine expres- and ubiquitin C-terminal hydrolase-L1 (UCHL-1) de-
sion and augmentation of endothelial nitric oxide release scribed in rare forms of hereditary PD [23], may lead to
[6]. Relative antioxidant activities among tea catechins impairment in the activity of the UPS. More recently,
have been found to be EGCG = (–)-epicatechin-3-gallate recessive mutations in DJ-1 [24] and PINK1 (PTEN-
(ECG) 1 (–)-epigallocatechin (EGC) 1 (–)-epicatechin induced kinase 1) [25] were proposed to play a role in
(EC) [7]. EGCG accounts for more than 10% of the ex- cellular response to OS, supporting a pathogenic role of
tract dry weight (30–130 mg per cup of tea) followed by ROS in the etiology of neurodegenerative diseases.
EGC 1 EC 6 ECG [2]. In addition to their radical scav- Therefore, it is not surprising that antioxidants were the
enging action, green tea catechins possess well-established first drugs to be studied in an attempt to retard the prog-
metal-chelating properties. Structurally important fea- ress of PD. Recently, coenzyme Q10, an intrinsic com-
tures defining their chelating potential are the 3,4-dihy- ponent of the mitochondrial respiratory chain acting as
droxyl group in the B ring [8] as well as the gallate group a bioenergizer and an antioxidant, was studied as a pu-
[9, 10], which may neutralize ferric iron to form redox- tative neuroprotective agent in PD. This double-blind,
inactive iron, thereby protecting cells against oxidative placebo-controlled pilot study demonstrated that high
damage [11]. However, a wealth of data is accumulating doses of coenzyme Q10 (1,200 mg/d) were associated
and indicating that the antioxidant/metal chelating attri- with a reduced rate of deterioration in motor function
butes of the catechin polyphenols are unlikely to be the from baseline over the 16-month course of this trial
sole explanation for their neuroprotective and neurores- [26].
cue capacity. Thus, catechin polyphenols were found to Neuropathological and neurochemical studies on sub-
invoke a wide spectrum of different mechanisms of ac- stantia nigra (SN) from PD brains and its animal models
tion responsible for cell survival [for our recent reviews, [23, 27, 28] and our recent gene expression profiling of
see 12, 13]. human SN pars compacta (pc) from PD patients [29]
There is evidence that polyphenol metabolites and demonstrate the existence of a ‘domino’ cascade of neu-
their parent compounds have access to the brain. Studies rotoxic events, which can be initiated at any point in the
with radioactively labeled EGCG in mouse or chemilu- cascade. These series of events may act independently or
minescence-based detection of EGCG in rats demon- cooperatively during the course of the disease, leading
strated its incorporation into brain, as well as in various eventually to the demise of dopaminergic neurons. This
organs including kidney, heart, liver, spleen and pancreas has led to the current notion that drugs directed against
[14, 15]. Furthermore, it has been shown that the methyl- a single target will be ineffective and rather a single drug
ated and glucuronidated derivatives of epicatechin are or cocktail of drugs with pluripharmacological properties
both detected in rat brain following oral administration may be more suitable to be employed. According to this
[16]. belief, green tea catechins well fulfill the requirements for
Significant body of evidence support the hypothesis a putative neuroprotective drug having diverse pharma-
that brain iron dysregulation and oxidative stress (OS), cological activities. Thus, it is not surprising that they

Multifunctional Catechins for Neurosignals 2005;14:46–60 47


Neuroprotection
have attracted increasing interest as therapeutic cytopro- [35] cerebral ischemia in gerbils. Insights into the possible
tective agents for the treatment of neurodegenerative and mechanism of neuroprotection by EGCG in the infarct
other diseases. area of ischemic rats, revealed that it acts by reducing
In this article the state of the art of the diverse mo- iNOS expression, infiltration and peroxynitrite forma-
lecular mechanisms and cell signaling pathways partici- tion [36], by increasing endothelial and neuronal NOS
pating in the neuroprotective action of green tea catechin and preservation of mitochondrial complex activity and
polyphenols is reviewed. Particular attention has been integrity [37]. In this context, the decrease in the activity
paid to their iron-chelating properties with respect to the of the transcription factor signal transducer and activator
potential promise for iron chelation therapy, as a novel of transcription-1alpha (STAT-1alpha) by EGCG in isch-
treatment for neurodegenerative diseases. emic rat cardiac myocytes may well account for the re-
duced mRNA levels of iNOS, a target of STAT-1 [38].
Other investigators have recently shown that EGCG re-
Neuroprotective Effects of Catechins: duced brain inflammation and neuronal damage in ex-
Insights from in vivo and in vitro Studies perimental autoimmune encephalomyelitis (EAE), when
given at initiation or after the onset of EAE [39].
There is a growing recognition that polyphenolic cat- An extensive number of studies regarding neuropro-
echins exert a protective role in neurodegeneration. An tection by green tea flavonoids in cellular and animal
experimental study conducted in N-methyl-4-phenyl- models of neurodegenerative diseases are starting to ac-
1,2,3,6-tetrahydropyridine (MPTP) model of PD has cumulate. Hence, the flavonoid epicatechin was shown to
shown that both green tea extract and EGCG effectively attenuate the toxicity induced by oxidized low-density
prevent mice striatal dopamine (DA) depletion and SN lipoprotein in mouse-derived striatal neurons [40] or fi-
dopaminergic neuron loss [30]. The protection exerted by broblasts [41] and to confer protection to primary culture
green tea polyphenols in vivo may involve direct scaveng- of mesencephalic neurons challenged with 6-hydroxydo-
ing of ROS and regulation of antioxidant protective en- pamine (6-OHDA) [42]. Recently, catechin was shown to
zymes. EGCG was found to elevate the activity of two reduce injury produced by hydrogen peroxide, 4-hy-
major oxygen-radical species metabolizing enzymes, su- droxynonenal, rotenone and 6-OHDA in primary rat
peroxide dismutase (SOD) and catalase in mice striatum mesencephalic cultures, as shown by increases in cellular
[30]. This is supported by a previous finding where 1 viability and [3H]DA uptake [43]. Similarly, EGCG was
month’s administration of a catechin-containing antioxi- reported to protect human neuroblastoma cells from
dant preparation increased SOD activity in the mito- damage induced by 6-OHDA and MPP+ [44]. EGCG also
chondria fraction of striatum and midbrain and decreased protects primary hippocampal neurons [45] and rescues
thiobarbiturate reactive substance formation in the cor- rat pheochromocytoma (PC12) cells from amyloid- pep-
tex and cerebellum of aged rats [31]. The structural cat- tide (A)-induced toxicity [46]. More recently, EGCG
echol resemblance of EGCG may explain a recently re- was reported to exert a neurorescue activity in long-term
ported inhibitory effect of green tea polyphenols on the serum-deprived PC12 cells and to promote neurite out-
DA presynaptic transporters. This inhibition lead to 1- growth, as manifested by the expression of a surrogate
methyl-4-phenylpyridinium (MPP+) uptake blockade marker of cell differentiation, growth-associated protein
(because of competition for the vesicular transporter) GAP-43 (GAP-43) [47]. This could have important im-
thereby protecting DA-containing neurons against plications with regard to aging, PD and AD, suggesting a
MPP(+)-induced injury [32]. In addition, EGCG greatly potential therapeutic use of EGCG in regenerating in-
inhibited catechol-O-methyltransferase (COMT) activity jured neuronal cells.
in rat liver cytosol at a low IC50 concentration (0.2 M)
[33]. This action may be of particular significance for PD
patients, given that DA and related catecholamines are Neuroprotection and Neurotoxicity by Low
physiological substrates of COMT, thus its inhibition will and High Concentrations of Catechins and
result in increased DA in the synapse. Other Flavonoids
Green tea polyphenols have been also shown beneficial
in animal models of cerebral ischemia: intraperitoneal Studies from our and other laboratories have shown
injection of EGCG reduced hippocampal neuronal dam- that green tea polyphenols display a concentration-de-
age and brain edema caused by global [34] or unilateral pendent window of neuroprotective action: They protect

48 Neurosignals 2005;14:46–60 Mandel/Avramovich-Tirosh/Reznichenko/


Zheng/Weinreb/Amit/Youdim
at low micromolar concentrations, whereas they become tive effects against 6-OHDA- and A-induced cell death
pro-oxidant and pro-apoptotic when increasing the con- in SH-SY5Y and PC12 cells [44, 46] and for the neuro-
centrations over 10–20 M [44, 48]. This bell-shaped pat- rescue effect against long-term growth factors withdrawal
tern is typical of antioxidative drugs, such as vitamin C in PC12 cells [47]. This is supported by the observation
[49], R-apomorphine [50] and DA [51], being neuropro- that EGCG could not overcome cell death under PKC
tective at low (1–10 M) concentrations, while having pathway blockade, as determined both morphologically
pro-oxidant/pro-toxic activity at higher (10–50 M) con- and by monitoring various apoptotic markers, suggesting
centrations. The toxicity of not only green tea polyphe- that this cascade is essential for the neuronal protection
nols, but also of several other flavonoids, is responsible and rescue effects of EGCG. Consistent with these find-
for their antiproliferative and chemopreventive actions. ings, recent animal studies have shown that two weeks
The anticancer properties of polyphenols is attributed to consumption of EGCG (2 mg/kg) led to a highly signifi-
the ability to inhibit phase I and induce phase II carcino- cant up-regulation of PKC isoform in mice striatum [62]
gen metabolizing enzymes (in animals) that initiate car- and to a significant increase in PKC isoenzymes  and 
cinogenesis, inhibition of cell cycle progression effectors, in the membrane and cytosolic fractions of mice hippo-
promotion of ROS and nitrogen species (thereby collaps- campus [46]. The implication of PKC in neuronal sur-
ing the mitochondrial membrane potential), induction of vival by EGCG is further demonstrated in vitro by the
p53 and apoptogenic factors and inactivation of protein rapid translocation of PKC to the membrane compart-
kinases that contribute to survival-associated signal trans- ment in PC12 cells, in response to EGCG (fig. 1). PKC
duction [for extensive reviews see 5, 52, 53]. Since the is a well-established neuron cell survival factor participat-
present article applies to the mechanisms of neuroprotec- ing also in cell growth and differentiation [63, 64]. In sup-
tion by tea catechins, the literature regarding their anti- port, a recent report shows that treatment of human cells
carcinogenic or pro-apoptotic properties will not be re- with EGCG induces a specific translocation of PKC to
viewed. the membrane [65].
More direct evidence implicating PKC in EGCG
mechanistic action has come from a recent study employ-
Mechanism of Neuroprotection by Green Tea ing solid-state nuclear magnetic resonance, showing that
Catechins EGCG interacts with the head group region of the phos-
pholipids within lipid bilayers from liposomes [66]. The
Cell Signaling Pathways interaction pattern of EGCG in terms of rotational mo-
Selective Activation of Protein Kinase C (PKC) in tion within the lipid bilayers was similar to that described
Brain Neurons for 12-O-tetradecanoylphorbol-13-acetate (TPA) [67], a
PKC expression has been previously coupled with the phorbol ester. Phorbol esters are prototype activators of
preservation of cell survival and the formation and con- PKC, suggesting that direct interaction of green tea cat-
solidation of different types of memory [54–56]. The in- echins with cell membranes may be sufficient for the rap-
duction of PKC activity in neurons is thought to be a id activation of PKC by EGCG previously reported by us
prerequisite for neuroprotection against several exoge- [44]. The impact of EGCG on membrane fluidity may
nous insults. Indeed, PKC activation after ischemic pre- give rise to activation of other membrane-associated sig-
conditioning or pharmacologic preconditioning (with naling pathways (e.g. G proteins), which can contribute
PKC, NMDA, or A1AR agonists) was shown essential as well to its protective action. This clearly needs to be
for neuroprotection against oxygen/glucose deprivation examined.
in organotypic slice cultures [57]. In accordance, activa-
tion of PKC by estrogen or by the grape flavonoid resve- Modulation of Other Signal Transduction Pathways
ratrol, in rat cortical or hippocampal neurons, respective- and Intracellular Transducers
ly, protects against A toxicity [58, 59]. Also, we have In addition to PKC, other cell signaling pathways have
recently shown that the anti-Parkinson/monoamine oxi- been also implicated in the action of green tea catechins
dase-B (MAO-B) inhibitor drug, rasagiline (Teva Phar- such as the mitogen-activated protein kinases (MAPKs)
maceutical Industries) [60], prevented PC12 cell death and phosphatidylinositide 3-OH kinase (PI3K)/AKT
induced by serum deprivation via PKC signaling cascade signaling cascades. These cascades have been shown to
[61]. Similarly, we have reported that phosphorylative play central functions in neuronal protection against a
activation of PKC by EGCG is responsible for the protec- variety of extracellular insults and to be essential for neu-

Multifunctional Catechins for Neurosignals 2005;14:46–60 49


Neuroprotection
ronal differentiation and survival [68–70]. OS seems to
be a major stimulus for MAPK cascade, which might lead
to cell survival/cell death [for review see 71]. Among the
MAPKs the extracellular signal-regulated kinases
(ERK1/2) are mainly activated by mitogen and growth
factors [72], while p38 and c-jun-N-terminal kinase (JNK)
respond to stress stimuli [73]. However, there have been
reports where activation of ERK1/2 is thought to mediate
neuronal injury such as in focal ischemia [74], in gluta-
mate and oxidized-low-density-lipoprotein-induced tox-
icity [40, 75] and in cytotoxicity and activation of cas-
pase-3 in the extraneuronal hepatoma HepG2 [76] and
HeLa [77] cell lines, respectively. Increasing evidence
shows that catechins can protect against neuronal cell
death caused by exogenous OS-inducing agents through
modulation of ERK activity [40, 44]. In this regard, a
number of flavonoids and phenolic antioxidants, at their
respective low protective concentrations, were demon-
strated to activate the expression of some stress-response
genes, such as phase II drug-metabolizing enzymes, glu-
tathione-s-transferase and heme-oxygenase1 [77], likely
via activation of the MAPK pathway [78].
More recently, additional signaling pathways, includ-
ing PI3K/AKT, protein kinase A (PKA) and calcium,
have been implicated in the neuroprotective action of
catechin flavonoids. These studies have been conducted
mainly in extra-neuronal tissue such as skin and heart.
For example, topical application of EGCG induces pro-
liferation of human normal epidermal keratinocytes
through stimulation of ERK1/2 and AKT [79]. Other in-
vestigators reported a rapid activation of endothelial ni-
Fig. 1. EGCG activates p-PKC (pan) and induces a rapid activation
of PKC and translocation to the membrane. A Cell lysates from tric oxide synthase after EGCG treatment by a process
PC12 cells deprived of serum for 24 h before short-term (15 min) that involves PI3K, PKA and AKT in endothelial cells
exposure to EGCG (1–10 M) were subjected to SDS-PAGE and [80] and a decrease in iNOS expression via inactivation
Western blot, with p-PKC (pan) antibody. B Cultured PC12 cells of STAT-1alpha in epithelial and colon cell lines [81].
grown with full serum (FS) support (control) were deprived of se-
rum (SF) for 24 h before short-term (15 min) exposure to EGCG
Consistent with this, Townsend et al. [38] have recently
(1–10 M). The cells were fixed and permeabilized for subcellular reported that in cardiac myocytes EGCG protects against
localization of PKC by confocal microscopy, using an isoenzyme- ischemia/reperfusion-induced apoptosis through a mech-
specific antibody and FITC-conjugated secondary antibody (light anism involving reduction of STAT-1 phosphorylation
areas). DRAQ5 stains nuclei (dark areas). Under FS conditions (inactivation) and of his downstream pro-apoptotic target
(control) PKC is evenly confined to both plasma membrane (grey
arrow) and cytosol (white arrow) (a). Upon serum withdrawal, PK-
gene, Fas. The discrepancy or divergence in the different
C immunostaining is mosly cytosolic (b). In cells treated with signal pathway activation by EGCG may reflect differ-
EGCG PKC is, in its majority (1 M), or entirely (10 M) local- ences in cell tissue (e.g. neuronal vs. peripheral), or in the
ized to the cell membrane (arrows) (c, d). The images are represen- downstream pathways being under the control of the dif-
tative fields from 3 independent experiments, all showing the same ferent kinases, which may diverge into different respons-
results. Taken from Reznichenko et al. [47].
es, thereby providing cell function diversity.

Anti-Apoptotic Activity
EGCG has been reported to exert a biphasic mode of
action as a function of concentration. The low micromo-

50 Neurosignals 2005;14:46–60 Mandel/Avramovich-Tirosh/Reznichenko/


Zheng/Weinreb/Amit/Youdim
lar concentrations are responsible for the anti-apoptotic/ Metal-Chelating Activity
neuroprotective actions of EGCG. This receives further One cardinal feature of neurodegenerative diseases in-
recognition from an experiment employing customized cluding AD, PD, Huntington’s disease, ALS, Friedreich’s
cDNA microarray designed to clarify the molecular ataxia, multiple sclerosis, and aceruloplasminemia is the
mechanisms involved in the cell survival action of EGCG appearance of excessive iron at degenerative neuronal
[44]. The results revealed that a low EGCG concentration sites [17]. The buildup of an iron gradient in conjunction
decreased the expression of pro-apoptotic genes bax, bad, with ROS (superoxide, hydroxyl radical and nitric oxide)
caspases-1 and 6, cyclin-dependent kinase inhibitor p21, are thought to constitute a major trigger in the demise in
cell-cycle inhibitor gadd45, fas-ligand and tumor necrosis all theses diseases. Therefore, the chelation of free cellular
factor-related apoptosis-inducing ligand TRAIL, in SH- ferric and ferrous ions by the different metal chelators
SY5Y neuronal cells. The same group has reported re- make them potential agents to combat iron-induced gen-
cently that EGCG reduced the expression of several apop- eration of reactive oxygen radicals (by the Fenton and
togenic factors when given after long-term serum depri- Haber-Weiss reactions) and aggregation of alpha ()-
vation of PC12 cells [47]. These findings are supported synuclein and A in PD and AD, respectively.
by an in vivo study showing that two weeks oral consump-
tion of EGCG (2 mg/kg) alone caused a complete disap- Iron Chelation for AD
pearance of Bax immunoreactivity, specifically in the do- A significant body of evidence point to an ‘amyloid
paminergic neurons of the SNpc [62] and counteracted cascade’ event in the pathogenesis of AD, where amyloid
the robust increase of Bax protein when administered be- precursor protein (APP) is processed to A, by - and
fore MPTP intoxication in the same area. -secretases, which spontaneously self-aggregate in the
Bax alone, or in conjunction with the BH-3-only pro- presence of divalent metals (Fe2+, Cu2+) into neurotoxic
teins (e.g. Bad, Bid, Bim, Noxa, Puma), can trigger the amyloid fibrils in the neocortex [88]. Iron was shown to
opening of the mitochondrial megachannel permeability promote both deposition of A and OS, which is associ-
transition pore (mPTP), or a specific channel in the outer ated with the plaques [89]. In addition, iron has taken
mitochondrial membrane, both of which promote the fall center stage in AD as a consequence of the studies by
in mitochondrial membrane potential, leading to cyto- Rogers and coworkers [90] who described the existence
chrome c release and consequent cell death [82, 83]. The of an iron responsive element (IRE-type II) in the 5UTR
decline in Bax expression by EGCG may favor the in- region of APP mRNA. APP is post-transcriptionally reg-
crease in the ratio of Bcl-2/Bcl-xL to Bax/Bad proteins, ulated by iron regulatory proteins (IRPs), which are la-
thereby contributing to mitochondrial stability and regu- bile iron pool-sensitive cytosolic RNA proteins binding
lation of mPTP [84]. Protection of mitochondrial integ- specifically to the IRE located in the 5 or 3 untrans-
rity is of major importance, especially in the case of post- lated regions of iron metabolism-associated mRNAs.
mitotic cells such as neurons and heart muscle cells, which Changes in iron status (iron overload or depletion) lead
are commonly not renewed. Thus, it is not surprising that to compensating changes in the IRP/IRE system of
one of the major neuroprotective strategies in PD, AD translational control of iron homeostasis. For example,
and other neurodegenerative diseases, where increased the APP 5-UTR conferred translation was selectively
OS, perturbed cellular energy and ion homeostasis have down-regulated upon intracellular iron chelation, in a
been implicated, includes pharmacological agents direct- similar manner as the iron-storage protein ferritin,
ed to specific mitochondrial targets. In this respect, which also possesses an IRE in its 5-UTR mRNA
Ginkgo biloba extract EGb 761 or its individual compo- [90].
nents were shown to protect mitochondria integrity by Iron removal has effects suggestive of inhibition of key
protecting against uncoupling of oxidative phosphoryla- enzymes or other metalloproteins, for instance in mim-
tion, thereby increasing ATP levels and by increasing the icking hypoxia, whereas many of the effects of iron over-
expression of the mitochondrial DNA-encoded COX III load may be the result of signaling associated with OS.
subunit of cytochrome oxidase [for review, see 85]. Fla- Hypoxia and iron chelation have similar effects on genes
vonoids may also affect mitochondrial integrity by in- regulated by the transcription factor hypoxia-inducible
creasing GSH levels and preventing the influx of calcium, factor-1 (HIF-1), a master regulator orchestrating the
as previously reported [86, 87]. coordinated induction of an array of hypoxia-sensitive
genes [91]. The target genes of HIF are especially related
to angiogenesis, cell proliferation/survival and glucose/

Multifunctional Catechins for Neurosignals 2005;14:46–60 51


Neuroprotection
Fig. 2. Iron-induced neurodegeneration in AD via transcriptional turn, can initiate OH generation, causing oxidative stress (OS).
activation of APP mRNA. Neurodegeneration can result from ab- Increased iron and OS may activate the prolyl hydroxylase enzymes
normal serum iron transport to the neurons because a disruption which are key iron and oxygen sensors, leading to proteasomal-me-
in the blood-brain barrier (BBB) or from release from its storage diated degradation of the transcription factor hypoxia-inducible
protein ferritin, thereby increasing the free-labile iron pool (ionic factor 1, a master regulator orchestrating the coordinated induc-
iron). Labile iron can increase the production of amyloid precursor tion of a wide array of survival genes. It has been also suggested
protein (APP) by down-regulating the activities of iron regulatory that IRP2 can be a substrate for prolyl hydroxylase. Neuroprotec-
proteins (IRP1 and IRP2, inactivation and proteasomal degrada- tive agents that can be used to prevent iron-induced neurodegen-
tion, respectively), thereby promoting the translation of APP eration include M30 and HLA20 (bifunctional iron chelator-MAO
mRNA from its 5-UTR type II. Ionic iron may also cause aggrega- inhibitors), VK-28, EGCG and curcumin (iron chelators). For a
tion of amyloid- peptide (A) to form toxic aggregates, which, in more detailed explanation, read text.

iron metabolism [92]. The mechanism of HIF-1 activa- as HIF, signaling it for protein degradation. This suggests
tion by iron chelation is not well understood. Fe(II)/2- a convergence of both iron and OS to a common pathway
oxoglutarate-dependent dioxygenases have been identi- triggering the neurotoxic degenerative cascade (for a de-
fied that hydroxylate critical proline and asparagine resi- tailed explanation, see fig. 2).
dues in HIF and upon high oxygen levels and iron The involvement of metals in the plaques of AD pa-
overload, target HIF for degradation [93]. Thus, these tients and the presence of an IRE in the unstranslated
prolyl hydroxylase enzymes act as key iron and oxygen region of APP mRNA, have encouraged the development
sensors. This may explain the decrease in cell survival of iron chelators as a major new therapeutic strategy for
genes described in neurodegenerative diseases such as the treatment of AD. In fact, intramuscular administra-
phosphofructokinase and the angiogenic factor VEGF, tion of the prototype iron chelator desferrioxamine
both regulated by the HIF proteins [94]. Interestingly, the (DFO) slowed the clinical progression of AD dementia
free iron-induced proteasomal-mediated degradation of [97], and some success has also been achieved with an-
IRP2 involves also activation a prolyl hydroxylase and is other metal-complexing agent, clioquinol [98]. However,
inhibited by iron chelators [95, 96]. Thus, it is possible clioquinol is highly toxic [99] and DFO has poor penetra-
that IRP2 is a substrate for this enzyme, in a similar way tion across the blood-brain barrier [17].

52 Neurosignals 2005;14:46–60 Mandel/Avramovich-Tirosh/Reznichenko/


Zheng/Weinreb/Amit/Youdim
vivo [12, 101]. Our recent studies have shown that pro-
longed administration of EGCG to mice induced a reduc-
tion in holoAPP levels in the hippocampus [46]. Indeed,
this effect may be related to the iron-chelating properties
of EGCG, leading to a decrease in the free-iron pool. This
in turn results in the suppression of APP mRNA transla-
tion by targeting the IRE-II sequences in the APP 5-UTR
[90], as was recently shown for DFO and the bifunctional
amyloid-binding/metal-chelating drug XH1 [102] (fig. 2).
The concept of metal chelation as a neuroprotective
strategy has led us to the development of non-toxic, lipo-
philic, brain-permeable iron chelators for progressive
neurodegenerative diseases [103]. Compounds such as
VK-28 (Varinel) [104] and the multifunctional iron che-
lators HLA20 and M30 [105, 106], which possess the
propargylamine MAO inhibitory and neuroprotective
moiety of rasagiline, display good cell permeability and
are protective against 6-OHDA toxicity in differentiated
P19 cells [107]. Comparative analysis of the Fe2+-chelat-
ing potency of EGCG, the prototype DFO and other
Fig. 3. Comparison of the Fe2+-chelating potency of EGCG to oth-
er iron chelators. The metal-binding capacity of EGCG was com-
pharmacological iron chelators, has revealed similar
pared to that of DFO and the novel iron chelators VK28, HLA20 binding potency (fig. 3). Both EGCG and the iron chela-
and M30, by assessing their ability to compete with ferrozine for tor M30 were shown by us to induce a significant down-
the ferrous ions, resulting in decrease in the absorbance at 562 nm. regulation of membrane-associated holoAPP level in the
Ferrozine can quantitatively react with Fe2+ to form Fe2+-ferrozine mouse hippocampus (fig. 4), SH-SY5Y and CHO cells
complex with a strong absorbance at 562 nm. In the presence of
other chelating agents, the complex formation is disrupted with the
expressing the APP ‘Swedish’ mutation (data not shown).
result that the absorbance at 562 nm is decreased. 0.1 mM of drug This may have a direct influence on A levels and plaque
was mixed with 0.1 mM ferrozine in 5% ammonium acetate (pH formation, as shown in preliminary studies for XH1
7) followed by the addition of 0.02 mM FeSO4. After 2 h incuba- [102]. Indeed, using a nucleation-dependent polymeriza-
tion, the absorbance (at 562 nm) of resulting solutions was read. tion model, it has been shown that wine and green tea
Considering that the purpose of this assay was to evaluate the abil-
ity of drugs to compete with the iron indicator ferrozine, drugs polyphenols are able to inhibit formation, extension and
and ferrozine were used at equal concentrations. Chelating ef- destabilization of -amyloid fibrils [108].
fect of drug on Fe2+ was calculated as follows: Chelating effect Other potential beneficial effect of EGCG in AD may
(%) = [1–(absorbance of sample at 562 nm)/(absorbance of control be related to our previous studies demonstrating the abil-
at 562 nm)]! 100. The order of chelating potency for complexing ity of EGCG to promote the non-amyloidogenic pathway,
Fe2+ in solution is Desferal (DFO) 1 M30 6 VK28 6 HLA20 1
EGCG. via a PKC-dependent activation of -secretase, thereby
increasing sAPP [46]. sAPP has been demonstrated to
posses potent neuroprotective activities against excitotox-
ic and oxidative insults in various cellular models [109]
A possible novel promising therapeutic approach for and it was shown to protect against p53-mediated apop-
treating AD, PD and ALS with non-toxic, brain perme- tosis [110]. Moreover, sAPP promotes neurite outgrowth
able metal chelators could be the use of the naturally oc- [111], regulates synaptogenesis [112] and exerts trophic
curring polyphenols, such as EGCG and curcumin, which effects on cerebral neurons in culture. As sAPP and A
by being of natural origin may not exert toxic side effects are formed by two mutually exclusive mechanisms, stim-
inherent to synthetic drugs. Both compounds have well- ulation of the secretory processing of sAPP might pre-
characterized antioxidant, metal (iron and copper) che- vent the formation of the amyloidogenic A. Thus, EGCG
lating and anti-inflammatory activities [12, 100] and may influence A levels, either via translational inhibition
have been demonstrated to exert neuroprotective activity of APP or by regulating APP processing. Finally, iron che-
against a variety of neurotoxic insults, as well as to regu- lation by EGCG may ablate Fe3+-induced aggregation of
late APP processing and A burden in cell culture and in hyperphosphorylated tau (PHF), the major constituent

Multifunctional Catechins for Neurosignals 2005;14:46–60 53


Neuroprotection
A B

holoAPP ➤ holoAPP ➤

Fig. 4. Effect of EGCG and M30 on APP


processing in mice hippocampus. Repre- β-actin ➤ β-actin ➤
sentative Western blots of levels of holo-
APP in the membrane compartment, ob- 120 120
tained from hippocampus of mice treated
100 100
with EGCG (2 mg/kg) (A), or with M30

% of control

% of control
(5 mg/kg) (B) for 14 days detected with 80 80
22C11 antibody (directed to the to APP ✽✽
60 ✽✽ 60
N-terminus) or with a C-terminus APP an-
tibody, respectively. Densitometric analy- 40 40
sis is expressed as percent of the control, 20 20
untreated animals after normalizing to the
levels of -actin. Data are expressed as the 0
Control EGCG
0
Control M30
mean 8 SEM (n = 6 mice in each group). (2 mg/kg) (5 mg/kg)
** p ! 0.03 vs. control. Figure drawn using
information from Levites et al. [46].

Fig. 5. Proposed schematic model for


EGCG neuroprotective effect via regula-
tion of APP processing and A formation.
d Increased levels/activity, f decreased lev-
els/activity. For full explanation, see text.

of neurofibrillary tangles in AD brains [113] (a descrip- pacta (pc) of PD patients [17, 27, 114]. Specifically, re-
tive explanation is depicted in fig. 5). dox-active iron has been observed in the rim of Lewy
body, the morphological hallmark of PD, also composed
Iron Chelation for PD of lipids, aggregated -synuclein (concentrating in its pe-
Numerous studies have shown that there is a progres- ripheral halo) and ubiquitinated, hyperphosphorylated
sive accumulation of iron and ferritin in the SN pars com- neurofilament proteins [115]. -Synuclein associated

54 Neurosignals 2005;14:46–60 Mandel/Avramovich-Tirosh/Reznichenko/


Zheng/Weinreb/Amit/Youdim
Fig. 6. Possible mechanism of neurotoxin-induced iron uptake, glutathione (GHS), the rate-limiting cofactor of glutathione per-
release and interaction with -synuclein resulting in OS initiated oxidase, the main enzymatic pathway in the brain, to eliminate
neurodegeneration and its prevention by iron-chelating/antioxi- hydrogen peroxide. Labile pool of iron can also cause aggregation
dants. The mechanism by which 6-OHDA and MPTP induce in- of -synuclein to the neurotoxic form, which can also generate OH.
crease of iron in substantia nigra pars compacta and within the The net effect is oxidative-stress-dependent damage to neuron an-
melanin-containing neurons is not known. These neurotoxins may tioxidant mechanism, membrane lipid peroxidation, demise of cell
(a) activate the divalent metal transporter 1 (DMT1) which is re- and mitochondrial membrane, protein misfolding and ultimate cell
sponsible for iron transport into the brain across the cell membrane; death. Neuroprotective agents that can be used to prevent iron-in-
(b) alter the blood-brain barrier (BBB), thereby allowing iron access duced neurodegeneration include M30 and HLA20 (bifunctional
to the brain; (c) induce release of iron from ferritin which enters iron chelator-MAO inhibitors); desferal, VK-28, R-APO (R-apo-
the labile (redox-active) pool of iron. It is the labile pool of iron morphine) and EGCG (iron chelators); R-APO, EGCG, melatonin
which can initiate the Fenton chemistry in response to the presence and Vit E (vitamin E) (radical scavengers). Sharp arrows indicate
of hydrogen peroxide, thus generating the highly reactive hydroxyl positive inputs, whereas blunt arrows are for inhibitory inputs. Re-
radical (OH). The resultant effect is the depletion of cell-reduced produced, with minor modifications, from Youdim et al. [106].

with presynaptic membrane is not toxic; however, a num- on the key iron and oxygen sensor EGLN1 gene coding
ber of recent studies [116–118] have shown that it forms for an isoform of 2-oxoglutarate-dependent dioxygenase
toxic aggregates in the presence of iron and this is consid- hydroxylase (see previous section). Excessive production
ered to contribute to the formation of Lewy body via OS, of EGLN1 hydroxylase in the SNpc may lead to a fall in
being one of its constituents. IRP2 and subsequent decrease in transferrin receptor
Our recent high throughput gene expression study in (TfR) mRNA and increase in ferritin levels, both sub-
the SNpc of Parkinsonian brains employing Affymetrix jected to positive and negative transcriptional regulation
chip technology [29] has revealed a significant increase by IRP2, respectively [119, 120]. Recent studies in knock-

Multifunctional Catechins for Neurosignals 2005;14:46–60 55


Neuroprotection
out mice for IRP2 have revealed accumulation of iron in tects rats against kainate and 6-OHDA [129]. Figure 6
the striatum with substantial bradykinesia and tremor summarizes the mechanism of neurotoxin-induced iron
[121]. uptake, release and interaction with -synuclein resulting
Consistent with the pivotal role of iron in neurodegen- in OS initiated neurodegeneration and its prevention by
eration is the finding that the iron chelator desferal pre- iron-chelating/antioxidant agents.
vents cytochrome c-induced--synuclein aggregation and
toxicity in vitro [122] and attenuates dopaminergic neu-
rotoxicity in response to neurotoxins MPTP and 6-OHDA Conclusions
in vivo [123, 124]. In line with the iron-chelating feature
of EGCG, this polyphenol was shown to prevent -synu- It is likely that syndromes such as AD and PD will re-
clein accumulation and to attenuate IRP2 depletion, in quire multiple- drug therapy to address the varied patho-
the SNpc of mice intoxicated with MPTP, when given logical aspects of the disease. Therefore, the use of com-
orally for a period of 2 weeks [62]. This may be associ- pounds with poly-pharmacological activities or cocktail
ated to our previous studies where green tea extract or of drugs is a promising therapeutic approach for the treat-
EGCG prevented DA-containing neuron degeneration ment of neurodegenerative diseases. Indeed, a wealth of
and tyrosine hydroxylase activity decrease [30]. In spite new data indicates that green tea catechins are being rec-
of the absence of clinical trials regarding tea polyphenols ognized as multifunctional compounds for neuroprotec-
and PD, epidemiological studies have shown reduced risk tion. They act as radical scavengers, iron chelators and
of PD associated with consumption of 2 cups/day or more modulators of pro-survival genes, and PKC signaling
of tea [125] and a much lower prevalence of PD in Chi- pathway. The use of EGCG as a natural, non-toxic, lipo-
nese population than in white people [126, 127]. philic brain permeable neuroprotective drug is advocated
Additional studies examining the effect of iron chela- for ‘ironing out iron’ from those brain areas where it pref-
tion, by either transgenic expression of the iron-binding erentially accumulates in neurodegenerative diseases
protein, ferritin, or oral administration of the metal che- [106]. Thus, green tea catechins may have potential dis-
lator clioquinol, have shown significant attenuation of ease-modifying action. Future efforts in the understand-
MPTP-induced neurotoxicity [128]. These findings have ing of the protective mechanism of action of these poly-
now been substantiated with systemic injection of the phenols should concentrate on deciphering the cellular
brain-permeable iron chelator, VK-28, to rats in response targets affected by these compounds and other neuropro-
to 6-OHDA [104]. In line, nutritional iron deficiency pro- tectants.

References

1 Butterfield D, Castegna A, Pocernich C, Drake 6 Jiang F, Dusting GJ: Natural phenolic com- 11 Grinberg LN, Newmark H, Kitrossky N, Ra-
J, Scapagnini G, Calabrese V: Nutritional ap- pounds as cardiovascular therapeutics: Poten- hamim E, Chevion M, Rachmilewitz EA: Pro-
proaches to combat oxidative stress in Alz- tial role of their anti-inflammatory effects. tective effects of tea polyphenols against oxida-
heimer’s disease. J Nutr Biochem 2002; 13: Curr Vasc Pharmacol 2003;1:135–156. tive damage to red blood cells. Biochem
444. 7 Guo Q, Zhao B, Shen S, Hou J, Hu J, Xin W: Pharmacol 1997;54:973–978.
2 Wang ZY, Huang MT, Lou YR, Xie JG, Reuhl ESR study on the structure-antioxidant activity 12 Mandel S, Weinreb O, Amit T, Youdim MBH:
KR, Newmark HL, Ho CT, Yang CS, Conney relationship of tea catechins and their epimers. Cell signaling pathways in the neuroprotective
AH: Inhibitory effects of black tea, green tea, Biochim Biophys Acta 1999;1427:13–23. actions of the green tea polyphenol (–)-epigal-
decaffeinated black tea, and decaffeinated 8 Hider RC, Liu ZD, Khodr HH: Metal chela- locatechin-3-gallate: implications for neurode-
green tea on ultraviolet B light-induced skin tion of polyphenols. Methods Enzymol 2001; generative diseases. J Neurochem 2004; 88:
carcinogenesis in 7,12-dimethylbenz[a]anthra 335:190–203. 1555–1569.
cene-initiated SKH-1 mice. Cancer Res 1994; 9 Guo Q, Zhao B, Li M, Shen S, Xin W: Studies 13 Weinreb O, Mandel S, Amit T, Youdim MB:
54:3428–3455. on protective mechanisms of four components Neurological mechanisms of green tea poly-
3 Yang CS, Wang ZY: Tea and cancer. J Natl of green tea polyphenols against lipid peroxida- phenols in Alzheimer’s and Parkinson’s dis-
Cancer Inst 1993;85:1038–1049. tion in synaptosomes. Biochim Biophys Acta eases. J Nutr Biochem 2004;15:506–516.
4 Wiseman SA, Balentine DA, Frei B: Antioxi- 1996;1304:210–222. 14 Suganuma M, Okabe S, Oniyama M, Tada Y,
dants in tea. Crit Rev Food Sci Nutr 1997;37: 10 Kumamoto M, Sonda T, Nagayama K, Tabata Ito H, Fujiki H: Wide distribution of [3H](–)-
705–718. M: Effects of pH and metal ions on antioxida- epigallocatechin gallate, a cancer preventive
5 Higdon JV, Frei B: Tea catechins and polyphe- tive activities of catechins. Biosci Biotechnol tea polyphenol, in mouse tissue. Carcinogene-
nols: health effects, metabolism, and antioxi- Biochem 2001;65:126–132. sis 1998;19:1771–1776.
dant functions. Crit Rev Food Sci Nutr 2003;
43:89–143.

56 Neurosignals 2005;14:46–60 Mandel/Avramovich-Tirosh/Reznichenko/


Zheng/Weinreb/Amit/Youdim
15 Nakagawa K, Miyazawa T: Absorption and 27 Riederer P, Sofic E, Rausch WD, Schmidt B, 40 Schroeter H, Spencer JP, Rice-Evans C, Wil-
distribution of tea catechin, (–)-epigallocate- Reynolds GP, Jellinger K, Youdim MBH: liams RJ: Flavonoids protect neurons from
chin-3-gallate, in the rat. J Nutr Sci Vitaminol Transition metals, ferritin, glutathione, and oxidized low-density-lipoprotein-induced
(Tokyo) 1997;43:679–684. ascorbic acid in parkinsonian brains. J Neuro- apoptosis involving c-Jun N-terminal kinase
16 Abd El Mohsen MM, Kuhnle G, Rechner AR, chem 1989;52:515–520. (JNK), c-Jun and caspase-3. Biochem J 2001;
Schroeter H, Rose S, Jenner P, Rice-Evans CA: 28 Jenner P, Olanow CW: Oxidative stress and 358:547–557.
Uptake and metabolism of epicatechin and its the pathogenesis of Parkinson’s disease. Neu- 41 Spencer JP, Schroeter H, Kuhnle G, Srai SK,
access to the brain after oral ingestion. Free rology 1996;47:S161–170. Tyrrell RM, Hahn U, Rice-Evans C: Epicate-
Rad Biol Med 2002;33:1693–1702. 29 Grunblatt E, Mandel S, Jacob-Hirsch J, Zelig- chin and its in vivo metabolite, 3-O-methyl epi-
17 Zecca L, Youdim MB, Riederer P, Connor JR, son S, Amariglo N, Rechavi G, Li J, Ravid R, catechin, protect human fibroblasts from oxida-
Crichton RR: Iron, brain ageing and neurode- Roggendorf W, Riederer P, Youdim MB: Gene tive-stress-induced cell death involving
generative disorders. Nat Rev Neurosci 2004; expression profiling of parkinsonian substantia caspase-3 activation. Biochem J 2001;354:493–
5:863–873. nigra pars compacta: Alterations in ubiquitin- 500.
18 Blum D, Torch S, Lambeng N, Nissou M, Ben- proteasome, heat shock protein, iron and oxi- 42 Nobre Junior HV, Cunha GM, Maia FD,
abid AL, Sadoul R, Verna JM: Molecular path- dative stress regulated proteins, cell adhesion/ Oliveira RA, Moraes MO, Rao VS: Catechin
ways involved in the neurotoxicity of 6-OHDA, cellular matrix and vesicle trafficking genes. J attenuates 6-hydroxydopamine (6-OHDA)-in-
dopamine and MPTP: Contribution to the Neural Transm 2004;111:1543–1573. duced cell death in primary cultures of mesen-
apoptotic theory in Parkinson’s disease. Prog 30 Levites Y, Weinreb O, Maor G, Youdim MBH, cephalic cells. Comp Biochem Physiol [C]
Neurobiol 2001;65:135–172. Mandel S: Green tea polyphenol (–)-epigallo- 2003;136:175–180.
19 Linazasoro G: Neuroprotection in Parkinson’s catechin-3-gallate prevents N-methyl-4-phe- 43 Mercer LD, Kelly BL, Horne MK, Beart PM:
disease: Love story or mission impossible? Ex- nyl-1,2,3,6-tetrahydropyridine-induced dopa- Dietary polyphenols protect dopamine neu-
pert Rev Neurotherapeut 2002;2:403–416. minergic neurodegeneration. J Neurochem rons from oxidative insults and apoptosis: In-
20 McNaught KS, Belizaire R, Jenner P, Olanow 2001;78:1073–1082. vestigations in primary rat mesencephalic cul-
CW, Isacson O: Selective loss of 20S protea- 31 Komatsu M, Hiramatsu M: The efficacy of an tures. Biochem Pharmacol 2005;69:339–345.
some alpha-subunits in the substantia nigra antioxidant cocktail on lipid peroxide level and 44 Levites Y, Amit T, Youdim MBH, Mandel S:
pars compacta in Parkinson’s disease. Neuro- superoxide dismutase activity in aged rat brain Involvement of protein kinase C activation
sci Lett 2002;326:155–158. and DNA damage in iron-induced epilepto- and cell survival/cell cycle genes in green tea
21 Olanow CW, Youdim MB: Iron and neurode- genic foci. Toxicology 2000;148:143–148. polyphenol (–)-epigallocatechin-3-gallate neu-
generation: Prospects for neuroprotection; in 32 Pan T, Fei J, Zhou X, Jankovic J, Le W: Effects roprotective action. J Biol Chem 2002; 277:
Olanow CW, Jenner P, Youdim MB (eds): of green tea polyphenols on dopamine uptake 30574–30580.
Neurodegeneration and Neuroprotection in and on MPP+-induced dopamine neuron in- 45 Choi YT, Jung CH, Lee SR, Bae JH, Baek WK,
Parkinson’s Disease. London, Academic Press, jury. Life Sci 2003;72:1073–1083. Suh MH, Park J, Park CW, Suh SI: The green
1996, pp 55–69. 33 Lu H, Meng X, Yang CS: Enzymology of meth- tea polyphenol (–)-epigallocatechin gallate at-
22 Mandel S, Grunblatt E, Youdim MBH: cDNA ylation of tea catechins and inhibition of cat- tenuates beta-amyloid-induced neurotoxicity
microarray to study gene expression of dopa- echol-O-methyltransferase by (–)-epigallocat- in cultured hippocampal neurons. Life Sci
minergic neurodegeneration and neuroprotec- echin gallate. Drug Metab Dispos 2003; 31: 2001;70:603–614.
tion in MPTP and 6-hydroxydopamine mod- 572–579. 46 Levites Y, Amit T, Mandel S, Youdim MBH:
els: Implications for idiopathic Parkinson’s 34 Lee S, Suh S, Kim S: Protective effects of the Neuroprotection and neurorescue against am-
disease. J Neural Transm Suppl 2000;60:117– green tea polyphenol (-)-epigallocatechin gal- yloid beta toxicity and PKC-dependent re-
124. late against hippocampal neuronal damage af- lease of non-amyloidogenic soluble precusor
23 Dauer W, Przedborski S: Parkinson’s disease: ter transient global ischemia in gerbils. Neuro- protein by green tea polyphenol (–)-epigallo-
Mechanisms and models. Neuron 2003; 39: sci Lett 2000;287:191–194. catechin-3-gallate. FASEB J 2003; 17: 952–
889–909. 35 Lee H, Bae JH, Lee SR: Protective effect of 954.
24 Bonifati V, Rizzu P, van Baren MJ, Schaap O, green tea polyphenol EGCG against neuronal 47 Reznichenko L, Amit T, Youdim MB, Man-
Breedveld GJ, Krieger E, Dekker MC, Squi- damage and brain edema after unilateral cere- del S: Green tea polyphenol (–)-epigallocate-
tieri F, Ibanez P, Joosse M, van Dongen JW, bral ischemia in gerbils. J Neurosci Res 2004; chin-3-gallate induces neurorescue of long-
Vanacore N, van Swieten JC, Brice A, Meco G, 77:892–900. term serum-deprived PC12 cells and promotes
van Duijn CM, Oostra BA, Heutink P: Muta- 36 Suzuki M, Tabuchi M, Ikeda M, Umegaki K, neurite outgrowth. J Neurochem 2005;in
tions in the DJ-1 gene associated with autoso- Tomita T: Protective effects of green tea cate- press.
mal recessive early-onset parkinsonism. Sci- chins on cerebral ischemic damage. Med Sci 48 Weinreb O, Mandel S, Youdim MBH: cDNA
ence 2003;299:256–259. Monit 2004;10:BR166–174. gene expression profile homology of antioxi-
25 Valente EM, Abou-Sleiman PM, Caputo V, 37 Sutherland BA, Shaw OM, Clarkson AN, Jack- dants and their anti-apoptotic and pro-apop-
Muqit MM, Harvey K, Gispert S, Ali Z, Del son DM, Sammut IA, Appleton I: Neuropro- totic activities in human neuroblastoma cells.
Turco D, Bentivoglio AR, Healy DG, Albanese tective effects of (–)-epigallocatechin gallate FASEB J 2003;17:935–937.
A, Nussbaum R, Gonzalez-Maldonado R, Del- after hypoxia-ischemia-induced brain damage: 49 Halliwell B: Vitamin C: antioxidant or pro-oxi-
ler T, Salvi S, Cortelli P, Gilks WP, Latchman novel mechanisms of action. Faseb J 2004. dant in vivo? Free Rad Res 1996;25:439–454.
DS, Harvey RJ, Dallapiccola B, Auburger G, 38 Townsend PA, Scarabelli TM, Davidson SM, 50 Gassen M, Pinchasi B, Youdim MB: Apomor-
Wood NW: Hereditary early-onset Parkinson’s Knight RA, Latchman DS, Stephanou A: phine is a potent radical scavenger and protects
disease caused by mutations in PINK1. Sci- STAT-1 interacts with p53 to enhance DNA cultured pheochromocytoma cells from 6-
ence 2004;304:1158–1160. damage-induced apoptosis. J Biol Chem 2004; OHDA and H2O2-induced cell death. Adv
26 Shults CW, Oakes D, Kieburtz K, Beal MF, 279:5811–5820. Pharmacol 1998;42:320–324.
Haas R, Plumb S, Juncos JL, Nutt J, Shoulson 39 Aktas O, Prozorovski T, Smorodchenko A, 51 Gassen M, Gross A, Youdim MB: Apomor-
I, Carter J, Kompoliti K, Perlmutter JS, Reich Savaskan NE, Lauster R, Kloetzel PM, Infan- phine enantiomers protect cultured pheochro-
S, Stern M, Watts RL, Kurlan R, Molho E, te-Duarte C, Brocke S, Zipp F: Green tea epi- mocytoma (PC12) cells from oxidative stress
Harrison M, Lew M: Effects of coenzyme Q10 gallocatechin-3-gallate mediates T cellular NF- induced by H2O2 and 6-hydroxydopamine.
in early Parkinson disease: Evidence of slowing kappa B inhibition and exerts neuroprotection Mov Disord 1998;13:242–248.
of the functional decline. Arch Neurol 2002; in autoimmune encephalomyelitis. J Immunol
59:1541–1550. 2004;173:5794–5800.

Multifunctional Catechins for Neurosignals 2005;14:46–60 57


Neuroprotection
52 Galati G, O’Brien PJ: Potential toxicity of fla- 65 Kim SY, Ahn BH, Kim J, Bae YS, Kwak JY, 76 Yu R, Jiao JJ, Duh JL, Gudehithlu K, Tan TH,
vonoids and other dietary phenolics: Signifi- Min G, Kwon TK, Chang JS, Lee YH, Yoon Kong AN: Activation of mitogen-activated
cance for their chemopreventive and antican- SH, Min do S: Phospholipase C, protein kinase protein kinases by green tea polyphenols: Po-
cer properties. Free Radic Biol Med 2004; 37: C, Ca2+/calmodulin-dependent protein kinase tential signaling pathways in the regulation
287–303. II, and redox state are involved in epigallocat- of antioxidant-responsive element-mediated
53 Wiseman S, Mulder T, Rietveld A: Tea flavo- echin gallate-induced phospholipase D activa- phase II enzyme gene expression. Carcinogen-
noids: Bioavailability in vivo and effects on cell tion in human astroglioma cells. Eur J Biochem esis 1997;18:451–456.
signaling pathways in vitro. Antioxid Redox 2004;271:3470–3480. 77 Chen C, Yu R, Owuor ED, Kong AN: Activa-
Signal 2001;3:1009–1021. 66 Kumazawa S, Kajiya K, Naito A, Saito H, Tuzi tion of antioxidant-response element (ARE),
54 Durkin JP, Tremblay R, Chakravarthy B, S, Tanio M, Suzuki M, Nanjo F, Suzuki E, Na- mitogen-activated protein kinases (MAPKs)
Mealing G, Morley P, Small D, Song D: Evi- kayama T: Direct evidence of interaction of a and caspases by major green tea polyphenol
dence that the early loss of membrane protein green tea polyphenol, epigallocatechin gallate, components during cell survival and death.
kinase C is a necessary step in the excitatory with lipid bilayers by solid-state nuclear mag- Arch Pharm Res 2000;23:605–612.
amino acid-induced death of primary cortical netic resonance. Biosci Biotechnol Biochem 78 Owuor ED, Kong AN: Antioxidants and oxi-
neurons. J Neurochem 1997;68:1400–1412. 2004;68:1743–1747. dants regulated signal transduction pathways.
55 Maher P: How protein kinase C activation pro- 67 Saito H, Tabeta R, Kodama M, Nagata C, Sato Biochem Pharmacol 2002;64:765–770.
tects nerve cells from oxidative stress-induced Y: Direct evidence of incorporation of 12-O- 79 Chung JH, Han JH, Hwang EJ, Seo JY, Cho
cell death. J Neurosci 2001;21:2929–2938. [20-2H1]tetradecanoylphorbol-13-acetate into KH, Kim KH, Youn JI, Eun HC: Dual mecha-
56 Vianna MR, Barros DM, Silva T, Choi H, artificial membranes as determined by deute- nisms of green tea extract-induced cell surviv-
Madche C, Rodrigues C, Medina JH, Izquier- rium magnetic resonance. Cancer Lett 1984; al in human epidermal keratinocytes. FASEB
do I: Pharmacological demonstration of the 22:65–69. J 2003;17:1913–1915
differential involvement of protein kinase C 68 Gary DS, Milhavet O, Camandola S, Mattson 80 Lorenz M, Wessler S, Follmann E, Michae-
isoforms in short- and long-term memory for- MP: Essential role for integrin linked kinase in lis W, Dusterhoft T, Baumann G, Stangl K,
mation and retrieval of one-trial avoidance in Akt-mediated integrin survival signaling in Stangl V: A constituent of green tea, epigallo-
rats. Psychopharmacology (Berl) 2000; 150: hippocampal neurons. J Neurochem 2003;84: catechin-3-gallate, activates endothelial nitric
77–84. 878–890. oxide synthase by a PI3K-, PKA-, and Akt-de-
57 Lange-Asschenfeldt C, Raval AP, Dave KR, 69 Kermer P, Klocker N, Labes M, Bahr M: Insu- pendent pathway, and leads to endothelial-de-
Mochly-Rosen D, Sick TJ, Perez-Pinzon MA: lin-like growth factor-I protects axotomized rat pendent vasorelaxation. J Biol Chem 2003.
Epsilon protein kinase C mediated ischemic retinal ganglion cells from secondary death via 81 Tedeschi E, Menegazzi M, Yao Y, Suzuki H,
tolerance requires activation of the extracellu- PI3-K-dependent Akt phosphorylation and in- Forstermann U, Kleinert H: Green tea inhibits
lar regulated kinase pathway in the organotyp- hibition of caspase-3 in vivo. J Neurosci 2000; human inducible nitric-oxide synthase expres-
ic hippocampal slice. J Cereb Blood Flow 20:2–8. sion by down-regulating signal transducer and
Metab 2004;24:636–645. 70 Singer CA, Figueroa-Masot XA, Batchelor activator of transcription-1alpha activation.
58 Cordey M, Gundimeda U, Gopalakrishna R, RH, Dorsa DM: The mitogen-activated pro- Mol Pharmacol 2004;65:111–120.
Pike CJ: Estrogen activates protein kinase C in tein kinase pathway mediates estrogen neuro- 82 Bernardi P, Petronilli V, Di Lisa F, Forte M: A
neurons: Role in neuroprotection. J Neuro- protection after glutamate toxicity in primary mitochondrial perspective on cell death.
chem 2003;84:1340–1348. cortical neurons. J Neurosci 1999; 19: 2455– Trends Biochem Sci 2001;26:112–117.
59 Han YS, Zheng WH, Bastianetto S, Chabot JG, 2463. 83 Cory S, Adams JM: The Bcl2 family: Regula-
Quirion R: Neuroprotective effects of resvera- 71 Schroeter H, Boyd C, Spencer JP, Williams RJ, tors of the cellular life-or-death switch. Nat
trol against beta-amyloid-induced neurotoxic- Cadenas E, Rice-Evans C: MAPK signaling in Rev Cancer 2002;2:647–656.
ity in rat hippocampal neurons: involvement neurodegeneration: Influences of flavonoids 84 Merry DE, Korsmeyer SJ: Bcl-2 gene family in
of protein kinase C. Br J Pharmacol 2004;141: and of nitric oxide. Neurobiol Aging 2002;23: the nervous system. Annu Rev Neurosci 1997;
997–1005. 861–880. 20:245–267.
60 Youdim MBH: Rasagiline: An anti-Parkinson 72 Vaudry D, Stork PJ, Lazarovici P, Eiden LE: 85 DeFeudis FV, Drieu K: Ginkgo biloba extract
drug with neuroprotective activity. Expert Rev Signaling pathways for PC12 cell differentia- (EGb 761) and CNS functions: Basic studies
Neurotherapeut 2003;3:737–749. tion: Making the right connections. Science and clinical applications. Curr Drug Targets
61 Weinreb O, Bar-Am O, Amit T, Chillag-Tal- 2002;296:1648–1649. 2000;1:25–58.
mor O, Youdim MB: Neuroprotection via pro- 73 Harris CA, Deshmukh M, Tsui-Pierchala B, 86 Ishige K, Schubert D, Sagara Y: Flavonoids
survival protein kinase C isoforms associated Maroney AC, Johnson EM Jr: Inhibition of the protect neuronal cells from oxidative stress by
with Bcl-2 family members. FASEB J 2004;18: c-Jun N-terminal kinase signaling pathway by three distinct mechanisms. Free Radic Biol
1471–1473. the mixed lineage kinase inhibitor CEP-1347 Med 2001;30:433–446.
62 Mandel S, Maor G, Youdim MBH: Iron and (KT7515) preserves metabolism and growth of 87 Lee JH, Song DK, Jung CH, Shin DH, Park J,
alpha-synuclein in the substantia Nigra of trophic factor-deprived neurons. J Neurosci Kwon TK, Jang BC, Mun KC, Kim SP, Suh SI,
MPTP-treated mice: Effect of neuroprotective 2002;22:103–113. Bae JH: (–)-Epigallocatechin gallate attenuates
drugs R-apomorphine and green tea polyphe- 74 Alessandrini A, Namura S, Moskowitz MA, glutamate-induced cytotoxicity via intracellu-
nol (–)-epigallocatechin-3-gallate. J Mol Neu- Bonventre JV: MEK1 protein kinase inhibi- lar Ca modulation in PC12 cells. Clin Exp
rosci 2004;24:401–416. tion protects against damage resulting from fo- Pharmacol Physiol 2004;31:530–536.
63 Ruvolo PP, Deng X, Carr BK, May WS: A cal cerebral ischemia. Proc Natl Acad Sci USA 88 Bush AI: The metallobiology of Alzheimer’s
functional role for mitochondrial protein ki- 1999;96:12866–12869. disease. Trends Neurosci 2003;26:207–214.
nase Calpha in Bcl2 phosphorylation and sup- 75 Yun HY, Gonzalez-Zulueta M, Dawson VL, 89 Atwood CS, Obrenovich ME, Liu T, Chan H,
pression of apoptosis. J Biol Chem 1998; 273: Dawson TM: Nitric oxide mediates N-methyl- Perry G, Smith MA, Martins RN: Amyloid-
25436–25442. D-aspartate receptor-induced activation of beta: A chameleon walking in two worlds: A
64 Jiffar T, Kurinna S, Suck G, Carlson-Bremer p21ras. Proc Natl Acad Sci USA 1998; 95: review of the trophic and toxic properties of
D, Ricciardi MR, Konopleva M, Andreeff M, 5773–5778. amyloid-beta. Brain Res Brain Res Rev 2003;
Ruvolo PP: PKC alpha mediates chemoresis- 43:1–16.
tance in acute lymphoblastic leukemia through
effects on Bcl2 phosphorylation. Leukemia
2004;18:505–512.

58 Neurosignals 2005;14:46–60 Mandel/Avramovich-Tirosh/Reznichenko/


Zheng/Weinreb/Amit/Youdim
90 Rogers JT, Randall JD, Cahill CM, Eder PS, 102 Dedeoglu A, Cormier K, Payton S, Tseitlin 113 Yamamoto A, Shin RW, Hasegawa K, Naiki
Huang X, Gunshin H, Leiter L, McPhee J, KA, Kremsky JN, Lai L, Li X, Moir RD, Tan- H, Sato H, Yoshimasu F, Kitamoto T: Iron
Sarang SS, Utsuki T, Greig NH, Lahiri DK, zi RE, Bush AI, Kowall NW, Rogers JT, (III) induces aggregation of hyperphosphory-
Tanzi RE, Bush AI, Giordano T, Gullans SR: Huang X: Preliminary studies of a novel bi- lated tau and its reduction to iron (II) revers-
An iron-responsive element type II in the 5- functional metal chelator targeting Alzheim- es the aggregation: implications in the forma-
untranslated region of the Alzheimer’s amy- er’s amyloidogenesis. Exp Gerontol 2004;39: tion of neurofibrillary tangles of Alzheimer’s
loid precursor protein transcript. J Biol Chem 1641–1649. disease. J Neurochem 2002;82:1137–1147.
2002;277:45518–45528. 103 Youdim MB, Fridkin M, Zheng H: Bifunc- 114 Gerlach M, Ben-Shachar D, Riederer P,
91 Sharp FR, Bernaudin M: HIF1 and oxygen tional drug derivatives of MAO-B inhibitor Youdim MB: Altered brain metabolism of
sensing in the brain. Nat Rev Neurosci 2004; rasagiline and iron chelator VK-28 as a more iron as a cause of neurodegenerative diseases?
5:437–448. effective approach to treatment of brain age- J Neurochem 1994;63:793–807.
92 Lee JW, Bae SH, Jeong JW, Kim SH, Kim ing and ageing neurodegenerative diseases. 115 Jellinger KA: Neuropathological spectrum of
KW: Hypoxia-inducible factor (HIF-1)alpha: Mech Age Dev 2005;126:317–326. synucleinopathies. Mov Disord 2003; 18
Its protein stability and biological functions. 104 Shachar DB, Kahana N, Kampel V, War- (suppl 6):2–12.
Exp Mol Med 2004;36:1–12. shawsky A, Youdim MB: Neuroprotection by 116 Turnbull S, Tabner BJ, El-Agnaf OM, Moore
93 Jaakkola P, Mole DR, Tian YM, Wilson MI, a novel brain permeable iron chelator, VK- S, Davies Y, Allsop D: Alpha-Synuclein im-
Gielbert J, Gaskell SJ, Kriegsheim A, Hebe- 28, against 6-hydroxydopamine lesion in rats. plicated in Parkinson’s disease catalyses the
streit HF, Mukherji M, Schofield CJ, Max- Neuropharmacology 2004;46:254–263. formation of hydrogen peroxide in vitro. Free
well PH, Pugh CW, Ratcliffe PJ: Targeting of 105 Youdim MB, Fridkin M, Zheng H: Novel bi- Radic Biol Med 2001;30:1163–1170.
HIF-alpha to the von Hippel-Lindau ubiqui- functional drugs targeting monoamine oxi- 117 Ostrerova-Golts N, Petrucelli L, Hardy J, Lee
tylation complex by O2-regulated prolyl hy- dase inhibition and iron chelation as an ap- JM, Farer M, Wolozin B: The A53T alpha-
droxylation. Science 2001;292:468–472. proach to neuroprotection in Parkinson’s synuclein mutation increases iron-dependent
94 Minchenko O, Opentanova I, Caro J: Hypox- disease and other neurodegenerative diseas- aggregation and toxicity. J Neurosci 2000;20:
ic regulation of the 6-phosphofructo-2-ki- es. J Neural Transm 2004;111:1455–1471. 6048–6054.
nase/fructose-2,6-bisphosphatase gene family 106 Youdim MBH, Buccafusco JJ: Multi-func- 118 Ebadi M, Govitrapong P, Sharma S, Murali-
(PFKFB-1-4) expression in vivo. FEBS Lett tional drugs for various CNS targets in the krishnan D, Shavali S, Pellett L, Schafer R,
2003;554:264–270. treatment of neurodegenerative disorders. Albano C, Eken J: Ubiquinone (coenzyme
95 Hanson ES, Leibold EA: Regulation of the Trends Pharmacol Sci 2005;26:27–35. q10) and mitochondria in oxidative stress of
iron regulatory proteins by reactive nitrogen 107 Zheng H, Weiner LM, Bar-Am O, Epsztejn S, Parkinson’s disease. Biol Signals Recept
and oxygen species. Gene Expr 1999;7:367– Cabantchik ZI, Warshawsky A, Youdim MB, 2001;10:224–253.
376. Fridkin M: Design, synthesis, and evaluation 119 Ponka P: Hereditary causes of disturbed iron
96 Wang J, Chen G, Muckenthaler M, Galy B, of novel bifunctional iron-chelators as poten- homeostasis in the central nervous system.
Hentze MW, Pantopoulos K: Iron-mediated tial agents for neuroprotection in Alzhei- Ann NY Acad Sci 2004;1012:267–281.
degradation of IRP2, an unexpected pathway mer’s, Parkinson’s, and other neurodegenera- 120 Meyron-Holtz EG, Ghosh MC, Iwai K, La-
involving a 2-oxoglutarate-dependent oxy- tive diseases. Bioorg Med Chem 2005; 13: Vaute T, Brazzolotto X, Berger UV, Land W,
genase activity. Mol Cell Biol 2004; 24: 954– 773–783. Ollivierre-Wilson H, Grinberg A, Love P,
965. 108 Ono K, Yoshiike Y, Takashima A, Hasegawa Rouault TA: Genetic ablations of iron regula-
97 Crapper McLachlan DR, Dalton AJ, Kruck K, Naiki H, Yamada M: Potent anti-amy- tory proteins 1 and 2 reveal why iron regula-
TP, Bell MY, Smith WL, Kalow W, Andrews loidogenic and fibril-destabilizing effects of tory protein 2 dominates iron homeostasis.
DF: Intramuscular desferrioxamine in pa- polyphenols in vitro: Implications for the pre- EMBO J 2004;23:386–395.
tients with Alzheimer’s disease. Lancet 1991; vention and therapeutics of Alzheimer’s dis- 121 LaVaute T, Smith S, Cooperman S, Iwai K,
337:1304–1308. ease. J Neurochem 2003;87:172–181. Land W, Meyron-Holtz E, Drake SK, Miller
98 Ritchie CW, Bush AI, Mackinnon A, Macfar- 109 Mattson MP, Barger SW, Furukawa K, Bruce G, Abu-Asab M, Tsokos M, Switzer R 3rd,
lane S, Mastwyk M, MacGregor L, Kiers L, AJ, Wyss-Coray T, Mark RJ, Mucke L: Cel- Grinberg A, Love P, Tresser N, Rouault TA:
Cherny R, Li QX, Tammer A, Carrington D, lular signaling roles of TGF beta, TNF alpha Targeted deletion of the gene encoding iron
Mavros C, Volitakis I, Xilinas M, Ames D, and beta APP in brain injury responses and regulatory protein-2 causes misregulation of
Davis S, Beyreuther K, Tanzi RE, Masters Alzheimer’s disease. Brain Res Rev 1997;23: iron metabolism and neurodegenerative dis-
CL: Metal-protein attenuation with iodo- 47–61. ease in mice. Nat Genet 2001;27:209–214.
chlorhydroxyquin (clioquinol) targeting a 110 Xu X, Yang D, Wyss-Coray T, Yan J, Gan L, 122 Hashimoto M, Takeda A, Hsu LJ, Takenou-
beta amyloid deposition and toxicity in Al- Sun Y, Mucke L: Wild-type but not Alzhei- chi T, Masliah E: Role of cytochrome c as a
zheimer disease: A pilot phase 2 clinical trial. mer-mutant amyloid precursor protein con- stimulator of alpha-synuclein aggregation in
Arch Neurol 2003;60:1685–1691. fers resistance against p53-mediated apopto- Lewy body disease. J Biol Chem 1999; 274:
99 Meade TW: Subacute myelo-optic neuropa- sis. Proc Natl Acad Sci USA 1999; 96: 28849–28852.
thy and clioquinol: An epidemiological case- 7547–7552. 123 Ben-Shachar D, Eshel G, Finberg JP, Youdim
history for diagnosis. Br J Prev Soc Med 1975; 111 Small DH, Nurcombe V, Reed G, Clarris H, MB: The iron chelator desferrioxamine (Des-
29:157–169. Moir R, Beyreuther K, Masters CL: A hepa- feral) retards 6-hydroxydopamine-induced
100 Baum L, Ng A: Curcumin interaction with rin-binding domain in the amyloid protein degeneration of nigrostriatal dopamine neu-
copper and iron suggests one possible mecha- precursor of Alzheimer’s disease is involved rons. J Neurochem 1991;56:1441–1444.
nism of action in Alzheimer’s disease animal in the regulation of neurite outgrowth. J Neu- 124 Lan J, Jiang DH: Desferrioxamine and vita-
models. J Alzheimers Dis 2004; 6: 367–377; rosci 1994;14:2117–2127. min E protect against iron and MPTP-in-
discussion 443–369. 112 Morimoto T, Ohsawa I, Takamura C, Ishigu- duced neurodegeneration in mice. J Neural
101 Lim GP, Chu T, Yang F, Beech W, Frautschy ro M, Kohsaka S: Involvement of amyloid Transm (Budapest) 1997;104:469–481.
SA, Cole GM: The curry spice curcumin re- precursor protein in functional synapse for-
duces oxidative damage and amyloid pathol- mation in cultured hippocampal neurons. J
ogy in an Alzheimer transgenic mouse. J Neu- Neurosci Res 1998;51:185–195.
rosci 2001;21:8370–8377.

Multifunctional Catechins for Neurosignals 2005;14:46–60 59


Neuroprotection
125 Checkoway H, Powers K, Smith-Weller T, 127 Zhang ZX, Roman GC: Worldwide occur- 129 Shoham S, Youdim MB: Nutritional iron
Franklin GM, Longstreth WT Jr, Swanson rence of Parkinson’s disease: An updated re- deprivation attenuates kainate-induced neu-
PD: Parkinson’s disease risks associated with view. Neuroepidemiology 1993;12:195–208. rotoxicity in rats: implications for involve-
cigarette smoking, alcohol consumption, and 128 Kaur D, Yantiri F, Rajagopalan S, Kumar J, ment of iron in neurodegeneration. Ann NY
caffeine intake. Am J Epidemiol 2002; 155: Mo JQ, Boonplueang R, Viswanath V, Jacobs Acad Sci 2004;1012:94–114.
732–738. R, Yang L, Beal MF, DiMonte D, Volitaskis
126 Li SC, Schoenberg BS, Wang CC, Cheng XM, I, Ellerby L, Cherny RA, Bush AI, Andersen
Rui DY, Bolis CL, Schoenberg DG: A preva- JK: Genetic or pharmacological iron chela-
lence survey of Parkinson’s disease and other tion prevents MPTP-induced neurotoxicity
movement disorders in the People’s Republic in vivo: A novel therapy for Parkinson’s dis-
of China. Arch Neurol 1985;42:655–657. ease. Neuron 2003;37:899–909.

60 Neurosignals 2005;14:46–60 Mandel/Avramovich-Tirosh/Reznichenko/


Zheng/Weinreb/Amit/Youdim
Review

Neurosignals 2005;14:61–70 Received: December 6, 2004


Accepted after revision: March 1, 2005
DOI: 10.1159/000085386

Unique Properties of Polyphenol Stilbenes


in the Brain: More than Direct Antioxidant
Actions; Gene/Protein Regulatory Activity

Sylvain Doré
Johns Hopkins University, School of Medicine, Baltimore, Md., USA

Key Words protection against models of in vitro and in vivo oxida-


Bilirubin  Biliverdin  Blood flow  Carbon monoxide  tive stress injury. Resveratrol could provide cellular re-
Hemin  Iron sistance against insults; although more work is neces-
sary before it is prescribed as a potential prophylactic in
models of either acute or chronic conditions, such as
Abstract stroke, amyotrophic lateral sclerosis, Parkinson, Alz-
The ‘French Paradox’ has been typically associated with heimer, and a variety of age-related vascular disorders.
moderate consumption of wine, especially red wine. A Copyright © 2005 S. Karger AG, Basel

polyphenol 3,4’,5-trihydroxy-trans-stilbene (a member


of the non-flavonoids family), better known as resvera-
trol, has been purported to have many health benefits. The ‘French Paradox,’ so named after a population
A number of these valuable properties have been attrib- studied in France, is defined by a low incidence of cardio-
uted to its intrinsic antioxidant capabilities, although the vascular problems despite a diet that is relatively high in
potential level of resveratrol in the circulation is likely not saturated fat [1]. Because of the typical levels of wine in-
enough to neutralize free radical scavenging. The brain take of the French, it has also been postulated that a mod-
and the heart are uniquely vulnerable to hypoxic condi- erate consumption of red wine could be associated with
tions and oxidative stress injuries. Recently, evidence this paradox. A vast amount of recent literature proposes
suggests that resveratrol could act as a signaling mole- that it is the polyphenols in red wine that provide the
cule within tissues and cells to modulate the expression beneficial effect, mainly attributable to their potential to
of genes and proteins. Stimulation of such proteins and act as antioxidants. These polyphenols are divided into
enzymes could explain some the intracellular antioxida- two main categories: flavonoids and non-flavonoids. The
tive properties. The modulation of genes could suffice as flavonoids, found in natural extracts of plants and fruits,
an explanation of some of resveratrol’s cytoprotective are considered to be the most abundant polyphenols,
actions, as well as its influence on blood flow, cell death, whereas the non-flavonoid stilbenes are considered to be
and inflammatory cascades. Resveratrol stimulation of a minor class. The naturally occurring polyphenol stil-
the expression of heme oxygenase is one example. In- bene, resveratrol (3,4,5-trihydroxy-trans-stilbene), has
creased heme oxygenase activity has led to significant been proposed to possess most of the beneficial health ef-

© 2005 S. Karger AG, Basel Sylvain Doré, PhD


Johns Hopkins University, School of Medicine
Fax +41 61 306 12 34 ACCM Department, Neuro Research Division, 720 Rutland Ave
E-Mail karger@karger.ch Accessible online at: Ross Research Building 364-365, Baltimore, MD 21205 (USA)
www.karger.com www.karger.com/nsg Tel. +1 410 614 4859, Fax +1 410 955 7271, E-Mail sdore@jhmi.edu
CEREBRAL ISCHEMIA [11,12]
Reduces infarct size (rat)
Reduces focal ischemia damage (rat)
AMYOTROPHIC LATERAL SCLEROSIS [13]
Directly stimulates BK(Ca) channel activity in
vascular endothelial cells (human cells)
AGING [22,23]
PARKINSON DISEASE [14] Mimics beneficial effects of
Protects embryonic mesencephalic cells (rat) caloric restriction (yeast)

SPINAL CORD LESION [8] CHEMOPREVENTION [16,17]


Protects spinal cord from OH Inhibits critical cell cycle regulatory proteins (mice)
ischemia-reperfusion injury (rabbit) Converts into an anticancer agent by
cytochrome P 450 enzymes (human cells)
HO

PAIN [20] OH trans -resveratrol


Decreases carrageenan-
COGNITIVE IMPAIRMENT [21]
induced hyperalgesia (rat) Prevents ICV streptozotocin-induced
cognitive impairment (rat)

BRAIN EDEMA [9] BRAIN TUMORS [15]


Inhibits expression of Reduces paclitaxel-induced apoptosis in
NFkB p65 (rat) neuroblastoma SH-SY5Y cells (human cells)

SEIZURE [10,18,19]
Reduces generalized tonic-clonic convulsions (rat)
Delays FeCl 3-induced epileptiform EEG changes (rat)
Reduces kainate-induced lesions in hippocampus (rat)

Fig. 1. Experimental neurologic benefits of resveratrol.

fects of red wine, considering that resveratrol is mainly By degradation of heme, a prooxidant, into biliverdin/
found in the skin of the grapes, and the skin and seeds are bilirubin, antioxidants, modulation of HO activity and
generally not used in processing white wines. Conse- levels would seem to be cytoprotective against free radi-
quently, it has been proposed that resveratrol would po- cal damage. Using in vitro and in vivo models, we have
tentially be the most active ingredient [2–5]. Given all of also shown that HO can be neuroprotective [7]. In addi-
this, the antioxidant properties of red wine, then, must tion, HO and its metabolites have been associated with
be associated with the actions of resveratrol, and we pro- antiapoptotic and antiinflammatory actions and are
pose that the protective properties are likely due to a known to have a vasodilatory effect.
unique cascade of intracellular events leading to activa- Several other enzymatic systems have been suggested
tion of unique antioxidant pathways. This hypothesis is to be either directly or indirectly modified by different
being proposed, taking into consideration that the modest members of the stilbene family. Here, we propose that
amount of resveratrol in the blood is not likely to reach a protective properties of HO are the mechanisms that
high enough plasmatic level to neutralize free radicals. provide the brain’s resistance to a variety of neurologic
Consequently, resveratrol is likely to stimulate an intra- stresses. Resveratrol has been shown to have a unique ef-
cellular signaling pathway, leading to cytoprotection. fect on neuronal cell death and inflammatory processes,
As represented here, several genes and proteins have which are all important therapeutic targets in the devel-
been shown to be potential targets for resveratrol modu- opment of either acute and/or chronic neurodegenerative
lation. Heme oxygenase (HO) is a possible candidate. We diseases [8–10]. These vascular properties become espe-
have shown that resveratrol is a potent inducer of HO cially important when considering that reduction in cere-
protein levels, activity, and cytoprotection [6]. HO’s main bral blood flow (CBF), followed by a reperfusion phase,
function is to cleave heme (Iron-Protoporphyrin-IX), is likely to affect specific neurons and/or the cell types that
which then liberates iron, generating carbon monoxide are especially vulnerable to free radical damage. Conse-
and biliverdin, which is rapidly converted to bilirubin. quently, preventing cell death is likely to have a beneficial

62 Neurosignals 2005;14:61–70 Doré


effect on the rate of neuroinflammation and its conse-
quences. Considering all of this together, one can make a OH

valid hypothesis that polyphenol stilbenes can precondi- HO HO

tion neurons against induced stress damage. light-induced


isomerization OH OH
Figure 1 contains a list of potential neurologic benefits
OH
associated with models of neurologic diseases treated trans-resveratrol cis-resveratrol
with resveratrol. Of importance is the fact that resveratrol
has been shown to be protective in several species. Fol-
lowing publication in Science and Nature of initial reports
Fig. 2. UV-light-induced isomerization of trans- to cis-resvera-
that suggested a potential biologic role of resveratrol [24,
trol.
25], more than 900 original research articles have been
published to support the beneficial effect of resveratrol.
It is somewhat paradoxical that such a simple, natural
component, extracted from plants and fruits, can have tion of an antioxidant intracellular pathway. Considering
such a variety of actions. The flavonoids constitute the the direct antioxidant properties associated with resvera-
majority of phenols in red wine and are mainly divided trol and taking into account the absorption rate and mod-
into three classes: the flavanols, the flavonols, and the an- ifications/conjugations, one would have to consume liters
thocyanins. The known flavonoids (hydroxycinnamic ac- of these drinks in order to achieve the necessary plasmat-
ids, benzoic acids, and stilbenes) are less abundant, and ic molar ratio to neutralize the free radicals and achieve
the stilbenes are only a minor class of the known flavo- the beneficial health effects. To highlight the variability
noids. of resveratrol’s effects, figure 3 shows the intracellular
Interestingly, during the production of red wine, com- consequences of cells treated with the maximal concen-
plex sugars from the grapes are fermented into alcohol, tration of 25 M, although the list is not exhaustive. We
and it is believed that alcohol would be considered a good eliminated experiments in which the concentration was
solvent for extracting polyphenols from the skins and higher than 25 M. We believe that it is reasonable to
seeds. Considering that resveratrol is mainly found in the think that a level ^25 M could potentially be achieved
skin of the grapes, it has been reported that the amount under normal conditions and not typically under phar-
of phenols within white wine would be much less than in macologic conditions.
red wine. Although the reported amounts of resveratrol Figure 3 shows the expression regulated by resveratrol
in wine vary, they suggest that approximately 7 mg/l are of different genes and proteins and describes some of the
present in most reds, as compared to 0.5 mg/l in whites potential functions. Living organisms under aerobic con-
[26, 27]. For purposes of comparison, the flavonoid con- ditions are continuously exposed to potential damage
centrations in red wine have been in the range of 1,300– caused by reactive oxygen species (ROS). ROS are pro-
1,500 mg/l. Once again, considering all of the beneficial duced naturally during normal cellular activity and mi-
properties of resveratrol, its minimal amount available in tochondrial function. In addition, induced stress is likely
wine, if active, is likely to be mediated via activation of to modify these normal functions and simulate the gen-
intracellular pathways. eration of free radical damage. Many of the studies using
It is interesting to note, also, that resveratrol exists in polyphenols have required pretreatment with resveratrol
two isomers, the cis and the trans, as shown in figure 2. to exert these beneficial biologic functions. Such pretreat-
Both of these are found in wine, and, although it appears ment would require either an increase in the concentra-
that only the trans isomer is found in grapes, direct light tion of this polyphenol or stimulate a cascade leading to
could cause its isomerization from trans to cis. In our pre- activation of an endogenous antioxidant system. This ac-
liminary observations in neurons, we indicated that the tivation is critically important to cytoprotection in tissue
active isoform would be mainly the trans resveratrol. with a weak, endogenous, antioxidant system. The heart
The fact that resveratrol, a simple active ingredient, and the brain are two unique examples of tissues with
which has been proposed to be responsible for the ratio- weak defenses, as evidenced by infarct damage following
nale behind the French Paradox, is present in such small ischemia/reperfusion.
amounts in wine or juice, and the theory concerning its Considering the antioxidant properties associated
bioactivity by its direct antioxidant capacity deserve re- with resveratrol, we have concentrated on the neuropro-
consideration. Thus, it led us to hypothesize the activa- tective effect of resveratrol and the possibility that this

Unique Properties of Polyphenol Stilbenes Neurosignals 2005;14:61–70 63


in the Brain
Fig. 3. Example of resveratrol-regulated genes/proteins affected in cells (!25 M). HO1 = Heme oxygenase 1;
COX-2 = cyclooxygenase 2; eNOS = endothelial nitric oxide synthase; iNOS = inducible nitric oxide synthase;
ET-1 = endothelin-1; GADD45 = growth-arrest- and DNA-damage-induced protein 45; TNF = tumor necrosis
factor-alpha; ATF3 = activating transcription factor 3; IGFBP = insulin-like growth factor-binding protein.

Fig. 4. Example of potential outcomes from gene/protein regulation (i.e., HO1) and potential enzymatic role in
the brain. Note: This list of potential actions would occur at physiologic levels, while abnormal or pharmacologic
levels of these compounds could have deleterious effects.

64 Neurosignals 2005;14:61–70 Doré


Fe 3+-Ferritin OH O
3O2 + H2O + O OH
NADPH NADP + + Ferritin
Cyt P450 Red CO Fe2+
N

N Fe 2+ N N HN
HEME OXYGENASE
N
NH HN
O RESVERATROL
HO OO
O

Heme OH O
OH O
OH Biliverdin
(Fe2+ -Protoporphyrin IX) NADPH + H+
other bilirubin
BV Red metabolites

NADP +
NH HN
ROS
Bilirubin
NH HN

OO

Fig. 5. Resveratrol mediates heme degrada-


tion by induction of heme oxygenase lev-
els.

pathway could involve the induction of an antioxidant stresses, in order to defend against disruption of any sys-
system, such as heme oxygenase. Regulation of HO activ- tem homeostasis. HO1 has been suggested to have differ-
ity has been shown to be protective in acute and/or chron- ent functions in the brain. It is one of the heat shock pro-
ic neurodegenerative conditions. Figure 4 summarizes teins, which are stress proteins that are induced in differ-
some of the potential outcomes of regulation of the HO ent cells and following different stimuli [37], including
protein and its activity. hypothermia [38], global ischemia [41], subarachnoid
As mentioned above, HO catalyzes the degradation of hemorrhage [42], Parkinson disease [47], Alzheimer dis-
heme, which is mainly a prooxidant, into iron, biliverdin/ ease [39, 40], and several other acute and chronic neuro-
bilirubin, and carbon monoxide (fig. 5). Two isoforms of logic conditions [43–46].
HO have been isolated and characterized [7, 33–35]. A In a model of transient ischemia, we and others have
third isoform has been reported, although it does not shown that HO mRNA and proteins are induced [43].
seem to be translated into a protein [36]. HO1, the first Reports have indicated that induction of HO proteins in
to be isolated [34], is the inducible enzyme and appears neurons would be protective [49], and our preliminary
to be concentrated mainly in the liver and spleen, an un- data indicate that resveratrol could induce HO levels
derstandable observation considering the high turnover within neurons, potentially affording neuroprotection.
of hemoglobin, which has heme in its core. Under basal Consequently, we believe that modulation of HO levels
conditions, HO1 is barely detectable in the brain, al- and its activity could be a pathway by which resveratrol
though several reports suggest that, under a variety of present in red wine or other concentrated extracts could
stimuli, it can be induced within brain tissue [37–48]. potentially protect the nervous system against induced
HO2 is an isoform that is constitutively expressed and oxidative stress damage. Many heme-containing enzymes
appears to be concentrated mainly in the brain and testis are located in the mitochondria, the cytosol, and the en-
[48]. Its protein expression level appears to be extremely doplasmic reticulum; and they presumably undergo rap-
stable, which is likely to respond to normal cellular ho- id turnover during oxidative stress. HO is the main en-
meostasis. zyme, which can degrade free heme, a prooxidant, and
Regulation of the HO1 protein and its activity has elic- maintain levels that would not reach toxicity. Following
ited a great deal of interest. Regulation of HO1 has been an ischemic event, tissue injury has been proposed to be
suggested to be in response to many cellular and organ mainly due to increased oxidative stress by free radicals

Unique Properties of Polyphenol Stilbenes Neurosignals 2005;14:61–70 65


in the Brain
generated during the reperfusion phase. We have previ- iron outside the cell [52]. Homeostasis of iron is a key
ously shown that the infarct volume in HO2 knockout factor in controlling cell toxicity. As one example, free
(HO2–/–) mice versus wild-type (WT) mice after stroke is iron has been considered to be a key ingredient in the
approximately double. HO2 is the isoform present under Fenton reaction, in which by reacting with H2O2, it gen-
normal basal conditions. Consequently, increasing the erates free radicals. Regulation of cellular homeostasis of
activity of heme oxygenase is likely to be protective in iron is a complex and tightly regulated system. It is regu-
stroke. lated by many proteins, a number of which are still under
Interestingly, the group led by Maines has demonstrat- extensive characterization. Rapid upregulation of HO1
ed significant reduction in infarct size after stroke in a by resveratrol in neurons could potentially directly affect
transgenic mouse model on which HO1 was over-ex- the intracellular iron level.
pressed using a neuron-specific promoter [50]. In addi- It has been previously demonstrated that decrease in
tion, we have recently accumulated evidence suggesting HO1 activity would be sufficient to change its iron level
that pre-treatment with resveratrol would be a most po- within the cell. For example, by using HO–/– mice, it has
tent inducer of HO1 in mouse primary neuronal cultures, been shown that iron accumulates in several organs [53].
that it would be sufficient to prevent induced neurotoxic- In addition, numerous iron-binding proteins are regulat-
ity, and that this neuroprotection was significantly atten- ed by intracellular levels of free iron [54]. As an example,
uated by the use of a heme oxygenase inhibitor [51]. All ferritin in the cell can sequester numerous molecules of
together, these results suggest that an increase in HO1 iron, and its intracellular level is tightly regulated to free
protein levels and its activities within neurons is likely to iron.
provide neuroprotection and promote cell survival. Therapeutic implications of controlling iron levels
within tissues or within cells are numerous. As an exam-
Heme ple, administration of desferoxamine, a trivalent iron
Heme metabolism is a crucial metabolic process. It has chelator, over a 2-year period slows the clinical progres-
been postulated that free heme can rapidly be generated sion of symptoms associated with Alzheimer disease [55].
from an induced turnover of heme-containing proteins/ Further study may bear out that the regulation of HO1
enzymes, for example, catalase, glutathione peroxidase, levels also regulates the cellular iron homeostasis. Inter-
superoxide dismutase, cytochrome, guanylate cyclase, ni- estingly, very little has been investigated regarding the
tric oxide synthase, etc. It has been further suggested that potential role of resveratrol in regulating iron and deter-
during hypoxia, ischemic injury could trigger significant mining its potential effect in neurologic disorders.
amounts of heme being released into the intracellular
pool. When stimulation of these hemoproteins is degrad- Carbon Monoxide
ed, heme becomes free; probably not salvaged, it should Carbon monoxide (CO) is a gas that is almost exclu-
be rapidly degraded. HO is the enzyme that can rapidly sively generated in cells by degradation of heme by heme
cleave prooxidant through heme and limit its capacity to oxygenase [56, 57]. In that it is a gas, carbon monoxide
enter into generation of a free radical cycle – notably, can travel freely through intracellular and extracellular
through its iron molecule. Consequently, HO could be compartments. The CO literature is vast and complex
considered an antioxidant enzyme by degradation of the with many controversies that have yet to be resolved [58–
prooxidant heme. Our previous observations indicate 63]. CO is better known to be toxic at high levels. It can
that resveratrol could specifically induce HO1 within also cause death [64]. Interestingly, in the recent litera-
neurons and potentially protect cells against oxidative ture, low concentrations of CO have been suggested to be
stress injury. Such a pathway is an interesting target for protective. Although CO has an affinity slightly lower to
resveratrol, considering that it can regulate the redox state its homologue, nitric oxide (NO), which is another gas, it
of the cell and prevent cells from dying through an oxida- appears that its half-life would be significantly longer.
tive stress-induced cascade. Such an observation could allow carbon monoxide, by
binding to heme moiety present on several key enzymes,
Iron to modulate their function [65]. Within the cell, physio-
Degradation of heme by HO1 also generates a mole- logic/normal levels of CO generated from degradation of
cule of iron. Evidence suggests that regulation of HO1 intracellular heme are likely to have multiple biologic
protein levels could modulate the level of intracellular functions on several heme-containing proteins. For ex-
heme. It is postulated that HO can stimulate the efflux of ample, CO has been shown to act as a vasodilator by po-

66 Neurosignals 2005;14:61–70 Doré


tentially binding with soluble guanylate cyclase (sGC)
and modulating its vasoactive activities. It can act on cal-
Antioxidant properties
cium-activated potassium channels (KCa channels) and Most neurologic disorders [6]
regulate their opening [66, 67]. CO has also been recent-
Inflammation reduction
ly reported to have specific antiapoptotic and antiinflam- Most neurologic disorders [77,78]
matory actions [68, 69]. Rapid induction of HO1 could
Restoration of normal blood flow
be a means by which resveratrol can increase CO levels Cerebral Ischemia [79]
within physiologic concentrations and allow cells and tis- AD (age-related vascular dementia) [7]

sue to benefit from many of CO’s biologic actions, espe- Reduction of neuronal cell death
cially in scenarios in which blood flow is reduced and cell In stroke models [43]
In induced apoptosis [80]
survival is compromised. Cholinergic neuron in AD models [81]
Dopaminergic neurons in PD models [47]
Bilirubin Regulation of iron homeostasis
Bilirubin is known for its toxicity, at high micromolar Parkinsonism [82]
Ataxia [83]
concentrations, in the central nervous system (CNS), es- Movement disorders, tremors [84]
pecially in neonates. Although under physiologic concen- Restless Leg Syndrome [85]
trations, bilirubin can act as an endogenous antioxidant Hallervorden-Spatz Syndrome [86]
Aceruloplasminemia [87]
[70, 71]. In testing a series of antioxidants, bilirubin was Neuroferritinopathy [88]
shown to have significant superoxide and hydroxyl radi-
Others
cal scavenger activity [72]. Using an animal model of hy- Sleep pattern [89]
perbilirubinemia, a protective effect against cerebral isch- Circadian rhythm [90,91]
emia was demonstrated. We have also observed, using Amyotrophic lateral sclerosis [92]
primary hippocampal and cortical neuronal cultures, that Spinal cord lesion [93]
Head trauma [94]
bilirubin can be protective at low concentrations [73, 74].
Brain edema [50]
Moreover, in previous observations on the potential role
Huntington [95]
of HO in Alzheimer disease pathology [39, 75], levels of Kernicterus [94]
bilirubin derivatives in the cerebral spinal fluid were re- Brain tumors [97]
ported to be significantly increased in brains of AD pa- Chemoprevention [98]

tients as compared to control [76]. An increase in HO1 Pain [99]


Seizure [100]
within AD brains was reported. Increased HO1 levels
Atherosclerosis [101]
could potentially increase the bilirubin level within a Age-related cognitive impairment [102]
physiologic range, and such a pathway could explain
some of the antioxidant properties associated with resve-
ratrol in relation to neurologic deficits in age-related de-
Fig. 6. Non-exhaustive list of potential neurologic benefits of res-
mentia. veratrol-regulating HO1.
Figure 6 briefly summarizes some of the neurologic
disorders that can be beneficially affected by the regula-
tion of HO activity. Regulation of genes by resveratrol,
such as in the case of HO1, presents a potential mecha- cally consume moderate amounts of red wine support the
nism by which its prophylactic use might be considered, prospect of a prophylactic role for resveratrol. Conse-
under either acute or chronic neurologic disorders. For quently, a better understanding of the mechanisms by
example, under ischemic stroke conditions, reperfusion which polyphenols and red wine can protect against isch-
frequently occurs after focal ischemia, particularly in the emic and toxic insults would be of great interest.
case of cerebral embolism and transient ischemic attack.
These can be warning signs of impending stroke. Recur-
rence is a prevalent phenomenon in patients who have Acknowledgements
suffered one episode of stroke. Therefore, availability of
This work is supported in part by the NIH grants (AA014911
a prophylactic approach in these patients is an important and AT002113), the Wine Institute, and the ABMR Foundation.
goal of preventive medicine. Reports that coronary heart The author would like to thank Tzipora Sofare, MA, for her assis-
disease appears to be reduced in patients who chroni- tance in preparing the manuscript.

Unique Properties of Polyphenol Stilbenes Neurosignals 2005;14:61–70 67


in the Brain
References

1 Renaud S, de Lorgeril M: Wine, alcohol, plate- 16 Reagan-Shaw S, Afaq F, Aziz MH, Ahmad N: 28 Subbaramaiah K, Chung WJ, Michaluart P,
lets, and the French paradox for coronary heart Modulations of critical cell cycle regulatory Telang N, Tanabe T, Inoue H, Jang M, Pez-
disease. Lancet 1992;339:1523–1526. events during chemoprevention of ultraviolet zuto JM, Dannenberg AJ: Resveratrol inhibits
2 Sato M, Ray PS, Maulik G, Maulik N, Engel- B-mediated responses by resveratrol in SKH-1 cyclooxygenase-2 transcription and activity in
man RM, Bertelli AA, Bertelli A, Das DK: hairless mouse skin. Oncogene 2004;23:5151– phorbol ester-treated human mammary epi-
Myocardial protection with red wine extract. J 5160. thelial cells. J Biol Chem 1998; 273: 21875–
Cardiovasc Pharmacol 2000;35:263–268. 17 Potter GA, Patterson LH, Wanogho E, Perry 21882.
3 Soleas GJ, Diamandis EP, Goldberg DM: Res- PJ, Butler PC, Ijaz T, Ruparelia KC, Lamb JH, 29 Cheung CY, Chen J, Chang TK: Evaluation of
veratrol: A molecule whose time has come? Farmer PB, Stanley LA, Burke MD: The can- a real-time polymerase chain reaction method
And gone? Clin Biochem 1997;30:91–113. cer preventative agent resveratrol is converted for the quantification of CYP1B1 gene expres-
4 Goldberg D, Tsang E, Karumanchiri A, Dia- to the anticancer agent piceatannol by the cy- sion in MCF-7 human breast carcinoma cells.
mandis E, Soleas G, Ng E: Method to assay the tochrome P450 enzyme CYP1B1. Br J Cancer J Pharmacol Toxicol Methods 2004; 49: 97–
concentrations of phenolic constituents of bio- 2002;86:774–778. 104.
logical interest in wines. Anal Chem 1996;68: 18 Gupta YK, Chaudhary G, Srivastava AK: Pro- 30 Imamura G, Bertelli AA, Bertelli A, Otani H,
1688–1694. tective effect of resveratrol against pentylene- Maulik N, Das DK: Pharmacological precon-
5 Celotti E, Ferrarini R, Zironi R, Conte LS: Res- tetrazole-induced seizures and its modulation ditioning with resveratrol: An insight with
veratrol content of some wines obtained from by an adenosinergic system. Pharmacology iNOS knockout mice. Am J Physiol Heart Circ
dried Valpolicella grapes: Recioto and Ama- 2002;65:170–174. Physiol 2002;282:H1996–H2003.
rone. J Chromatogr [A] 1996;730:47–52. 19 Gupta YK, Chaudhary G, Sinha K, Srivastava 31 Liu JC, Chen JJ, Chan P, Cheng CF, Cheng
6 Zhuang H, Kim YS, Koehler RC, Doré S: Po- AK: Protective effect of resveratrol against in- TH: Inhibition of cyclic strain-induced endo-
tential mechanism by which resveratrol, a red tracortical FeCl3-induced model of posttrau- thelin-1 gene expression by resveratrol. Hyper-
wine constituent, protects neurons. Ann NY matic seizures in rats. Methods Find Exp Clin tension 2003;42:1198–1205.
Acad Sci 2003;993:276–286. Pharmacol 2001;23:241–244. 32 Shi T, Liou LS, Sadhukhan P, Duan ZH,
7 Doré S: Decreased activity of the antioxidant 20 Gentilli M, Mazoit JX, Bouaziz H, Fletcher D, Novick AC, Hissong JG, Almasan A, DiDo-
heme oxygenase enzyme: Implications in isch- Casper RF, Benhamou D, Savouret JF: Resve- nato JA: Effects of resveratrol on gene expres-
emia and in Alzheimer’s disease. Free Radic ratrol decreases hyperalgesia induced by car- sion in renal cell carcinoma. Cancer Biol Ther
Biol Med 2002;32:1276–1282. rageenan in the rat hind paw. Life Sci 2001;68: 2004;3:1538–4047.
8 Kiziltepe U, Turan NN, Han U, Ulus AT, Akar 1317–1321. 33 Ewing JF, Maines MD: Histochemical local-
F: Resveratrol, a red wine polyphenol, protects 21 Sharma M, Gupta YK: Chronic treatment with ization of heme oxygenase-2 protein and
spinal cord from ischemia-reperfusion injury. trans resveratrol prevents intracerebroventric- mRNA expression in rat brain. Brain Res
J Vasc Surg 2004;40:138–145. ular streptozotocin induced cognitive impair- Brain Res Protoc 1997;1:165–174.
9 Wang YJ, He F, Li XL: The neuroprotection ment and oxidative stress in rats. Life Sci 2002; 34 Shibahara S, Muller R, Taguchi H, Yoshida T:
of resveratrol in the experimental cerebral isch- 71:2489–2498. Cloning and expression of cDNA for rat heme
emia. Zhonghua Yi Xue Za Zhi 2003;83:534– 22 Howitz KT, Bitterman KJ, Cohen HY, Lam- oxygenase. Proc Natl Acad Sci USA 1985; 82:
536. ming DW, Lavu S, Wood JG, Zipkin RE, 7865–7869.
10 Virgili M, Contestabile A: Partial neuroprotec- Chung P, Kisielewski A, Zhang LL, Scherer B, 35 Maines MD: The heme oxygenase system: A
tion of in vivo excitotoxic brain damage by Sinclair DA: Small molecule activators of sir- regulator of second messenger gases. Annu Rev
chronic administration of the red wine anti- tuins extend Saccharomyces cerevisiae life- Pharmacol Toxicol 1997;37:517–554.
oxidant agent, trans-resveratrol in rats. Neuro- span. Nature 2003;425:191–196. 36 Zhuang H, Pin S, Li X, Doré S: Regulation of
sci Lett 2000;281:123–126. 23 Wood JG, Rogina B, Lavu S, Howitz K, Hel- heme oxygenase expression by cyclopentenone
11 Huang SS, Tsai MC, Chih CL, Hung LM, Tsai fand SL, Tatar M, Sinclair D: Sirtuin activators prostaglandins. Exp Biol Med 2003; 228: 499–
SK: Resveratrol reduction of infarct size in mimic caloric restriction and delay ageing in 505.
Long-Evans rats subjected to focal cerebral metazoans. Nature 2004;430:686–689. 37 Ewing JF, Maines MD: Rapid induction of
ischemia. Life Sci 2001;69:1057–1065. 24 Jang M, Cai L, Udeani GO, Slowing KV, heme oxygenase 1 mRNA and protein by hy-
12 Sinha K, Chaudhary G, Gupta YK: Protective Thomas CF, Beecher CW, Fong HH, Farn- perthermia in rat brain: Heme oxygenase 2 is
effect of resveratrol against oxidative stress in sworth NR, Kinghorn AD, Mehta RG, Moon not a heat shock protein. Proc Natl Acad Sci
middle cerebral artery occlusion model of RC, Pezzuto JM: Cancer chemopreventive ac- USA 1991;88:5364–5368.
stroke in rats. Life Sci 2002;71:655–665. tivity of resveratrol, a natural product derived 38 Ewing JF, Haber SN, Maines MD: Normal and
13 Wu SN: Large-conductance Ca2+-activated K+ from grapes. Science 1997;275:218–220. heat-induced patterns of expression of heme
channels: Physiological role and pharmacolo- 25 Corder R, Douthwaite JA, Lees DM, Khan oxygenase-1 (HSP32) in rat brain: Hyperther-
gy. Curr Med Chem 2003;10:649–661. NQ, Viseu Dos Santos AC, Wood EG, Carrier mia causes rapid induction of mRNA and pro-
14 Karlsson J, Emgard M, Brundin P, Burkitt MJ: MJ: Endothelin-1 synthesis reduced by red tein. J Neurochem 1992;58:1140–1149.
Trans-resveratrol protects embryonic mesen- wine. Nature 2001;414:863–864. 39 Schipper HM, Cisse S, Stopa EG: Expression
cephalic cells from tert-butyl hydroperoxide: 26 Ribeiro de Lima MT, Waffo-Teguo P, Teisse- of heme oxygenase-1 in the senescent and Alz-
Electron paramagnetic resonance spin trap- dre PL, Pujolas A, Vercauteren J, Cabanis JC, heimer-diseased brain. Ann Neurol 1995; 37:
ping evidence for a radical scavenging mecha- Merillon JM: Determination of stilbenes 758–768.
nism. J Neurochem 2000;75:141–150. (trans-astringin, cis- and trans-piceid, and cis- 40 Smith MA, Kutty RK, Richey PL, Yan SD,
15 Nicolini G, Rigolio R, Scuteri A, Miloso M, and trans-resveratrol) in Portuguese wines. J Stern D, Chader GJ, Wiggert B, Petersen RB,
Saccomanno D, Cavaletti G, Tredici G: Effect Agric Food Chem 1999;47:2666–2670. Perry G: Heme oxygenase-1 is associated with
of trans-resveratrol on signal transduction 27 Wallerath T, Deckert G, Ternes T, Anderson the neurofibrillary pathology of Alzheimer’s
pathways involved in paclitaxel-induced apop- H, Li H, Witte K, Forstermann U: Resveratrol, disease. Am J Pathol 1994;145:42–47.
tosis in human neuroblastoma SH-SY5Y cells. a polyphenolic phytoalexin present in red
Neurochem Int 2003;42:419–429. wine, enhances expression and activity of en-
dothelial nitric oxide synthase. Circulation
2002;106:1652–1658.

68 Neurosignals 2005;14:61–70 Doré


41 Takeda A, Kimpara T, Onodera H, Itoyama Y, 54 Quinlan GJ, Chen Y, Evans TW, Gutteridge 69 Brouard S, Berberat PO, Tobiasch E, Seldon
Shibahara S, Kogure K: Regional difference in JM: Iron signalling regulated directly and MP, Bach FH, Soares MP: Heme oxygenase-1-
induction of heme oxygenase-1 protein follow- through oxygen: Implications for sepsis and the derived carbon monoxide requires the activa-
ing rat transient forebrain ischemia. Neurosci acute respiratory distress syndrome. Clin Sci tion of transcription factor NF-kappa B to pro-
Lett 1996;205:169–172. (Lond) 2001;100:169–182. tect endothelial cells from tumor necrosis
42 Kuroki M, Kanamaru K, Suzuki H, Waga S, 55 Crapper McLachlan DR, Dalton AJ, Kruck factor-alpha-mediated apoptosis. J Biol Chem
Semba R: Effect of vasospasm on heme oxy- TP, Bell MY, Smith WL, Kalow W, Andrews 2002;277:17950–17961.
genases in a rat model of subarachnoid hemor- DF: Intramuscular desferrioxamine in patients 70 Gopinathan V, Miller NJ, Milner AD, Rice-
rhage. Stroke 1998; 29: 683–688; discussion with Alzheimer’s disease. Lancet 1991; 337: Evans CA: Bilirubin and ascorbate antioxidant
688–689. 1304–1308. activity in neonatal plasma. FEBS Lett 1994;
43 Doré S, Sampei K, Goto S, Alkayed NJ, Guas- 56 Vreman HJ, Wong RJ, Sanesi CA, Dennery 349:197–200.
tella D, Blackshaw S, Gallagher M, Traystman PA, Stevenson DK: Simultaneous production 71 Stocker R, Glazer AN, Ames BN: Antioxidant
RJ, Hurn PD, Koehler RC, Snyder SH: Heme of carbon monoxide and thiobarbituric acid activity of albumin-bound bilirubin. Proc Natl
oxygenase-2 is neuroprotective in cerebral reactive substances in rat tissue preparations Acad Sci USA 1987;84:5918–5922.
ischemia. Mol Med 1999;5:656–663. by an iron-ascorbate system. Can J Physiol 72 Farrera JA, Jauma A, Ribo JM, Peire MA,
44 Koistinaho J, Miettinen S, Keinanen R, Var- Pharmacol 1998;76:1057–1065. Parellada PP, Roques-Choua S, Bienvenue E,
tiainen N, Roivainen R, Laitinen JT: Long- 57 Yoshida T, Noguchi M, Kikuchi G: The step Seta P: The antioxidant role of bile pigments
term induction of haem oxygenase-1 (HSP-32) of carbon monoxide liberation in the sequence evaluated by chemical tests. Bioorg Med Chem
in astrocytes and microglia following transient of heme degradation catalyzed by the reconsti- 1994;2:181–185.
focal brain ischaemia in the rat. Eur J Neurosci tuted microsomal heme oxygenase system. J 73 Doré S, Takahashi M, Ferris CD, Zakhary R,
1996;8:2265–2272. Biol Chem 1982;257:9345–9348. Hester LD, Guastella D, Snyder SH: Bilirubin,
45 Nimura T, Weinstein PR, Massa SM, Panter S, 58 Alkadhi KA, Al-Hijailan RS, Malik K, Hogan formed by activation of heme oxygenase-2,
Sharp FR: Heme oxygenase-1 (HO-1) protein YH: Retrograde carbon monoxide is required protects neurons against oxidative stress inju-
induction in rat brain following focal ischemia. for induction of long-term potentiation in rat ry. Proc Natl Acad Sci USA 1999; 96: 2445–
Brain Res Mol Brain Res 1996;37:201–208. superior cervical ganglion. J Neurosci 2001;21: 2450.
46 Matz P, Weinstein P, States B, Honkaniemi J, 3515–3520. 74 Doré S, Snyder SH: Neuroprotective action of
Sharp FR: Subarachnoid injections of lysed 59 Verma A, Hirsch DJ, Glatt CE, Ronnett GV, bilirubin against oxidative stress in primary
blood induce the hsp70 stress gene and pro- Snyder SH: Carbon monoxide: A putative neu- hippocampal cultures. Ann NY Acad Sci 1999;
duce DNA fragmentation in focal areas of the ral messenger. Science 1993;259:381–384. 890:167–172.
rat brain. Stroke 1996;27:504–512; discussion 60 Maines M: Carbon monoxide and nitric oxide 75 Takahashi M, Doré S, Ferris CD, Tomita T,
513. homology: Differential modulation of heme Sawa A, Wolosker H, Borchelt DR, Iwatsubo
47 Yoo MS, Chun HS, Son JJ, DeGiorgio LA, oxygenases in brain and detection of protein T, Kim SH, Thinakaran G, Sisodia SS, Snyder
Kim DJ, Peng C, Son JH: Oxidative stress reg- and activity. Methods Enzymol 1996; 268: SH: Amyloid precursor proteins inhibit heme
ulated genes in nigral dopaminergic neuronal 473–488. oxygenase activity and augment neurotoxicity
cells: correlation with the known pathology in 61 Meffert MK, Haley JE, Schuman EM, Schul- in Alzheimer’s disease. Neuron 2000;28:461–
Parkinson’s disease. Brain Res Mol Brain Res man H, Madison DV: Inhibition of hippocam- 473.
2003;110:76–84. pal heme oxygenase, nitric oxide synthase, and 76 Kimpara T, Takeda A, Yamaguchi T, Arai H,
48 Takizawa S, Hirabayashi H, Matsushima K, long-term potentiation by metalloporphyrins. Okita N, Takase S, Sasaki H, Itoyama Y: In-
Tokuoka K, Shinohara Y: Induction of heme Neuron 1994;13:1225–1233. creased bilirubins and their derivatives in ce-
oxygenase protein protects neurons in cortex 62 Poss KD, Thomas MJ, Ebralidze AK, O’Dell rebrospinal fluid in Alzheimer’s disease. Neu-
and striatum, but not in hippocampus, against TJ, Tonegawa S: Hippocampal long-term po- robiol Aging 2000;21:551–554.
transient forebrain ischemia. J Cereb Blood tentiation is normal in heme oxygenase-2 mu- 77 Zhuang H, Kim YS, Namiranian K, Doré S:
Flow Metab 1998;18:559–569. tant mice. Neuron 1995;15:867–873. Prostaglandins of J series control heme oxygen-
49 Doré S, Law A, Blackshaw S, Gauthier S, 63 Linden DJ, Narasimhan K, Gurfel D: Proto- ase expression: Potential significance in modu-
Quirion R: Alteration of expression levels of porphyrins modulate voltage-gated Ca current lating neuroinflammation. Ann N Y Acad Sci
neuronal nitric oxide synthase and haem oxy- in AtT-20 pituitary cells. J Neurophysiol 1993; 2003;993:208–216.
genase-2 messenger RNA in the hippocampi 70:2673–2677. 78 Bishop A, Cashman NR: Induced adaptive re-
and cortices of young adult and aged cogni- 64 Cowan RL, Doré S: Toxicity and neuroprotec- sistance to oxidative stress in the CNS: A dis-
tively unimpaired and impaired Long-Evans tive effects of carbon monoxide: Consequences cussion on possible mechanisms and their ther-
rats. Neuroscience 2000;100:769–775. to suicide and survival. Encycl Psychol Behav apeutic potential. Curr Drug Metab 2003; 4:
50 Panahian N, Yoshiura M, Maines MD: Over- Sci 2004;3:994–997. 171–184.
expression of heme oxygenase-1 is neuropro- 65 Hartsfield CL: Cross talk between carbon mon- 79 Goto S, Sampei K, Alkayed NJ, Doré S,
tective in a model of permanent middle cere- oxide and nitric oxide. Antioxid Redox Signal Koehler RC: Characterization of a new double-
bral artery occlusion in transgenic mice. J 2002;4:301–307. filament model of focal cerebral ischemia in
Neurochem 1999;72:1187–1203. 66 Wang R, Wu L: The chemical modification of heme oxygenase-2-deficient mice. Am J Physi-
51 Zhuang H, Kim YS, Greenberg DL, Doré S: KCa channels by carbon monoxide in vascular ol Regul Integr Comp Physiol 2003;285:R222–
Potential mechanism of stilbene neuroprotec- smooth muscle cells. J Biol Chem 1997; 272: R230.
tion. Soc Neurosc Abstr 2004;216:12. 8222–8226. 80 Doré S, Goto S, Sampei K, Blackshaw S, Hes-
52 Ferris CD, Jaffrey SR, Sawa A, Takahashi M, 67 Wu L, Cao K, Lu Y, Wang R: Different mech- ter LD, Ingi T, Sawa A, Traystman RJ, Koehler
Brady SD, Barrow RK, Tysoe SA, Wolosker H, anisms underlying the stimulation of K(Ca) RC, Snyder SH: Heme oxygenase-2 acts to pre-
Baranano DE, Doré S, Poss KD, Snyder SH: channels by nitric oxide and carbon monoxide. vent neuronal cell death in brain cultures and
Haem oxygenase-1 prevents cell death by regu- J Clin Invest 2002;110:691–700. following transient cerebral ischemia. Neuro-
lating cellular iron. Nat Cell Biol 1999;1:152– 68 Otterbein LE: Carbon monoxide: Innovative science 2000;99:587–592.
157. anti-inflammatory properties of an age-old gas 81 Calabrese V, Butterfield DA, Stella AM: Nutri-
53 Poss KD, Tonegawa S: Heme oxygenase 1 is molecule. Antioxid Redox Signal 2002;4:309– tional antioxidants and the heme oxygenase
required for mammalian iron reutilization. 319. pathway of stress tolerance: Novel targets for
Proc Natl Acad Sci USA 1997; 94: 10919– neuroprotection in Alzheimer’s disease. Ital J
10924. Biochem 2003;52:177–181.

Unique Properties of Polyphenol Stilbenes Neurosignals 2005;14:61–70 69


in the Brain
82 Schipper HM, Cisse S, Stopa EG: Expression 89 Reid G: Association of sudden infant death 97 Hara E, Takahashi K, Tominaga T, Kumabe
of heme oxygenase-1 in the senescent and Alz- syndrome with grossly deranged iron metabo- T, Kayama T, Suzuki H, Fujita H, Yoshimo-
heimer-diseased brain. Ann Neurol 1995; 37: lism and nitric oxide overload. Med Hypoth- to T, Shirato K, Shibahara S: Expression of
758–768. eses 2000;54:137–139. heme oxygenase and inducible nitric oxide
83 Wagner KR, Sharp FR, Ardizzone TD, Lu A, 90 Artinian LR, Ding JM, Gillette MU: Carbon synthase mRNA in human brain tumors. Bio-
Clark JF: Heme and iron metabolism: Role in monoxide and nitric oxide: Interacting mes- chem Biophys Res Commun 1996;224:153–
cerebral hemorrhage. J Cereb Blood Flow sengers in muscarinic signaling to the brain’s 158.
Metab 2003;23:629–652. circadian clock. Exp Neurol 2001; 171: 293– 98 Solowiej E, Kasprzycka-Guttman T, Fiedor
84 Thompson K, Menzies S, Muckenthaler M, 300. P, Rowinski W: Chemoprevention of cancer-
Torti FM, Wood T, Torti SV, Hentze MW, 91 Dioum EM, Rutter J, Tuckerman JR, Gonza- ogenesis – the role of sulforaphane. Acta Pol
Beard J, Connor J: Mouse brains deficient in lez G, Gilles-Gonzalez MA, McKnight SL: Pharm 2003;60:97–100.
H-ferritin have normal iron concentration but NPAS2: a gas-responsive transcription factor. 99 Liang DY, Li X, Clark JD: Formalin-induced
a protein profile of iron deficiency and in- Science 2002;298:2385–2387. spinal cord calcium/calmodulin-dependent
creased evidence of oxidative stress. J Neurosci 92 Ilzecka J, Stelmasiak Z: Serum bilirubin con- protein kinase II alpha expression is modu-
Res 2003;71:46–63. centration in patients with amyotrophic lateral lated by heme oxygenase in mice. Neurosci
85 Krieger J, Schroeder C: Iron, brain and restless sclerosis. Clin Neurol Neurosurg 2003; 105: Lett 2004;360:61–64.
legs syndrome. Sleep Med Rev 2001; 5: 277– 237–240. 100 Carratu P, Pourcyrous M, Fedinec A, Leffler
286. 93 Gordh T, Sharma HS, Azizi M, Alm P, West- CW, Parfenova H: Endogenous heme oxy-
86 Ponka P: Hereditary causes of disturbed iron man J: Spinal nerve lesion induces upregula- genase prevents impairment of cerebral vas-
homeostasis in the central nervous system. tion of constitutive isoform of heme oxygenase cular functions caused by seizures. Am J
Ann NY Acad Sci 2004;1012:267–281. in the spinal cord. An immunohistochemical Physiol 2003;285:H1148–H1157.
87 Xu X, Pin S, Gathinji M, Fuchs R, Harris ZL: investigation in the rat. Amino Acids 2000;19: 101 Zhuang H, Littleton-Kearney MT, Doré S:
Aceruloplasminemia: An inherited neurode- 373–381. Characterization of heme oxygenase in adult
generative disease with impairment of iron ho- 94 Beschorner R, Adjodah D, Schwab JM, Mit- rodent platelets. Curr Neurovasc Res 2004;in
meostasis. Ann NY Acad Sci 2004;1012:299– telbronn M, Pedal I, Mattern R, Schluesener press.
305. HJ, Meyermann R: Long-term expression of 102 Law A, Doré S, Blackshaw S, Gauthier S,
88 Crompton DE, Chinnery PF, Fey C, Curtis heme oxygenase-1 (HO-1, HSP-32) following Quirion R: Alteration of expression levels of
AR, Morris CM, Kierstan J, Burt A, Young F, focal cerebral infarctions and traumatic brain neuronal nitric oxide synthase and haem oxy-
Coulthard A, Curtis A, Ince PG, Bates D, Jack- injury in humans. Acta Neuropathol (Berl) genase-2 messenger RNA in the hippocampi
son MJ, Burn J: Neuroferritinopathy: A win- 2000;100:377–384. and cortices of young adult and aged cogni-
dow on the role of iron in neurodegeneration. 95 Browne SE, Ferrante RJ, Beal MF: Oxidative tively unimpaired and impaired Long-Evans
Blood Cells Mol Dis 2002;29:522–531. stress in Huntington’s disease. Brain Pathol rats. Neuroscience 2000;100:769–775.
1999;9:147–163.
96 Cooke RW: New approach to prevention of
kernicterus. Lancet 1999;353:1814–1815.

70 Neurosignals 2005;14:61–70 Doré


Review

Received: September 23, 2004


Neurosignals 2005;14:71–82
Accepted after revision: November 8, 2004
DOI: 10.1159/000085387

Neuroprotective Effects of Huperzine A


A Natural Cholinesterase Inhibitor for the Treatment of Alzheimer’s Disease

Rui Wang Xi Can Tang


State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Shanghai Institutes for
Biological Sciences, Chinese Academy of Sciences, Zhangjiang Hi-Tech Park, Shanghai, China

Key Words chondria, and interfere with APP metabolism. Antagoniz-


Huperzine A W Alzheimer’s disease W ing effects on NMDA receptors and potassium currents
Acetylcholinesterase W Cholinesterase inhibitor W may contribute to the neuroprotection as well. It is also
Cognitive enhancer W Neuroprotection W Oxidative stress W possible that the non-catalytic function of AChE is in-
Beta-amyloid W Amyloid precursor protein W Cerebral volved in neuroprotective effects of HupA. The thera-
ischemia W Apoptosis W Apoptotic-related gene W peutic effects of HupA on AD or VD are probably exerted
Mitochondria W Glutamate W NMDA receptor W Potassium via a multi-target mechanism.
current Copyright © 2005 S. Karger AG, Basel

Abstract Alzheimer’s disease (AD) is a progressive, neurodegen-


Huperzine A (HupA), isolated from Chinese herb Huper- erative disorder associated with a global impairment of
zia serrata, is a potent, highly specific and reversible higher mental function, and presenting an impairment of
inhibitor of acetylcholinesterase. It has been found to memory as the cardinal symptom [1]. Histopathological
reverse or attenuate cognitive deficits in a broad range of hallmarks of the disease are the extracellular deposition of
animal models. Clinical trials in China have demon- amyloid ß-peptide (Aß) in senile plaques, the appearance
strated that HupA significantly relieves memory deficits of intracellular neurofibrillary tangles (NFT), a loss of
in aged subjects, patients with benign senescent forget- cholinergic neurons, and extensive synaptic changes in
fulness, Alzheimer’s disease (AD) and vascular dementia the cerebral cortex, hippocampus and other areas of brain
(VD), with minimal peripheral cholinergic side effects essential for cognitive functions.
compared with other AChEIs in use. HupA possesses the To date, the cause and the mechanism by which neu-
ability to protect cells against hydrogen peroxide, ß-amy- rons die in AD remain unclear, but Aß has been estab-
loid protein (or peptide), glutamate, ischemia and stau- lished as a crucial factor in AD pathogenesis. Aß deposi-
rosporine-induced cytotoxicity and apoptosis. These tion may cause neuronal death via a number of possible
protective effects are related to its ability to attenuate mechanisms, including oxidative stress, excitotoxicity,
oxidative stress, regulate the expression of apoptotic energy depletion, inflammation and apoptosis. Despite
proteins Bcl-2, Bax, P53 and caspase-3, protect mito- this multifactorial etiology, genetics plays a key role in

© 2005 S. Karger AG, Basel Prof. Xi Can Tang, State Key Laboratory of Drug Research
ABC 1424–862X/05/0142–0071$22.00/0 Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences
Fax + 41 61 306 12 34 Chinese Academy of Sciences, 555 Zu Chong Zhi Road
E-Mail karger@karger.ch Accessible online at: Zhangjiang Hi-Tech Park, Shanghai 201203 (China)
www.karger.com www.karger.com/nsg Tel. +86 21 5080 6710, Fax +86 21 5080 7088, E-Mail xctang@mail.shcnc.ac.cn
disease progression. However, environmental factors (e.g. ly impaired by age [37,79] or by treatment with scopol-
cytokines, neurotoxins) may be even more important in amine [16, 23, 36, 54, 59, 65, 98], AF64A [73, 9], electro-
the development and progression of AD. Several lines of shock [59, 99], cycloheximide [99], NaNO2 [99], or CO2
evidence support the involvement of oxidative stress [2, [37, 98]. In addition, HupA improved cognition in cholin-
40]. Oxidative damage, mediated by reactive oxygen spe- ergically lesioned rats [37, 99], and it reduced the spatial
cies (ROS) generated following cell lysis, oxidative bursts, working memory deficit induced by lesion of the nucleus
or an excess of free transition metals, has been hypothe- basalis magnocellularis [74]. These effects are attributable
sized to play a pivotal role in AD neurodegeneration. On to increased synaptic ACh. Early on, HupA was found to
the other hand, postmortem studies provide direct mor- cause a significant increase in ACh level in rat brain [55,
phological and biochemical evidence that some neurons 100]. More recently, a rise in cortical ACh levels has been
in the AD brain degenerate via an apoptotic mechanism demonstrated by microdialysis in awake, free-moving
[30, 51], which may or may not be linked to ROS. The rats. In this study HupA was 8- and 2-fold more potent
biological mechanism underlying the formation of AD is than donepezil and rivastigmine, respectively, and its
clearly complex, with many factors contributing to the effect was longer lasting [34].
neuropathology. Thus it is not surprising that a number of Clinical trials of HupA in China have demonstrated a
different intervention therapies are currently being re- meaningful improvement in the memory of aged subjects,
searched to address distinct aspects of the disease. individuals with benign senescent forgetfulness and pa-
Huperzine A (HupA) is a novel Lycopodium alkaloid tients with AD. The results indicate minimal peripheral
with recognized medicinal properties. In China the folk cholinergic side effects compared to other AChEIs in use;
medicine Huperzia serrata (Qian Ceng Ta) (fig. 1), a even more important, HupA lacks the dose-limiting hepa-
source of HupA, has been used for centuries in the treat- totoxicity induced by tacrine [75–77, 84, 90]. Adverse
ment of contusions, strains, swelling, schizophrenia, etc. effects in the clinical trials were reported at a very low rate
The active principle, HupA, is a potent, reversible, and and are mainly cholinergic. Examples are dizziness, nau-
selective inhibitor of acetylcholinesterase (AChE) [56]. Its sea, gastroenteric symptoms, headaches and depressed
potency in AChE inhibition is similar or superior to that heart rate. Several clinical trials have addressed the effects
of physostigmine, galanthamine, donepezil and tacrine of HupA on vascular dementia (VD). VD is currently con-
[60, 67]. AChE exists in multiple molecular forms that can sidered to be the second most common form of dementia
be distinguished by their subunit associations and hydro- in Europe and the USA. However, in Asia and many
dynamic properties [6, 41]. In mammalian brain, the bulk developing countries including China, the incidence of
of AChE occurs as a tetrameric, G4 form (10S) together VD exceeds that of AD. Multicenter, randomized, dou-
with much smaller amounts of a monomeric, G1 (4S) ble-blind, placebo-controlled clinical trials in China
form [4, 22]. This phenomenon led us to investigate the proved that HupA markedly improved the cognitive func-
possibility of HupA on differential inhibition of AChE tion of VD [32, 38, 39, 90]. This article will summarize
forms [93]. We observed that HupA preferentially inhibit- and discuss current research focused on elucidating the
ed tetrameric AChE (G4 form), while tacrine and rivastig- neuroprotective effect of HupA.
mine preferentially inhibited monomeric AChE (G1
form). Donepezil showed pronounced selectivity for G1
AChE in striatum and hippocampus, but not in cortex. Protection of HupA against Hydrogen Peroxide
Physostigmine showed no form-selectivity in any brain and ß-Amyloid Protein-Induced Injury by
region [93]. Attenuating Oxidative Stress
HupA has been found to reverse or attenuate cognitive
deficits in a broad range of animal models. Enhancement Several neurodegenerative disorders such as AD, cere-
of learning and memory performance was documented in bral ischemia-reperfusion and head injury are thought to
the passive footshock avoidance paradigm [37, 54, 98, be related to changes in oxidative metabolism. Increased
99], the classic water maze escape task [36, 79], and spa- oxidative stress, resulting from free radical damage to cel-
tial discrimination in the radial arm maze [72]. Similarly, lular function, can be involved in the events leading to
cognition enhancement was found in a delayed-response AD, and is also connected with lesions called tangles and
task in aged monkeys [78] and in reserpine- or yohimbine- plaques. Plaques are caused by the deposition of Aß and
treated monkeys [44]. Beneficial effects were seen not are observed in the brains of AD patients [7, 40, 45, 48].
only in intact adult rodents, but also in rodents cognitive- Studies show that oxygen radicals initiate amyloid build-

72 Neurosignals 2005;14:71–82 Wang/Tang


1

Fig. 1. Chinese herb Huperzia serrata (Qian Ceng Ta).


Fig. 5. Anti-apoptotic mechanism of HupA. Black real line arrows = apoptotic-inducing pathway; red real line ar-
rows = affecting sites of HupA; red dash arrows = speculative pathways; (–) = inhibiting; (+) = promoting.

Hup A: A Natural Cholinesterase Inhibitor Neurosignals 2005;14:71–82 73


for Alzheimer’s Disease
In rat studies, an intracerebroventricular (i.c.v.) infu-
Ab 25-35 sion of ß-amyloid 1–40 (800 pmol !3) induced a signifi-
M Con
HupA cant cognitive deficit, morphologic signs of injury and a
6 12 24 24 (h)
decrease of cortical choline acetyltransferase activity [64].
Daily i.p. administration of HupA for 12 consecutive days
produced partial reversal of the ß-amyloid-induced deficit
in learning a water maze task. This treatment ameliorated
the loss of choline acetyltransferase activity in cerebral
cortex and the neuronal degeneration induced by ß-amy-
loid 1–40 [64]. HupA also reduced the level of lipid perox-
idation and superoxide dismutase in the hippocampus,
cerebral cortex and serum of aged rats [49]. In a rat model
of chronic cerebral hypo-perfusion, HupA significantly
reduced the increases in SOD and lipid peroxide while
restoring lactate and glucose to their normal levels [61]. A
clinical study also demonstrated a reduction of oxygen
free radicals in plasma and erythrocytes from AD patients
[77]. These findings indicate that HupA has protective
Fig. 2. Reduction of Aß25–35-induced DNA fragmentation by effects against free radical and Aß-induced cell toxicity,
HupA. Neurons were treated with 20 ÌM of Aß25–35 with or with- which might be beneficial in the treatment of patients
out 1 ÌM of HupA. Fragmented DNA was isolated by NucleoBond
with various kinds of dementia.
DNA and RNA purification kit, electrophoresed with agarose gel,
and finally stained with ethidium bromide. M = DNA size marker; Experience with HupA enantiomers has shown that
Con = control. the neuroprotective properties have no relation to anti-
cholinesterase potency. Thus, preincubation with (+)-
HupA or (–)-HupA (0.1–10 ÌM ) protected cells with simi-
lar potency against Aß toxicity and similar enhancement
up leading to neurodegeneration [19]. HupA has been of survival [86]. This result contrasted with the stereosel-
found to protect against H2O2 and Aß-induced cell lesion, ectivity of cholinesterase inhibition in vitro and in vivo,
to decrease the level of lipid peroxidation, and to increase in which (–)-HupA is approximately 50-fold more potent
antioxidant enzyme activities in rat PC12 and primary than (+)-HupA. In another study, we examined drug
cultured cortical neurons [68–70]. Following a 6-hour effects on the apoptosis induced by incubation with
exposure of the cells to H2O2 (200 ÌM) or a 48-hour expo- Aß25–35 and on the increase of AChE activity accompa-
sure of the cells to Aß25–35 (1 ÌM ), a marked reduction nying this reaction. We observed that inhibiting the
in cell survival and the activities of glutathione peroxi- hydrolyzing activity of AChE without decreasing AChE
dase (GSH-Px) and catalase (CAT) was observed, along expression itself did not attenuate the Aß25–35 induced
with an increased production of malondialdehyde apoptosis [85]. In other words, the ability of HupA to
(MDA). Pretreatment of the cells with HupA (0.1–10 ÌM) block the catalytic activity of AChE did not parallel its
2 h before H2O2 and Aß exposure caused a significant neuroprotective effect. Therefore, the cytoprotective ef-
increase in cell survival. HupA also partly reversed the fect of HupA enantiomers may relate to some kind of non-
H2O2- and Aß-induced decrease of GSH-Px and CAT catalytic actions on AChE, or to actions on other cellular
activity, as well as the increase in production of MDA and targets.
SOD. All these effects indicate a neuroprotective action.
The protective effect of HupA on Aß-induced cell
lesion was also observed in NG108–15 cells [86] and pri- Anti-Apoptotic Effect of HupA
mary cultured cortical neurons [71]. In addition to elevat-
ing cell survival and GSH-Px and CAT activities while Apoptosis is the process by which neurons die during
decreasing the level of MDA [70], HupA (0.1–10 ÌM) sig- normal development and is also a feature of chronic and
nificantly reduced Aß25–35-induced the formation of acute neurodegenerative diseases and stroke [82]. The cel-
reactive oxygen species (ROS) in rat primary cultured cor- lular commitment to apoptosis is regulated by the Bcl-2
tical neurons [71]. family of proteins. High levels of Bcl-2 expression will

74 Neurosignals 2005;14:71–82 Wang/Tang


Fig. 3. Effects of HupA on H2O2-induced 1 2 3 4 5 6 7 8 9

expression of bcl-2, bax and p53 in PC12 b-Actin


cells by RT-PCR. Cells were exposed to
100 ÌM H2O2, and total RNA was extracted
after the indicated recovery period and then Bcl-2
subjected to RT-PCR. The PCR products
were normalized by ß-actin mRNA. Lane 1: Bax
non-treated intact control; lanes 2–5: 0, 2, 6
and 12 h after 30 min H2O2-treatment, re-
spectively. Lanes 6–9 represent the same P53
time points as lanes 2–5 but preincubated
with 1 ÌM HupA before H2O2 exposure.

inhibit apoptosis. In contrast, an increased expression of characterized by chromatin condensation, nucleus frag-
P53 and Bax is associated with the initiation of apoptosis mentation and DNA laddering, accompanied by altered
[29]. In accordance with previous reports, studies from levels of mRNA for c-jun, p53, bcl-2 and bax. In this mod-
our lab demonstrate typical apoptotic changes when neu- el, HupA significantly attenuated apoptosis and reduced
ron-like cells are exposed to stressors such as H2O2, Aß the up-regulation of c-jun and bax as well as the down-
peptide, oxygen-glucose deprivation (OGD), serum depri- regulation of bcl-2 [94].
vation, or the PKC inhibitor, staurosporine. These In the mitochondrial-mediated cell death pathway, a
changes include DNA laddering, cell shrinkage, the gener- key step is transient opening of the mitochondrial perme-
ation of nuclear apoptotic bodies, TUNEL positive stain- ability transition (MPT), involving a non-specific in-
ing, and other classic hallmarks of apoptosis (fig. 2) [63, crease in the permeability of the inner mitochondrial
71, 87, 94]. Such abnormalities are markedly relieved by membrane [25, 28, 53]. In this process, cytochrome c
HupA. For example, in rats that received i.c.v. injections moves from the intermembrane space into the cytoplasm
of ß-amyloid1–40 (800 pmol ! 3), administration of [5] where it binds to another factor (Apaf-1). In the pres-
HupA (0.1, 0.2 mg/kg, i.p.) for 12 consecutive days gave ence of dATP, this complex polymerizes into an oligomer
substantial neuroprotection in the brain. This treatment known as the apoptosome. The apoptosome activates the
greatly reduced the number of apoptotic-like neurons and protease, caspase-9, which in turn activates caspase-3.
partly reversed the down-regulation of Bcl-2 and up-regu- The cascade of proteolytic reactions also activates
lation of Bax and P53. Anti-apoptotic effects of HupA DNAases, which leads to cell death [83].
were also found in primary cultured neurons. Preincuba- When PC12 cells were pre-incubated with HupA at
tion with HupA at concentrations higher than 0.01 ÌM concentrations above 0.01 ÌM, there was a marked neuro-
led to a large, dose-dependent attenuation of cell toxicity protection against apoptosis induced by ß-amyloid, with a
induced by Aß25–35 (20 ÌM). Moreover, HupA (1 ÌM) significant reduction in mitochondrial swelling and an
caused large reductions in the amounts of subdiploid improvement in mitochondrial membrane potentials [un-
DNA detected in a flow cytometry assay and weakened publ. data of this lab]. Pretreatment of rat cortical neu-
the ladder pattern on agarose gel electrophoresis, typically rons with HupA (0.01–10 ÌM) significantly elevated cell
seen after exposure to Aß (fig. 2). survival and reduced all signs of apoptosis resulting from
The anti-apoptotic actions of HupA may involve inhi- exposure to Aß25–35.
bition of the production or the effects of ROS [71]. We Further studies focused on caspase activation in pri-
found that preincubation of PC12 cells with HupA before mary cultures of rat cortical neurons subjected to a variety
exposure to H2O2 substantially reduced apoptosis, by any of stresses. Measurements of caspase-3-like fluorogenic
of several measures. The same treatment also attenuated cleavage demonstrated that HupA (1 ÌM) attenuated an
H2O2-induced overexpression of bax and p53, while res- Aß25–35-induced increase in caspase-3 activity at 6, 12,
toring bcl-2 to normal levels (fig. 3) [63]. HupA was also 24, and 48 h [71]. Western blot analyses confirmed these
effective in the OGD paradigm. Exposure to OGD for 3 h results at the protein level. HupA also inhibited caspase-3
followed by reoxygenation for 24 h triggers apoptosis activation in models of apoptosis by serum deprivation

Hup A: A Natural Cholinesterase Inhibitor Neurosignals 2005;14:71–82 75


for Alzheimer’s Disease
a b Serum deprivation for 24 h
80
✽✽ Control Huperzine A Vehicle

Caspase-3 activity (U/mg protein)


COX4
Mitochondrial
60 fraction

Cytosolic
fraction
40
Cytochrome c
Mitochondrial
20 fraction

Cytosolic
0 fraction
Control Vehicle Huperzine A

Fig. 4. Effect of HupA on caspase-3 activity (a) and Western blot vation group. b The mitochondrial and cytosolic fractions were iso-
detection of cytochrome c and COX4 (b) in primary cortical neu- lated using an ApoAlert cell fractionation kit. They were then pro-
rons. Neurons under serum deprivation for 24 h. HupA at a concen- cessed using the standard Western blot procedure on 12% SDS-
tration of 1 ÌM added to the culture 2 h in advance. a Data expressed PAGE and probe with COX4 antibody (F17 kDa) or cytochrome c
as means B SD. Statistical comparison was made using ANOVA fol- antibody (F15 kDa). The presence of cytochrome c in the cytosolic
lowed by Duncan’s test. There was a significant difference between fraction after induction indicates that apoptosis involves mitochon-
the serum deprivation group and the untreated control group. ## p ! drial release of cytochrome c to the cytosol.
0.01 compared to control group. * p ! 0.05 compared to serum depri-

and staurosporine treatment. The apoptosis induced by In light of these findings and the effects of HupA on
24 h of serum deprivation was accompanied by enhanced apoptosis-related genes, we propose that HupA blocks
caspase-3 activity and a release of mitochondrial cyto- apoptosis by antagonizing the mitochondrial-dependent
chrome c into the cytosol [97]. HupA (0.1–10 ÌM) im- caspase pathway, directly or indirectly (fig. 5). The effects
proved neuronal survival in this model, inhibiting the rise of HupA on the intrinsic caspase-3 pathway might be
in caspase-3 activity and protein expression [97]. Like- downstream consequences of altered expression of bcl-2
wise, cell survival was greatly enhanced when HupA (0.1– family genes. Functionally, bcl-2 is a potent cell death
100 ÌM) was introduced 2 h before a 24-hour exposure to suppressor, whose over-expression can prevent cell death
0.5 ÌM staurosporine. Incubation with HupA at dose of in response to a variety of stimuli. It is well known that
1 ÌM also reduced staurosporine-induced DNA fragmen- Bcl-2 suppresses apoptosis by inhibiting cytochrome c
tation, up-regulation of the pro-apoptotic gene, bax, down- release from the mitochondria. On the other hand, bax is
regulation of the anti-apoptotic gene, bcl-2, and decrease a death-promoting factor, whose translocation to the mi-
in caspase-3 proenzyme protein level (fig. 4) [87]. tochondrial membrane leads to a loss of mitochondrial
A potassium channel with delayed rectifier characteris- membrane potential and increases mitochondrial perme-
tics may play an important role in Aß-mediated toxicity. ability. Increased mitochondrial permeability results in
The up-regulation of an outward K+ current known as Ik the release of cytochrome c followed by activation of cas-
mediates several forms of neuronal apoptosis and could pase-3 [21]. We consider it likely that HupA owes some of
contribute to the pathogenesis of Aß-induced neuronal its anti-apoptotic effects to an effective antagonism of the
death. Exposure to a 20-ÌM concentration of Aß25–35 or up-regulation of bax and the down-regulation of bcl-2,
Aß1–42 is known to enhance the apoptosis-related cur- which impairs mitochondria-dependent caspase pathway.
rent, Ik [81]. Interestingly, HupA will reversibly inhibit At present, however, direct effects of HupA on cyto-
the fast transient current, IA, and the sustained potassium chrome c and caspase-3 and other possible targets are not
current, Ik, in CA1 pyramidal neurons acutely dissociated excluded.
from rat hippocampus [33]. Such effects might contribute
to this agent’s anti-apoptotic effect.

76 Neurosignals 2005;14:71–82 Wang/Tang


Effects of HupA on Secretory Amyloid Our recent results provide the first demonstration that
Precursor Protein and Protein Kinase C-· HupA can reduce the disturbance of PKC and APPs both
in rats and in an isolated cell line. The effect of HupA to
Aß is a self-aggregating 39–43-amino acid peptide that enhance non-amyloidogenic processing of APP and ele-
originates from a larger polypeptide termed Alzheimer’s vate APPs levels likely depends on the activation of mus-
amyloid precursor protein (APP). Alternate pathways for carinic receptors and the PLC/PKC cascade. A number of
APP processing have been described: the non-amyloido- biological activities such as cell proliferation, promotion
genic secretory pathway, which releases a soluble ectodo- of cell-substratum adhesion, neurite outgrowth and the
main (APPs) and prevents Aß formation [15], and the prevention of intracellular calcium accumulation and cell
endosomal-lysosomal pathway, which produces amyloi- death have been attributed to APPs [47]. Since PKC is a
dogenic products [24]. The amyloid hypothesis of AD [17, key enzyme in signal transduction, and since APPs itself
46] is focused on the potential toxic role of an excessive has neuroprotective effects, modulating the levels of these
production of Aß and suggests that the aberrant metabo- two proteins by HupA may well be beneficial in AD thera-
lism of APP is a central pathogenetic mechanism for the py (fig. 6).
disease.
Several factors can affect the secretory non-amyloido-
genic pathway of APP. For example, the stimulation of Protection of HupA against Hypoxic-Ischemic
phospholipase C (PLC)-coupled receptors, such as musca- and Glutamate Induced Brain Injury and
rinic m1 and m3, has been shown to potentiate the secre- Cytotoxicity
tion of APP in cell cultures. These effects are probably
mediated mainly by protein kinase C (PKC) [43]. It has Apart from AD, the most common dementia in the
also been reported that several anticholinesterases affect elderly is VD. This disorder, like AD, presents a clinical
APP processing in addition to the catalytic function of syndrome of intellectual decline produced by ischemia,
AChE [18, 42]. hypoxia, or hemorrhagic brain lesion. Cerebral ischemia
Our own studies showed that HupA could alter APP in rats with permanent bilateral ligation of the common
processing in the brains of rats given i.c.v. infusions of carotid arteries (CCA) provides a useful model of VD, in
Aß1–40, and in otherwise untreated human embryonic which to investigate the effects of HupA. These animals
kidney 293 (HEK293sw) cells [88]. In the Aß treated rats, experience a significant reduction of cerebral blood flow
levels of APPs and PKC· were significantly decreased by and exhibit learning and memory impairments and neu-
treatment with Aß1–40. These decreases were much re- ronal damage resembling those in VD. Daily oral admin-
duced by 12 consecutive days of HupA treatment (0.2 mg/ istration of HupA (0.1 mg/kg) to such rats for 14 days pro-
kg, i.p.), but HupA in normal rats caused no change in duced significant improvement in the learning of a water
either APPs or PKC·. In normal HEK293sw cells, on the maze task. Simultaneously there was marked recovery
other hand, the levels of APPs and PKC· rose progres- from the decrease in choline acetyltransferase activity in
sively during an 18-hour exposure to HupA (1 ÌM). How- hippocampus and a restoration of SOD, lipid peroxide,
ever, no significant alternations in the levels of PKC‰ and lactate and glucose to normal levels [61]. Similar protec-
PKCÂ were found after HupA treatment. Taken together tion was also observed in gerbils given subchronic oral
these findings suggest that HupA may affect the process- doses of HupA (0.1 mg/kg, twice daily for 14 days) follow-
ing of APP by up-regulating PKC, especially PKC·. ing 5 min of global ischemia [96].
In an attempt to clarify the receptor mechanisms An in vitro model of neuronal ischemia is the rat
involved in such effects, we treated HEK293wt cells with pheochromocytoma PC12 cell treated with OGD for
cholinergic receptor antagonists [unpubl. data]. The non- 30 min. In our hands, this treatment causes death in more
selective muscarinic antagonist, scopolamine, partly than 50% of the cells in culture, along with major changes
blocked the HupA-induced rise in levels of APPs and in morphology and biochemistry, including elevated lev-
PKC·. By contrast the nicotinic antagonist, mecamyl- els of lipid peroxide, SOD activity and lactate. Cells pre-
amine, had little effect. These results suggest that musca- treated for 2 h with HupA (0.1, 1 and 10 ÌM), however,
rinic ACh receptors may mediate, at least in part, the showed increased survival and reduced biochemical and
effects of HupA on the regulation of APPs and PKC· in morphologic signs of toxicity. HupA protected PC12 cells
HEK293sw cells. against OGD-induced toxicity, most likely by alleviating
disturbances of oxidative and energy metabolism [95].

Hup A: A Natural Cholinesterase Inhibitor Neurosignals 2005;14:71–82 77


for Alzheimer’s Disease
Fig. 6. Protective effects of HupA through affecting APP metabolism. Black arrows show the process of AD. Blue real
line arrows show the proved pathway; blue dash arrow represents speculative pathway; (–) means reducing or inhib-
iting.

These findings suggested that HupA might be beneficial water maze performance, as shown by an increased escape
for VD therapy through its effects on the cholinergic sys- latency and a reduced time spent in the target quadrant.
tem, the oxygen free radical system and energy metabo- The combination of CCA ligation and exposure to a hyp-
lism. oxic environment also led to morphologic damage in the
A protective effect of HupA on hypoxic–ischemic (HI) ipsilateral striatum, cortex, and also hippocampus, where
brain injury has also been found in neonatal rats [62]. A it produced 12% neuronal loss in the CA1 region. Treat-
unilateral HI brain injury was produced in 7-day-old rat ment with HupA at a dose of 0.1 mg/kg conferred signifi-
pups by the ligation of the left CCA followed by 1 h hyp- cant protection against the behavioral and morphologic
oxia with 7.7% oxygen. After 5 weeks, the HI brain injury consequences of HI injury (fig. 7). The same treatment
in these pups caused working memory impairments in spared a significant fraction of the CA1 neurons relative

78 Neurosignals 2005;14:71–82 Wang/Tang


Fig. 7. Photomicrographs of coronal brain sections stained with cresyl violet at the levels of the striatum (a, b) and the
dorsal hippocampus (c, d) for representative saline-treated and huperzine A 0.1 mg/kg-treated rats. Intraperitoneal
administration of huperzine A or saline for 5 weeks after hypoxic-ischemic (HI) brain injury in neonatal rats. Note the
gross infarction and atrophy in left hemisphere of saline-treated HI rats (a, c) (n = 11) and the subtle reduction in the
left hemisphere in huperzine A-treated HI rats (b, d) (n = 12).

to saline-treated HI group. These results raise the possibil- 100 ÌM). In those experiments, HupA reduced gluta-
ity that HupA have potential utility in treating HI enceph- mate-induced calcium mobilization but did not affect the
alopathy in neonates. increase in intracellular free calcium induced by exposure
Glutamate is the main excitatory neurotransmitter in to high KCl or a calcium activator Bay-K-8644 [58].
the CNS, with important roles in neurotransmission and HupA dose-dependently inhibited the NMDA-induced
functional plasticity. Excitatory amino acid neurotrans- toxicity in primary neuronal cells, most likely by blocking
mitters are also involved in CNS pathology. The deleteri- NMDA ion channels and the subsequent Ca2+ mobiliza-
ous effects of overstimulation with excitatory amino acids tion at or near the PCP and MK-801 ligand sites [20].
have been implicated in a variety of acute and chronic Wang et al. [66] reported that HupA reversibly inhibited
neurodegenerative disorders such as ischemic brain dam- NMDA-induced current in acutely dissociated rat hippo-
age, AD and neuronal cell death [11] [for reviews, see 13, campal pyramidal neurons and blocked specific [3H]MK-
14, 26, 27, 35]. Glutamate-mediated overactivation of 801 binding in synaptic membranes from rat cerebral cor-
receptors induces excessive Ca2+ influx, which results in tex. Of all the AChE inhibitors tested, HupA is the most
elevated intracellular Ca2+ concentrations [10, 12] with powerful both in protecting mature neurons and in block-
serious consequences such as necrosis and apoptosis [31]. ing the binding of [3H]MK-801. Studies on the mecha-
Blockade of glutamate receptors prevents most of the Ca2+ nism of receptor inhibition showed that HupA reversibly
influx and neuronal cell death induced by glutamate expo- inhibited NMDA-induced currents. The effect was non-
sure [50, 57]. competitive, and showed neither ‘voltage-dependency’,
It has been reported that HupA protects against gluta- nor ‘use-dependency’ [89]. Studies of [3H]MK-801 bind-
mate-induced toxicity. HupA (100 ÌM) decreased neu- ing in cortex membranes suggest that HupA acts as a non-
ronal cell death caused by a toxic level of glutamate (also competitive antagonist of the NMDA receptors, via a

Hup A: A Natural Cholinesterase Inhibitor Neurosignals 2005;14:71–82 79


for Alzheimer’s Disease
competitive interaction with one of the polyamine bind- Neuronal cell death caused by overstimulation of glu-
ing sites [92]. Of interest, natural (–)-HupA and synthetic tamate receptors has been proposed as the final common
(+)-HupA reduced the binding of [3H]MK-801 with simi- pathway for a variety of neurodegenerative diseases in-
lar potencies [91] indicating that HupA inhibits NMDA cluding AD. The ability of HupA to attenuate glutamate-
receptors in rat cerebral cortex without stereoselectivity. mediated neurotoxicity may be one additional reason for
This result is in dramatic contrast with the stereoselective considering this agent as a potential therapeutic for de-
inhibition of acetylcholinesterase. mentia and as a means of slowing or halting the pathogen-
NMDA-receptor activation also mediates the genera- esis of AD at an early stage [20].
tion of long-term potentiation (LTP) – a cellular process
that underlies learning and memory [3, 52]. There is evi-
dence that the suppressive action of Aß on LTP in both Acknowledgements
CA1 and dentate gyrus operates via a NMDA receptor-
This work was supported in part by the grants from Ministry
independent pathway that involves cholinergic terminals
of Science and Technology of China (G199805110, G1998051115)
in the hippocampus. Of some interest, HupA (1.0 ÌM) and National Natural Science Foundation of China (39170860,
was found to enhance LTP, while a much lower dose 39770846, 3001161954, 30123005 and 30271494). The authors are
(0.1 ÌM) largely blocked the suppressive effects of Aß on grateful to Professor Stephen W. Brimijoin (Mayo Clinic, USA) for
LTP induction [8, 80]. English revision on the manuscript.

References

1 Bartus RT, Dean RL 3rd, Beer B, Lippa AS: 10 Choi DW: Calcium-mediated neurotoxicity: 20 Gordon RK, Nigam SV, Weitz JA, Dave JR,
The cholinergic hypothesis of geriatric memory Relationship to specific channel types and role Doctor BP, Ved HS: The NMDA receptor ion
dysfunction. Science 1982;217:408–414. in ischemic damage. Trends Neurosci 1988;11: channel: A site for binding of huperzine A. J
2 Behl C, Moosmann B: Antioxidant neuropro- 465–469. Appl Toxicol 2001;21(suppl 1):S47–S51.
tection in Alzheimer’s disease as preventive 11 Choi DW: Calcium and excitotoxic neuronal 21 Graham SH, Chen J: Programmed cell death in
and therapeutic approach. Free Radic Biol injury. Ann NY Acad Sci 1994;747:162–171. cerebral ischemia. J Cereb Blood Flow Metab
Med 2002;33:182–191. 12 Choi DW: Calcium: still center-stage in hypox- 2001;21:99–109.
3 Bliss TVP, Collingridge GL: A synaptic model ic-ischemic neuronal death. Trends Neurosci 22 Grassi J, Vigny M, Massoulie J: Molecular
of memory: long-term potentiation in the hip- 1995;18:58–60. forms of acetylcholinesterase in bovine caudate
pocampus. Nature 1993;361:31–39. 13 DiFiglia M: Excitotoxic injury of the neostria- nucleus and superior cervical ganglion: Solubil-
4 Bon S, Vigny M, Massoulie J: Asymmetric and tum: A model for Huntington’s disease. Trends ity properties and hydrophobic character. J
globular forms of AChE in mammals and birds. Neurosci 1990;13:286–289. Neurochem 1982;387:457–469.
Proc Natl Acad Sci USA 1979;76:2540–2550. 14 Dirnagl U, Iadecola C, Moskowitz MA: Patho- 23 Guan LC, Chen SS, Cui QG, Lu WH, Tang
5 Bossy-Wetzel E, Newmeyer DD, Green DR: biology of ischaemic stroke: An integrated XC: The effects of huperzine A on behavior
Mitochondrial cytochrome c release in apopto- view. Trends Neurosci 1999;22:391–397. and ECoG in animals. Acta Psychol Sin 1991;
sis occurs upstream of DEVD-specific caspase 15 Esch FS, Keim PS, Beattie EC, Blacher RW, 23:404–411.
activation and independently of mitochondrial Culwell AR, Oltersdorf T, McClure D, Ward 24 Haass C, Selkoe DJ: Cellular processing of
transmembrane depolarization. EMBO J 1998; PJ: Cleavage of amyloid beta-peptide during beta-amyloid precursor protein and the genesis
17:37–49. constitutive processing of its precursor. Science of amyloid-beta peptide. Cell 1993;75:1039–
6 Brimijoin S: Molecular forms of acetylcholines- 1990:248:1122–1124. 1042.
terase in brain, nerve and muscle: nature, local- 16 Gao Y, Tang XC, Guan LC, Kuang PZ: Huper- 25 Halestrap AP, McStay GP, Clarke SJ: The per-
ization and dynamics. Prog Neurobiol 1983; zine A reverses scopolamine- and muscimol- meability transition pore complex: Another
21:291–322. induced memory deficits in chick. Acta Phar- view. Biochimie 2000;84:153–166.
7 Butterfield DA, Howard B, Yatin S, Koppal T, macol Sin 2000;21:1169–1173. 26 Hossmann KA: Glutamate-mediated injury in
Drake J, Hensley K, Aksenov M, Subrama- 17 Gasparini L, Racchi M, Binetti G, Trabucchi focal cerebral ischemia: The excitotoxin hy-
niam R, Varadarajan S, Harris-White ME, Pe- M, Solerte SB, Alkon D, Etcheberrigaray R, pothesis revised. Brain Pathol 1994;4:23–36.
digo NW Jr, Carney JM: Elevated oxidative Gibson G, Blass J, Paoletti R, Govoni S: Pe- 27 Hynd MR, Scott HL, Dodd PR: Glutamate-
stress in models of normal brain aging and Alz- ripheral markers in testing pathophysiological mediated excitotoxicity and neurodegenera-
heimer’s disease. Life Sci 1999;65:1883–1892. hypotheses and diagnosing Alzheimer’s dis- tion in Alzheimer’s disease. Neurochem Int
8 Chen QS, Kagan BL, Hirakura Y, Xie CW: ease. FASEB J 1998;12:17–34. 2004;45:583–595.
Impairment of hippocampal long-term poten- 18 Giacobini E, Mori F, Lai CC: The effect of cho- 28 Kim JS, He L, Lemasters JJ: Mitochondrial
tiation by Alzheimer amyloid beta-peptides. J linesterase inhibitors on the secretion of APPs permeability transition: A common pathway to
Neurosci Res 2000;60:65–72. from rat brain cortex. Ann NY Acad Sci 1996; necrosis and apoptosis. Biochem Biophys Res
9 Cheng DH, Tang XC: Comparative studies of 777:393–398. Commun 2003;304:463–470.
huperzine A, E2020, and tacrine on behavior 19 Gilgun-Sherki Y, Melamed E, Offen D: Antiox- 29 Ko LJ, Prives C: p53: Puzzle and paradigm.
and cholinesterase activities. Pharmacol Bio- idant treatment in Alzheimer’s disease: Cur- Genes Dev 1996;10:1054–1072.
chem Behav 1998;60:377–386. rent state. J Mol Neurosci 2003;21:1–12.

80 Neurosignals 2005;14:71–82 Wang/Tang


30 Eckert A, Marques CA, Keil U, Schussel K, 46 Racchi M, Govoni S: Rationalizing a pharma- 62 Wang LS, Zhou J, Shao XM, Tang XC: Huper-
Muller WE: Increased apoptotic cell death in cological intervention on the amyloid precur- zine A attenuates cognitive deficits and brain
sporadic and genetic Alzheimer’s disease. Ann sor protein metabolism. Trends Pharmacol Sci injury in neonatal rats after hypoxia-ischemia.
NY Acad Sci 2003;1010:604–609. 1999;20:418–423. Brain Res 2002;949:162–170.
31 Lee JM, Zipfel GJ, Choi DW: The changing 47 Rossner S, Ueberham U, Schliebs R, Perez- 63 Wang R, Xiao XQ, Tang XC: Huperzine A
landscape of ischaemic brain injury mecha- Polo JR, Bigl V: The regulation of amyloid attenuates hydrogen peroxide-induced apopto-
nisms. Nature 1999;399(suppl):A7–A14. precursor protein metabolism by cholinergic sis by regulation expression of apoptosis-relat-
32 Li HN, Min QY: Huperzine A improved the mechanisms and neurotrophin receptor signal- ed genes in rat PC12 cells. NeuroReport 2001;
cognition of vascular dementia: A report of 30 ing. Prog Neurobiol 1998;56:541–569. 12:2629–2634.
patients in therapeutics. Xian Dai Kang Fu 48 Selkoe DJ, Abraham CR, Podlisny MB, Duffy 64 Wang R, Zhang HY, Tang XC: Huperzine A
2001;5:59 LK: Isolation of low-molecular-weight proteins attenuates cognitive dysfunction and neuronal
33 Li Y, Hu GY: Huperzine A inhibits the sus- form amyloid plaque fibers in Alzheimer’s dis- degeneration caused by beta-amyloid protein-
tained potassium current in rat dissociated hip- ease. J Neurochem 1986;46:1820–1834. (1–40) in rat. Eur J Pharmacol 2001;421:149–
pocampal neurons. Neurosci Lett 2002;329: 49 Shang YZ, Ye JW, Tang XC: Improving effects 156.
153–156. of huperzine A on abnormal lipid peroxidation 65 Wang T, Tang XC: Reversal of scopolamine-
34 Liang YQ, Tang XC: Comparative effects of and superoxide dismutase in aged rats. Acta induced deficits in radial maze performance by
huperzine A, donepezil and rivastigmine on Pharmacol Sin 1999;20:824–828. (–)-huperzine A: Comparison with E2020 and
cortical acetylcholine level and acetylcholines- 50 Simon RP, Swan JH, Griffiths T, Meldrum BS: tacrine. Eur J Pharmacol 1998;349:137–142.
terase activity in rats. Neurosci Lett 2004;361: Blockade of N-methyl-aspartate receptors may 66 Wang XD, Zhang JM, Yang HH, Hu GY:
56–59. protect against ischemic damage in the brain. Modulation of NMDA receptor by huperzine A
35 Lipton SA, Rosenberg PA: Excitatory amino Science 1984;226:850–852. in rat cerebral cortex. Acta Pharmacol Sin
acids as a final common pathway for neurologic 51 Smale G, Nichols NR, Brady DR, Finch CE, 1999;20:31–35.
disorders. N Engl J Med 1994;330:613–622. Horton WE Jr: Evidence for apoptotic cell 67 Wang YE, Yue DX, Tang XC: Anticholinester-
36 Liu J, Zhang HY, Tang XC, Wang B, He XC, death in Alzheimer’s disease. Exp Neurol 1995; ase activity of huperzine A. Acta Pharmacol
Bai DL: Effects of synthetic (–)-huperzine A on 133:225–230. Sin 1986;7:110–113.
cholinesterase activities and mouse water maze 52 Sucher NJ, Awobuluyi M, Choi YB, Lipton SA: 68 Xiao XQ, Yang JW, Tang XC: Huperzine A
performance. Acta Pharmacol Sin 1998;19: NMDA receptors: From genes to channels. protects rat pheochromocytoma cells against
413–416. Trends Pharmacol Sci 1996;17:348–355. hydrogen peroxide-induced injury. Neurosci
37 Lu WH, Shou J, Tang XC: Improving effect of 53 Szalai G, Krishnamurthy R, Hajnoczky G: Lett 1999;275:73–76.
huperzine A on discrimination performance in Apoptosis driven by IP(3)-linked mitochon- 69 Xiao XQ, Wang R, Han YF and Tang XC: Pro-
aged rats and adult rats with experimental cog- drial calcium signals. EMBO J 1999;18:6349– tective effects of huperzine A on ß-amyloid25–
nitive impairment. Acta Pharmacol Sin 1988; 6361. 25 induced oxidative injury in rat pheochro-
9:11–15. 54 Tang XC, Han YF, Chen XP, Zhu XD: Effects mocytoma cells. Neurosci Lett 2000;286:155–
38 Ma YX, Zhu Y, Gu YD, Yu ZY, Yu SM, Ye of huperzine A on learning and retrieval pro- 158.
YZ: Double-blind trial of huperzine-A (HUP) cess of discrimination performance in rats. 70 Xiao XQ, Wang R, Tang XC: Huperzine A and
on cognitive deterioration in 314 cases of be- Acta Pharmacol Sin 1986;7:507–511. tacrine attenuate ß-amyloid peptide induced
nign senescent forgetfulness, vascular demen- 55 Tang XC, De Sarno P, Sugaya K, Giacobini E: oxidative injury. J Neurosci Res 2000;61:564–
tia, and Alzheimer’s disease. Ann NY Acad Sci Effect of huperzine A, a new cholinesterase 569.
1998;854:506–507. inhibitor, on the central cholinergic system of 71 Xiao XQ, Zhang HY, Tang XC: Huperzine A
39 Pi X, Liu Y, Jiang ZY, Hu XQ, Zhu GZ: Clini- the rat. J Neurosci Res 1989;24:276–285. attenuates amyloid ß-peptide fragment 25–35-
cal observations on treatment of light and mod- 56 Tang XC, He XC, Bai DL: Huperzine A: A induced apoptosis in rat cortical neurons via
erate vascular dementia with meclofenoxate novel acetylcholinesterase inhibitor. Drug Fu- inhibiting reactive oxygen species formation
plus huperzine A. Shanghai Med Pharmaceut J ture 1999;24:647–663. and caspase-3 activation. J Neurosci Res 2002;
2004;255:409–411. 57 Turski L, Huth A, Sheardown M, McDonald F, 67:30–36.
40 Markesbery WR: Oxidative stress hypothesis Neuhaus R, Schneider HH, Dirnagl U, Wie- 72 Xiong ZQ, Tang XC: Effect of huperzine A, a
in Alzheimer’s disease. Free Rad Biol Med gand F, Jacobsen P, Ottow E: ZK200775: A novel acetylcholinesterase inhibitor, on radial
1997;23:134–147. phosphonate quinoxalinedione AMPA antago- maze performance in rats. Pharmacol Biochem
41 Massoulie J, Bon S: The molecular forms of nist for neuroprotection in stroke and trauma. Behav 1995;51:415–419.
cholinesterase and acetylcholinesterase in ver- Proc Natl Acad Sci USA 1998;95:10960– 73 Xiong ZQ, Han YF, Tang XC: Huperzine A
tebrate. Annu Rev Neurosci 1982;5:57–106. 10965. ameliorates the spatial working memory im-
42 Mori F, Lai CC, Fusi F, Giacobini E: Cholines- 58 Ved HS, Koening ML, Dave JR, Doctor BP: pairments induced by AF64A. NeuroReport
terase inhibitors increase secretion of APPs in Huperzine A, a potential therapeutic agent for 1995;6:2221–2224.
rat brain cortex. NeuroReport 1995;6:633– dementia, reduces neuronal cell death caused 74 Xiong ZQ, Cheng DH, Tang XC: Effects of
636. by glutamate. NeuroReport 1997;8:963–968. huperzine A on nucleus basalis magnocellularis
43 Nitsch RM, Slack BE, Wurtman RJ, Growdon 59 Vincent GP, Rumennik L, Cumin R, Martin J, lesion-induced spatial working memory deficit.
J: Release of Alzheimer amyloid precursor de- Sepinwall J: The effects of huperzine A, an ace- Acta Pharmacol Sin 1998;19:128–132.
rivative stimulated by activation of muscarinic tylcholinesterase inhibitor, on the enhance- 75 Xu SS, Gao ZX, Weng Z, Du ZM, Xu WA,
acetylcholine receptors. Science 1992;258: ment of memory in mice, rats and monkeys. Yang JS, Zhang ML, Tong ZH, Fang YS, Chai
304–307. Neurosci Abstr 1987;13:844. XS, Li SL: Efficacy of tablet huperzine A on
44 Ou LY, Tang XC, Cai JX: Effect of huperzine 60 Wang H, Tang XC: Anticholinesterase effects memory, cognition, and behavior in Alzhei-
A on working memory in reserpine- or yohim- of huperzine A, E2020, and tacrine in rats. Acta mer’s disease. Acta Pharmacol Sin 1995;16:
bine-treated monkeys. Eur J Pharmacol 2001; Pharmacol Sin 1998;19:27–30. 391–395.
433:151–156. 61 Wang LM, Han YF, Tang XC: Huperzine A 76 Xu SS, Xie HB, Du ZW, Tong ZH, Shi QC, Lu
45 Perry EK, Tomlinson VE, Blessed G, Bergman improves cognitive deficits caused by chronic KM, Li SL, Lin B: Efficacy of tablet huperzine
K, Gibson PH, Perry RH: Correlation of cho- cerebral hypoperfusion in rats. Eur J Pharmo- A on memory and cognition in patients with
linergic abnormalities with senile plaques and col 2000;398:65–72. benign senescent forgetfulness. Chin J Clin
mental scores. Br Med J 1978;ii:1457–1459. Pharmacol Ther 1997;2:1–4.

Hup A: A Natural Cholinesterase Inhibitor Neurosignals 2005;14:71–82 81


for Alzheimer’s Disease
77 Xu SS, Cai ZY, Qu ZW, Yang RM, Cai YL, 86 Zhang HY, Liang YQ, Tang XC, He XC, Bai 92 Zhang YH, Zhao XY, Chen XQ, Wang Y,
Wang GQ: Huperzine A in capsules and tablets DL: Stereoselectivities of enantiomers of hu- Yang HH, Hu GY: Spermidine antagonizes
for treating patients with Alzheimer’s disease. perzine A in protection against amyloid 25–35- the inhibitory effect of huperzine A on [3H]di-
Acta Pharmacol Sin 1999;20:486–490. induced injury in PC12 and NG108–15 cells zocilpine (MK-801) binding in synaptic
78 Ye JW, Cai JX, Wang LM, Tang XC: Improv- and cholinesterase inhibition in mice. Neurosci membrane of rat cerebral cortex. Neurosci
ing effects of huperzine A on spatial working Lett 2002;317:143–146. Lett 2002;319:107–110.
memory in aged monkeys and young adult 87 Zhang HY, Tang XC: Huperzine A attenuates 93 Zhao Q, Tang XC: Effects of huperzine A on
monkeys with experimental cognitive impair- the neurotoxic effect of staurosporine in prima- acetylcolinesterase isoforms in vitro: Com-
ment. J Pharmacol Exp Ther 1999;288:814– ry rat cortical neurons. Neurosci Lett 2003; parison with tacrine, donepezil, rivastigmine
819. 340:91–94. and physostigmine. Eur J Pharmacol 2002;
79 Ye JW, Shang YZ, Wang ZM, Tang XC: Hu- 88 Zhang HY, Yan H, Tang XC: Huperzine A 455:101–107.
perzine A ameliorates the impaired memory of enhances the level of secretory amyloid precur- 94 Zhou J, Fu Y, Tang XC: Huperzine A pro-
aged rat in the Morris water maze perfor- sor protein and protein kinase C-· in intracere- tects rat pheochromocytoma cells against oxy-
mance. Acta Pharmacol Sin 2000;21:65–69. broventricular ß-amyloid-(1–40) infused rats gen-glucose deprivation. NeuroReport 2001;
80 Ye L, Qiao JT: Suppressive action produced by and human embryonic kidney 293 Swedish 12:2073–2077.
beta-amyloid peptide fragment 31–35 on long- mutant cells. Neurosci Lett 2004;360:21–24. 95 Zhou J, Fu Y, Tang XC: Huperzine A and
term potentiation in rat hippocampus is N- 89 Zhang JM, Hu GY: Huperzine A, a nootropic donepezil protect rat pheochromocytoma
methyl-D-aspartate receptor-independent: it’s alkaloid, inhibits N-methyl-D-aspartate-in- cells against oxygen-glucose deprivation.
offset by (–)huperzine A. Neurosci Lett 1999; duced current in rat dissociated hippocampal Neurosci Lett 2001;306:53–56.
275:187–190. neurons. Neuroscience 2001;105:663–669. 96 Zhou J, Zhang HY, Tang XC: Huperzine A
81 Yu SP, Farhangrazi ZS, Ying HS, Yeh CH, 90 Zhang RW, Tang XC, Han YY, Sang GW, attenuates cognitive deficits and hippocam-
Choi DW: Enhancement of outward potassium Zhang YD, Ma YX, Zhang CL, Yang RM: pal neuronal damage after transient global
current may participate in beta-amyloid pep- Drug evaluation of huperzine A in the treat- ischemia in gerbils. Neurosci Lett 2001;313:
tide-induced cortical neuronal death. Neuro- ment of senile memory disorders. Acta Phar- 137–140.
biol Dis 1998;5(2):81–88. macol Sin 1991;12:250–252. 97 Zhou J, Tang XC: Huperzine A attenuates
82 Yuan J, Yankner BA: Apoptosis in the nervous 91 Zhang YH, Chen XQ, Yang HH, Jin GY, Bai apoptosis and mitochondria-dependent cas-
system. Nature 2000;407:802–809. DL, Hu GY: Similar potency of the enantio- pase-3 in rat cortical neurons. FEBS Lett
83 Zamzami N, Kroemer G: The mitochondrion mers of huperzine A in inhibition of [(3)H]di- 2002;526:21–25.
in apoptosis: How Pandora’s box opens. Na- zocilpine (MK-801) binding in rat cerebral cor- 98 Zhu XD, Tang XC: Facilitatory effects of
ture Reviews in Molecular and Cellular Biology tex. Neurosci Lett 2000;295:116–118. huperzine A and B on learning and memory
2001;21:67–71. of spatial discrimination in mice. Acta Phar-
84 Zhang CL, Wang GZ: Effects of huperzine A macol Sin 1987;22:812–817.
tablet on memory. New Drugs Clin Remed 99 Zhu XD, Tang XC: Improvement of im-
1990;9:339–341. paired memory in mice by huperzine A and
85 Zhang HY, Brimijoin S, Tang XC: Apoptosis huperzine B. Acta Pharmacol Sin 1988;9:
induced by ß-amyloid25–35 in acetylcholin- 492–497.
esterase-overexpressing neuroblastoma cells. 100 Zhu XD, Giacobini E: Second generation
Acta Pharmacol Sin 2003;24:853–858. cholinesterase inhibitors: Effect of (L)-huper-
zine A on cortical biogenic amines. J Neuro-
sci Res 1995;41:828–835.

82 Neurosignals 2005;14:71–82 Wang/Tang


Author Index Vol. 14, No. 1–2, 2005

Amit, T. 46
Avramovich-Tirosh, Y. 46
Doré, S. 61
Houghton, P.J. 6
Howes, M.-J. 6
Komatsu, K. 34
Kuboyama, T. 34
Mandel, S.A. 46
Reznichenko, L. 46
Suk, K. 23
Tang, X.C. 71
Tohda, C. 34
Wang, R. 71
Youdim, M.B.H. 46
Zheng, H. 46

© 2005 S. Karger AG, Basel 83


ABC
Fax + 41 61 306 12 34
E-Mail karger@karger.ch Accessible online at:
www.karger.com www.karger.com/nsg
Subject Index Vol. 14, No. 1–2, 2005

Acetylcholine 6 Galantamine 6
Acetylcholinesterase 71 Ginseng 34
Alzheimer’s disease 6, 34, 71 Glutamate 71
Amyloid 34, 71 Green tea catechins 46
– precursor protein 71
Apoptosis 71 Hemin 61
Ashwagandha 34 Huperzine A 6, 71
Astrocytes, neuroinflammation 23
Axon regeneration 34 Inflammation 23
Iron 61
Bilirubin 61 – chelation 46
Biliverdin 61
Blood flow 61 Microglia 23
Mitochondria 71
Carbon monoxide 61
Cell signaling 46 Neurite outgrowth 46
Central nervous system, herbal medicine Neuritic atrophy 34
23 Neurodegeneration 23, 46
Cerebral ischemia 71 Neuroglia 23
Cholinergic receptors 6 Neuroprotection 23, 46, 71
Cholinesterase inhibitors 6, 71 Neurorescue 46
Coffee beans, trigonelline 34 NMDA receptor 71
Cognitive enhancer, huperzine A 71
Oxidative stress 71
Dendrite regeneration 34
L-DOPA 6 Parkinson’s disease 6, 46
Dopamine 6 Physostigmine 6
Dopaminergic receptors 6 Polyphenol stilbenes 61
Potassium current 71
(–)-Epigallocatechin-3-gallate 46 Protein kinase C 46
Ergot alkaloids 6
Synaptic loss 34
Flavonoids 23
Withania somnifera 34

© 2005 S. Karger AG, Basel


ABC
Fax + 41 61 306 12 34
E-Mail karger@karger.ch Accessible online at:
www.karger.com www.karger.com/nsg

You might also like