Anda di halaman 1dari 25

bs_bs_banner

Review

Journal of Pharmacy
And Pharmacology

Alkaloids as a source of potential anticholinesterase


inhibitors for the treatment of Alzheimers disease
Eduardo Luis Konrath, Carolina dos Santos Passos, Luiz Carlos Klein-Jnior and Amlia T. Henriques
Programa de Ps-Graduao em Cincias Farmacuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil

Keywords
acetylcholinesterase inhibition; alkaloids;
Alzheimers disease; butyrylcholinesterase
inhibition; plants
Correspondence
Eduardo L. Konrath, Programa de Ps
Graduao em Cincias Farmacuticas,
Universidade Federal do Rio Grande do Sul,
Av. Ipiranga 2752, 90620-000 Porto Alegre
RS, Brazil.
E-mail: edukonrath@yahoo.com.br
Received February 28, 2013
Accepted May 12, 2013
doi: 10.1111/jphp.12090

Abstract
Objectives The inhibition of acetylcholinesterase (AChE), the key enzyme in the
breakdown of acetylcholine, is currently the main pharmacological strategy available for Alzheimers disease (AD). In this sense, many alkaloids isolated from
natural sources, such as physostigmine, have been long recognized as acetyl- and
butyrylcholinesterase (BChE) inhibitors. Since the approval of galantamine for the
treatment of AD patients, the search for new anticholinesterase alkaloids has escalated, leading to promising candidates such as huperzine A. This review aims to
summarize recent advances in current knowledge on alkaloids as AChE and BChE
inhibitors, highlighting structureactivity relationship (SAR) and docking studies.
Key findings Natural alkaloids belonging to the steroidal/triterpenoidal, quinolizidine, isoquinoline and indole classes, mainly distributed within Buxaceae,
Amaryllidaceae and Lycopodiaceae, are considered important sources of alkaloids
with anti-enzymatic properties. Investigations into the possible SARs for some
active compounds are based on molecular modelling studies, predicting the mode
of interaction of the molecules with amino acid residues in the active site of the
enzymes. Following this view, an increasing interest in achieving more potent and
effective analogues makes alkaloids good chemical templates for the development
of new cholinesterase inhibitors.
Summary The anticholinesterase activity of alkaloids, together with their structural diversity and physicochemical properties, makes them good candidate agents
for the treatment of AD.

Introduction
Alzheimers disease (AD) is a progressive age-related neurodegenerative disorder and is the most common form of
dementia among the elderly, generally diagnosed in individuals over the age of 65 years.[13] Considering the accelerated ageing of human society and the increase in life
expectancy worldwide, AD rates are predicted to increase
enormously, especially in developing regions. About 4.6
million new cases are added every year throughout the
world, and the World Health Organization (WHO) estimates that by 2040, 71% of the 81.1 million dementia cases
will occur in developing countries.[4,5]
The main pathological hallmarks of AD are the accumulation of amyloid plaques, or senile plaques, containing
extracellular deposits of b-amyloid peptide (Ab) and the
presence of intraneuronal neurofibrillary tangles (NFTs),
which result from hyperphosphorylated t-protein, both

producing neuronal cell loss in the nucleus basalis of


Meynert and in the hippocampus.[6,7] The most common
initial symptom of AD is deterioration of recent memory
with preservation of long term memory, leading to a progressive cognitive dysfunction, the main behavioural characteristic of AD.[8]
Although the exact pathogenesis of AD still remains to be
fully elucidated, it is currently considered to be a multifactorial disease. As such, different causes have been recognized
to be implicated in the course of AD, providing several
pharmacological strategies with multiple possible targets, all
of them under investigation.[9] Three main approaches have
been taken. The first involves the re-establishment of neurotransmitters levels, with the inhibition of cholinesterases,
acetylcholinesterase (AChE) and butyrylcholinesterase
(BChE), and also monoamine oxidase (MAO) enzymes.

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

1701

Anticholinesterase alkaloids for AD

Eduardo Luis Konrath et al.

The second concerns neuroprotection, wherein oxidative


stress is considered to be an early event in the pathological
cascade for the disease, thus suggesting the potential use of
antioxidants to limit the effects of free radicals on nerve
cells. The third approach deals with specific aspects related
to AD, including the decrease in the production or aggregation of Ab peptide, and inhibition of g- and b-secretase
enzymes, which play a critical role in the amyloidogenic
pathway, t-protein,[10,11] among others.
Though recent intensive efforts have been made to
understand the mechanism of neurodegeneration involved
in AD and to discover new drugs combating the symptoms,
at present there is a deficit in the number of efficient and
safe therapeutic agents to treat the disease. No new drugs
have been approved by the US Food and Drug Administration (FDA) since 2003, likely because the abnormal brain
deposits of Ab and t-proteins still cannot be considered
causes or by-products of the disease.[12] The few current
drugs available only address the symptoms of cognitive loss,
without delaying or modifying the disease progression. In
some cases, the medications only work for a limited time
and in some patients they do not offer relief at all. Another
point to consider is that there is no in-vivo model available
that is able to mimic all the cognitive, behavioural, biochemical and histopathological abnormalities observed in
the course of AD, making it difficult to investigate new beneficial agents.[13,14]
The underlying principle of this review is to afford an
outline on the current status of isolated alkaloids with anticholinesterase properties, the main strategy available for AD
treatment, summarizing the most recent work in this area.
Their potency of inhibition for both AChE and BChE
enzymes is also reviewed, with the description of SARs,
when available. This review covers the main alkaloid classes
containing active molecules, including steroidal and triterpenoidal, quinolizidine (focusing mainly the Lycopodium
alkaloids), isoquinoline, indole and miscellaneous alkaloids.

The neuropathology of AD
Histopathologically, AD is characterized by the presence of
extracellular deposits of Ab, known as amyloid or neuritic
plaques, and intracellular NFTs containing abnormally
hyperphosphorylated t-protein, both processes resulting in
marked atrophy of brain tissue.[15] The neuropathology of
the disease is complex, and much effort has been devoted to
elucidating the relationships among these main hallmarks
and the aetiology of AD.
The density of senile plaques in the cerebral cortex is significantly correlated with the degree of cognitive impairment observed in patients before death, although amyloid
deposits can also be found in healthy brain tissue.[13,16] The
hippocampus and the basal forebrain neurons are damaged
1702

early in the course of AD, especially affecting the cholinergic


neurons and also causing pronouncing alterations in other
neurotransmitter systems, such as serotonin and glutamate.
Interestingly, a potential link between the two pathological
hallmarks of AD, senile plaques and NFTs, was also
described, since Ab deposits may induce t-phosphorylation,
resulting in tangle formation and, finally, neuronal death.[17]
Moreover, overproduction of Ab is also able to decrease
t-protein solubility in some cell lines.[18]

The amyloid hypothesis


Senile plaques are a constant feature of AD and consist of a
central extracellular Ab peptide core surrounded by degenerative nerve endings.[19,20] Ab is a small peptide (3943
amino acids in length) generated from a much larger
protein, the amyloid precursor protein (APP), which is a
transmembrane glycoprotein, consisting of three major isoforms of about 700 amino acids with a large extracellular
region, and is widely expressed in the brain.[21]
The sequential cleavage of APP is a complex process,
divided into the a- and b-pathways and performed by three
types of enzymes called secretases.[22,23] The a-pathway
involves the sequential cleavage of APP by a-secretase and
g-secretase, producing soluble peptides of non-amyloid
nature, P3 and sAPP, the former playing an important
role in synaptic plasticity and protecting neurons against
excitotoxicity. However, the sequential proteolysis of APP
by b-secretase, followed by an additional cleavage by
g-secretase, generates the insoluble Ab, which is highly
fibrillogenic and readily able to be deposited in amyloid
plaques. Since amyloid fibrils are protease-resistant structures, their formation cannot be reversed once initiated and
they act as a nucleation site for further aggregation.[24] The
toxicity of Ab42, the main species formed through the
b-pathway, is due to its insoluble b-sheet conformation
preferentially formed instead of an a-helix.[25] Another
point to consider is that the expression and activity of the
b-secretase enzyme is known to be increased with increasing age,[10] and this tends to contribute to the comparatively
high incidence of dementia in the elderly.
Investigations have suggested that overproduction of Ab
results in neurotoxic effects in neurons, leading to synaptic
dysfunction, formation of NFTs and, eventually, neuronal
loss.[26] However, it is unclear whether the amyloid plaques
are the causal agent or merely a by-product of neuronal
death. The amyloid cascade hypothesis considers that Ab is
the initiating molecule in the pathological process observed
in AD, and that it disrupts calcium homeostasis leading to
the generation of free radicals, lipid peroxidation and
reduced synaptic integrity. Moreover, small amounts of
soluble Ab have also been shown to compromise cholinergic functions, independent of cell death.[27] Therefore, parts

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

Eduardo Luis Konrath et al.

of the neurotransmitter deficit observed in AD can be


attributed to this phenomenon rather than to cell death.
Another effect resulting from amyloid deposition is the activation of glial cells and the expression of inflammatory
mediators in microglia and astrocytes, which leads to a scenario of chronic neuroinflammation.[23]

t-Protein and neurofibrillary tangles


NFTs are bundles of abnormal fibres located within
neurons and consist of pairs of filaments wound into paired
helical filaments. These filaments are made of t-protein,
which is a normal neuronal microtubule-bound protein
involved in the assembly and stabilization of microtubules.
In AD, t-protein becomes hyperphosphorylated as a result
of an imbalance between kinase and phosphatase activity,
which normally tightly regulate its phosphorylation. In this
condition, t-protein dissociates itself from the microtubules, loses its ability to stabilize them and self-aggregates,
forming insoluble oligomers and eventually microscopic
tangles.[28] However, these tangles are not specific to AD,
also being found in other neurodegenerative diseases; also,
infrequent cases of tangle-poor AD have been described.[29]

Cholinergic hypothesis
There is considerable evidence to suggest a possible association between the memory deficits described in AD patients
and impairment of cholinergic neurotransmission in the
central nervous system (CNS), in an attempt to characterize the cognitive and behavioural abnormalities
observed.[8,26,30,31] The cholinergic system plays an essential
role in the processes of learning and memory, and the dysfunction of this whole process is consistent with the progressive severity of memory impairment observed in AD.[32]
Hence, the cholinergic hypothesis postulates that restoration of cholinergic neurotransmission is a helpful strategy
to enhance the availability of synaptic acetylcholine and
ameliorate impaired memory in AD patients.[33,34] The conceptualization of AD as a cholinergic deficiency syndrome
provides a useful outline to the search for new drugs but it
is also important to understand that cholinergic deficit is
not the only neurotransmitter dysfunction observed in AD,
since catecholaminergic, glutamatergic and serotonergic
systems are also involved.[35] However, the few medications
available, with the exception of the N-methyl d-aspartate
antagonist memantine, only address the symptoms of cognitive loss by targeting the AChE enzyme inhibition.
Acetylcholine released into the synaptic cleft is quickly
hydrolysed by AChE, and the blockade of this catabolic
process results in increased levels of the neurotransmitter,
which may partially correct the cholinergic deficiency seen
in AD. BChE, or pseudocholinesterase, plays only a minor
role in regulating the acetylcholine levels when compared

Anticholinesterase alkaloids for AD

with AChE,[36] but it does plays an important role in the


pathology of AD, since both enzymes are present in the
brain and are detected among NFTs and amyloid plaques.[37]
Various studies have indeed supported the hypothesis that
cholinesterase inhibitors can prevent Ab oligomerization,
thus displaying both anti-amyloid and neuroprotective
disease-modifying effect.[38] Moreover, in AD patients, the
level of AChE activity tends to decline and the activity of
BChE increases, with the ratio between BChE and AChE
changing from 0.6 in the normal brain to 11 in cortical
areas affected by the disease. In late AD stage, BChE represents the predominant cholinesterase in the brain, and for
this reason this enzyme is also targeted as an approach to
intercede the disease.[39,40]

The use of cholinesterase inhibitors


The currently approved treatment approach for AD focuses
on replacement therapy for deficits in central cholinergic
neurotransmission, with encouraging results obtained with
the use of anticholinesterases to amplify the physiological
action of acetylcholine in AD patients.[38,41,42] Three drugs
that target the cholinergic system are currently approved by
the FDA for AD: donepezil, rivastigmine and galantamine
(Figures 1 and 2).
Tacrine was the first medication approved by the FDA
and introduced in the market for the treatment of AD in

N
O

O
O

Donepezil

NH2

N
N

O
O

N
Rivastigmine
OH

Tacrine
NH2

N
Velnacrine
Figure 1
disease.

Some cholinesterase inhibitors used to treat Alzheimers

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

1703

Anticholinesterase alkaloids for AD

Eduardo Luis Konrath et al.

NH
2

10
10a

O
9

1
10b

D
6a

4
4a

11

OH
3

12

Galantamine

Physostigmine
H
N

NH2
Huperzine A
Figure 2 Some natural alkaloids used as cholinesterase inhibitors for
AD therapy.

1993, providing cognitive improvement in 540% of


patients with mild-to-moderate AD after treatment.[43,44]
However, gastrointestinal side effects and serious doserelated hepatotoxicity were reported, frequently preventing
dose escalation to 160 mg, the dosage required for
maximum proven efficacy on cognition.[45] A major
metabolite of tacrine, velnacrine, was also screened as a
cholinesterase inhibitor, although the molecule also demonstrated significant hepatotoxicity in vivo.[46]
In 1996, donepezil was cleared by the FDA for AD, and
was considered to have better properties than tacrine, with
lower toxicity and higher selectivity for AChE, and was also
able to increase the density of nicotinic receptors in the
brain. This piperidine derivative has a longer half-life than
tacrine, but has a more moderate efficacy than that produced by the maximum recommended dose of tacrine.
However, donepezil does not produce hepatotoxicity, and
has few side effects, all of them related to gastrointestinal
system.[47,48] Thereafter, the use of rivastigmine was
approved in 2000, being first marketed in Switzerland for
the treatment of AD. This physostigmine derivative, which
is administered twice daily, inhibits both cholinesterases
with a similar affinity, and it does not cause liver damage,
producing only cholinergic side effects such as nausea
and vomiting.[46,49] Galantamine, unlike the other anticholinesterases, is an alkaloid obtained from natural
sources, first isolated from Galanthus nivalis L and related
species belonging to the Amaryllidaceae family. This
product was first marketed in 2001 and is a long-acting,
relatively selective AChE inhibitor, with less BChE inhibitory activity and some gastrointestinal side effects
1704

described.[50,51] Moreover, galantamine also improves


cholinergic transmission by allosteric modulation of nicotinic receptors and has been used to treat the symptoms of
other forms of dementia.[52] It is currently isolated from
daffodil bulbs by the pharmaceutical industry, which has
engendered an exponential interest in the isolation and
characterization of alkaloids from Amaryllidaceae bulbs
(e.g. lycorine, homolycorine and hipeastrine), most of them
having anticholinesterase activity.[53]
The use of cholinesterase inhibitors appears to have beneficial impact on both behavioural and psychiatric symptoms, but there is no evidence for superiority of one agent
over another with respect to cognitive, behavioural or functional outcomes.[10,54] Several agents targeting AD are currently under clinical trials but failure rates are high, which is
an unfortunately common point for drugs that target the
brain. In this sense, researchers are currently focusing on
biomarkers to provide insight into disease progression,
hoping to find therapies that can be applied at parts of the
disease mechanism that occur at an early stage in the
brain.[12]

Alkaloids as anticholinesterase
inhibitors
A number of AChE and BChE inhibitors have been isolated
from various natural sources, as extensively described in the
literature.[5560] It is noteworthy that most of the compounds
have demonstrated only in-vitro activity; few of them have
been tested on animal models, which is imperative to prove
their capacity to cross the bloodbrain barrier and exert
beneficial effects in the brain.
Among these natural products, alkaloids are considered
to be the most promising candidates for use in the treatment of AD, due to their complex nitrogen-containing
structures.[61] In fact, one of the binding sites of AChE
involves the interaction of the positively-charged nitrogen,
even though other binding site exists, in order to allow the
inhibition by non-alkaloid compounds, mainly terpenes,
xanthones and coumarins.[56,57,62]
AChE inhibitors have been extensively studied as therapeutic options for AD after the initial observation that
physostigmine (eserine), an alkaloid obtained from the
seeds of Physostigma venenosum Balf. (Fabaceae), traditionally used as a ritual poison in Africa,[63] could reverse the
disruption of cognition produced by scopolamine in animal
models. This alkaloid has a rapid effect and although it acts
as a reversible inhibitor for both cholinesterases, it is more
selective for AChE. However, the use of physostigmine is
currently limited by its short half-life, its narrow therapeutic window and its gastrointestinal and orthostatic
effects.[64,65] Finally, molecular modifications of the alkaloidal structure of physostigmine have led to the development

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

Eduardo Luis Konrath et al.

of its analogue rivastigmine, a synthetic compound with a


better therapeutic and safety profile.
Other anticholinesterase alkaloids currently studied for
their use in AD include galantamine, as previously reported,
and huperzine A, a quinolizidine-type alkaloid isolated
from Huperzia serrata (Thunb) Trev, a Chinese species
traditionally used for the treatment of swelling, strains,
schizophrenia, myasthenia gravis and organophosphate poisoning.[66,67] Huperzine A has been found to be a potent,
reversible and selective AChE inhibitor, with oral bioavailability, able to cross the bloodbrain barrier and act with a
prolonged half-life.[68] Investigations conducted with this
alkaloid demonstrated it to exert a more potent effect in
increasing the cortical acetylcholine levels when compared
with either donepezil or rivastigmine and it also demonstrated higher in-vivo AChE inhibition.[69] Clinical trials at
phase IV, conducted in China, showed that huperzine A significantly improved memory shortages in elderly people
with benign senescent forgetfulness and in patients with
AD, with minimal peripheral cholinergic side effects and
toxicity.[70,71] However, recent phase II clinical trials conducted in the USA did not demonstrate any beneficial
cognitive effect of huperzine A in patients with mild-tomoderate AD, suggesting that further investigations must be
conducted to prove the efficacy of this compound.[72] In
this sense, Lycopodium alkaloids have become a focus of
research in recent years, resulting in many in-vitro and
in-vivo pharmacological studies aimed at cholinesterase
activity. However, all of the other Lycopodium alkaloids
identified so far have either shown no cholinesterase inhibitory activity or have possessed in-vitro activity significantly
lower than that of huperzine A.[73]
Many alkaloids that are active cholinesterase inhibitors
have already been described in different families, including
the isoquinoline-type alkaloids from Amaryllidaceae (especially found in Narcissus sp, Galanthus sp, Hyppeastrum
sp)[74,75] and Papaveraceae,[76,77] the steroidal alkaloids
from Buxaceae (genera Buxus and Sarcococca),[78] the
quinolizidine-type alkaloids from Lycopodiaceae (genera
Huperzia and Lycopodium)[7981] and the indole alkaloids
from Apocynaceae and Rubiaceae[82] among others.
Although the great interest in these products has inspired
developments in medicinal chemistry, leading to the synthesis of more potent synthetic analogues, reviews focusing
only on anticholinesterase alkaloids obtained from natural
sources are scarce in the literature.

Classes of anticholinesterase
alkaloids
Steroidal and triterpenoidal alkaloids
The active alkaloids belonging to this class are distributed in
species of Solanum, Veratrum and Fritillaria and in the Bux-

Anticholinesterase alkaloids for AD

aceae family, mainly in Buxus and Sarcococca genera, as


shown in Figure 3.[83] The Sarcococca genus is known for the
production of pregnane-type steroidal alkaloids. These
metabolites have mono- or dimethylamino substituents at
the C-3 and/or C-20 position in the basic steroidal skeleton.[84] Several Sarcococca steroidal alkaloids have been
reported to have anticholinesterase activity, with IC50
values in the range of 0.5-249 mm for AChE, (+)phulchowkiamide A being the most active, while for BChE
the IC50 varies from 0.3 to 200 mm, hookerianamide I possessing the lowest IC50 value (see Table S1).[8595] Through
LineweaverBurk and Dixon plots, Khalid et al.[84] demonstrated that the majority of the pregnane-type alkaloids
obtained from Sarcococca saligna have a pure noncompetitive type of inhibition for both cholinesterases.
Despite the similar activity range, this type of alkaloid is,
in general, more selective for BChE. The selectivity might be
explained by the larger size of the steroidal alkaloids, which
can easily diffuse into the larger aromatic gorge of BChE,
when compared with AChE. The residues of Phe-330 and
Tyr-121 in AChE form a bottleneck, making the access of
these compounds into the gorge difficult. Another important aspect that might also play a role in the selectivity of
these compounds is that hydrophobic interactions are more
prominent in BChE.[92] SAR studies demonstrated that the
amino nitrogens at C-3 and/or C-20 positions are the most
important features related to the potency of the compounds. At physiological pH, these nitrogens are expected
to be protonated and to have a similar behaviour to some
known quaternary and bisquaternary inhibitors.
Despite the higher selectivity for BChE, quantitative SAR
(QSAR) and docking studies of these Sarcococca alkaloids
focus on AChE. Through a 3D-QSAR study by comparative
molecular field analysis of 32 steroidal alkaloids obtained
from S. saligna, Zaheer-ul et al.[96] inferred that the presence
of negative density close to the substituent on the C-3
side chain and C-4 positions increase the AChE inhibitory
activity. On the other hand, negative density close to
C-2 decreases the inhibitory activity. For instance,
2b-hydroxyepipachysamine-D is almost three-fold less
active than epipachysamine-D, which does not have a
hydroxyl group at C-2. Furthermore, the presence of a
double bond between C-5 and C-6 also seems to increase
the activity, since saracocine, with a double bond in ring B,
is about two-fold more active than sarcodine, which does
not possess this unsaturation. Regarding the bulky groups,
their presence near the side chain at C-3, C-4, C-14 and
C-15 is not favourable for AChE inhibitory activity.
In a docking study using the crystal structure of Torpedo
californica AChE co-crystallized with decamethonium, a
series of pregnane-type steroidal alkaloids was evaluated,
demonstrating that all the 15 compounds adopted the same
binding mode. The ring A of the steroidal skeleton enters

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

1705

Anticholinesterase alkaloids for AD

Eduardo Luis Konrath et al.

NH

NH

H
H

O
H

H
N

NH

H
O
(+)Phulchowkiamide A

Hookerianamide I
21
20
18

11

19

HO

10

2
3

NH

H
6

12
13

17
16
14 15

H
O

NH

2-Hydroxyepipachysamine-D

Epipachysamine-D

Ac

H
H

H
N

H
Sarcodine

Saracocine

N
O
H

OH
O

MeO

N
H

NH
H

HO

OAc
OMe
O2 -Natafuranamine
Figure 3

1706

Buxakashmiramine

Steroidal and triterpenoidal alkaloids with anticholinesterase activity isolated from natural sources.
2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

Eduardo Luis Konrath et al.

Anticholinesterase alkaloids for AD

NH

NH

H
Buxamine C

Buxamine B

21
20
21

O
12

11

9
1

NH

10

2
4

13

14

18
17
15 16

8
7

19

H
(+)Buxabenzamidienine
(+)Buxamidine

27

25
26

N
22

18

H
19

10
5

14

20

16

21

17

HO

12

13

15

1
2
3

11
9

24
23

HO
H
O

Chuanbeinone

Ebeiedinone

21

N
18
19

11
9

10
5

3
4

H
6

12

H
8
7

20

H
17
16
15

13
14

Conessimin
Figure 3

Continued.

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

1707

Anticholinesterase alkaloids for AD

Eduardo Luis Konrath et al.

into the aromatic gorge and is placed into the bottom,


which might be due to the greater hydrophobicity of this
ring in comparison with the ring D. Also, the alkaloids are
buried inside the aromatic gorge, significantly contributing
to the stabilization of the complex, but these compounds
are not as deep into the aromatic gorge as the ligand.
Despite this, amino groups at C-3 and C-20 come close to
Tyr-331 and Phe-281, demonstrating a similar binding
position when superimposed with the co-crystal. Nevertheless, electrostatic interactions do not contribute significantly
to the binding energy, since the most important contributions are guaranteed by van der Waals interactions. The
binding of the alkaloid with the enzyme does not cause
major structural rearrangements, although the dynamics of
the gorge are considerably changed, since its flexibility is
reduced.[97]
Another well-studied genus from the Buxaceae family is
Buxus, considered to be a rich source of cycloartenol-type
triterpenoidal alkaloids. As in the pregnane-type alkaloids
obtained from Sarcococca, nitrogens atoms are also present
at C-3 or/and C-20,[98] and for this reason they are also recognized as a source of anti-AChE and -BChE compounds.
The IC50 values range from 3.0 (O2-natafuranamine) to
468 mm for AChE, while for BChE the values range from
0.74 (buxakashmiramine) to 350 mm.[99104] Also, Khalid
et al.[105] demonstrated that buxamine B and buxamine C
have a non-competitive inhibitory profile for AChE, as verified by LineweaverBurk plots.
Orhan et al.[106] reported the specificity of (+)buxabenzamidienine (10-fold) and (+)-buxamidine (more
than 320-fold) for AChE inhibition. However, Atta-urRahman et al.[101] and Choudhary et al.[100,102] observed that
usually the triterpenoidal alkaloids were more selective for
BChE. Some docking studies performed with Torpedo californica AChE and Buxus alkaloids demonstrated that, as
observed for the steroidal alkaloids, the C-3 and C-20
amino groups are essential for the activity, and that the
hydrophobicity of the compounds is also related to the
inhibitory capacity of the ligands.[105,106] Corroborating these
results, Choudhary et al.,[102] through SAR studies of nine
different triterpenoidal alkaloids, demonstrated that the
presence of Me2N moieties at C-3 and C-20 may be the
most important features for the anticholinesterase activity.
Moreover, the presence of substituents at C-16 and at C-31
decreases the inhibitory effect. On the other hand, unsaturated sites do not seem to influence the activity, since the
aromatic gorge of the enzymes are extremely flexible, facilitating the accommodation of different conformations of the
alkaloids.
Some Fritillaria species have also been studied due to the
anticholinesterase activity of their steroidal alkaloids.
Usually, these compounds possess a C27 cholestane carbon
skeleton, with five or six carbocyclic or heterocyclic
1708

rings.[107] As for the other steroidal alkaloids, the Fritillaria


alkaloids are more selective for BChE, with IC50 values
ranging from 0.7 to 121 mm, chuanbeinone being the most
active. For AChE, the IC50 varies from 5.7 (for ebeiedinone)
to 352 mm.[108,109] In a SAR analysis of a series of these steroidal alkaloids, it was observed that the potency is increased
by the presence of functional groups at C-3 and by the C-6keto function. In contrast, hydroxyl substitution at C-20
and methyl substitution at the N position are responsible
for decrease in the anticholinesterase activity.[108]
Yang et al.[110] isolated five different steroidal alkaloids,
sharing a fused-pyrrolidine ring at the preganane skeleton,
from the species Holarrhena antidysenterica and evaluated
their capacity to inhibit AChE, with IC50 ranging from 4 to
28 mm. The most active compound, conessimin, demonstrated a reversible and non-competitive mode of inhibition. The pyrrolidine moiety of the alkaloid forms a
hydrogen bound with Tyr-72. Through a SAR analysis of
the five alkaloids, it was observed that the N-Me substituent
of the pyrrolidine moiety seems to decrease the activity,
while the presence of the same substituents at C-3 increases
the anti-AChE effect.

Quinolizidine-type/Lycopodium alkaloids
Lycopodium alkaloids are quinolizine, pyridine or
a-pyridone alkaloids, which may be divided into four different classes: lycopodine, lycodine or flabellidine, fawcettimine and miscellaneous. The most prominent compound
belonging to this group, huperzine A, is a lycodine-type
alkaloid and was first isolated from the Chinese lycopod
Huperzia serrata (Thunb. Ex Murray); it is responsible for
the exponential increase in interest on investigations of
Lycopodiaceae species (Figure 4), as previously cited.[73]
Halldorsdottir et al.[111] isolated ten known lycopodane alkaloids from Lycopodium annotinum ssp. alpestre collected in
Iceland, and evaluated their in-vitro anticholinesterase
activity. As a result, the most potent inhibitors of AChE
were anhydrolycodoline (IC50 = 191 mm), and lycofoline
(IC50 = 600 mm), while for BChE only lycodoline could
inhibit the enzyme activity, with a very high IC50 value
(667 mm) (Table S2). In an attempt to understand the
reason behind the low anticholinesterase activity of the
lycopodane alkaloids (some of them have an IC50 value
higher than 2000 mm), investigations using docking studies
were made using Torpedo californica AChE co-crystallized
with huperzine B. Comparing the orientations assumed
by anhydrolycodoline and by huperzine B, the authors
observed that both were completely different, since the carbonyl groups of the two molecules were pointing in opposite directions. Also, the nitrogen of anhydrolycodoline was
shifted about 3.0 from the position of the nitrogens of
huperzine B, and no stabilizing hydrogen bonds were

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

Eduardo Luis Konrath et al.

Anticholinesterase alkaloids for AD

OH

HO

OH

Anhydrolycodoline

Lycofolin

Lycodoline

HO
O

N
H

NH

H2N
N
H

Cryptadine B

Carinatumin A

OH

OH

O
O

H
H

NH

HN

NH

OH

O
Lycoparin A

Cernuine

Sieboldine A

HO

Lycoparin B

OH

Lycocernuine

H
H

O
Acetyldihydrolycopodine
Figure 4

Lycopodine

Quinolizidine/Lycopodium alkaloids with anticholinesterase activity isolated from natural sources.

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

1709

Anticholinesterase alkaloids for AD

Eduardo Luis Konrath et al.

detected. Finally, the authors concluded that the lycopodane


alkaloids fit into the binding site of AChE, although they
are not able to form strong interactions with the amino
acids of this site, thus explaining their high inhibitory
concentrations.
Some miscellaneous Lycopodium alkaloids were also
evaluated due to their capacity to inhibit AChE. The most
active, cryptadine B (IC50 = 18.5 mm) isolated from L. cryptomerinum, consists of two octahydroquinoline rings and a
piperidine ring.[112] However, other alkaloids with the same
C27N3 pentacyclic skeleton inhibited the enzyme activity
with a higher IC50 value (>60 mm).[112,113] On the other
hand, lycodine alkaloids, in the class of huperzine A, as
expected were much more active (e.g. carinatumin A, isolated from L. carinatum, exhibited an IC50 value of
5.6 mm).[114] Even so, some other alkaloids of the same
class are considered inactive (IC50 > 200 mm), such as
lycoparins A and B.[115] Interestingly, one of the most
active Lycopodium alkaloids, with an IC50 value of 2.0 mm,
is sieboldine A, a fawcettimine-derived compound with
a fused tetracyclic ring system, consisting of an azacyclononane, a cyclohexanone, a cyclopentanone and a tetrahydrofuran ring.[116]
Recently, Konrath et al.[81] investigated some Lycopodiaceae species belonging to Huperzia and Lycopodiella
genera collected in Brazil, which resulted in interesting
profiles of anticholinesterase inhibition for the alkaloid
extracts. Some compounds isolated from these species were
also screened in vitro, but the results showed a lower
potency of inhibition when compared with the IC50 values
from the alkaloidal extracts, suggesting a synergism among
the compounds. For example, Lycopodiella cernua extract
showed an IC50 value of 42.6 mm, while its main alkaloids
cernuine and lycocernuine possessed IC50 values of 32.7 and
>250 mm, respectively. Moreover, acetyldihydrolycopodine,
one of the main alkaloids found in L. clavatum and L. thyoides collected in Brazil had an IC50 value of 84.7 mm, while
lycopodine had a much lower anticholinesterase activity,
suggesting that the acetyl moiety may have a beneficial
influence on enzyme inhibition.[117]

Isoquinoline-type alkaloids
Amaryllidaceae alkaloids
The Amaryllidaceae alkaloids represent a large group of isoquinoline alkaloids, the majority of which are known to
occur exclusively in this family. These alkaloids can be classified into nine skeleton types, for which the representative
ones are: norbelladine, lycorine, homolycorine, crinine, haemanthamine, narciclasine, tazettine, montanine and galantamine.[118] Regarding AChE inhibition, the most active
compounds, displaying IC50 values close to 10-6 m, belong to
the galantamine type (Table S3). In a study evaluating 23
1710

Amaryllidaceae alkaloids belonging to the lycorine, homolycorine, haemanthamine, galantamine, tazettine and miscellaneous types, Lopz et al.[74] demonstrated that sanguinine,
galantamine, 11a-hydroxygalantamine, and epinorgalantamine inhibited AChE from the electric eel, Electrophorus
electricus (EeAChE), with IC50 values varying from 0.10 to
9.60 mm. The authors suggest that the AChE inhibitory
effects seem to be related to structural characteristics
among the different skeleton types, as only the alkaloids
included in the galantamine and lycorine (assoanine,
oxoassoanine, pseudolycorine) groups exhibited such
inhibitory activity. The higher potency of hydroxygalantamine for AChE inhibition, in comparison with galantamine, may be due to the presence of an additional
hydroxyl group in sanguinine, which could aid in the stabilization of the ligand in the enzyme binding pocket by classical and non-classical hydrogen bonds.[74,119] Chlidanthine,
a positional isomer of galantamine, is about 10-fold less
potent for AChE inhibition (EeAChE IC50 = 24.1 mm).[120]
This difference in activity could be assigned to the stereochemistry of chlidanthine, which hinders the establishment of intramolecular hydrogen bonds between the
hydroxyl group at C-3 and the oxygen atom of the dihydrofuran ring, such as occurs in galantamine ring.[120,121]
Among the other subclasses of Amaryllidaceae alkaloids,
the most potent cholinesterase inhibitors seem to be those
compounds belonging to the lycorine type. Assoanine
(EeAChE IC50 = 3.87 mm), oxoassonine (EeAChE IC50 =
47.2 mm)[74] and 1-O-acetyllycorine (IC50 = 0.96 mm)[53] are
the most potent AChE inhibitors in this group, their
potency being attributed to (i) the presence of an aromatic
ring C, for assoanine and oxoassonine, and (ii) the acetoxy
and hydroxyl substituents attached to C-1 and C-2 of 1-Oacetyllycorine. Ungeremine, a quaternary lycorine-type
alkaloid with the ring C aromatic, inhibited EeAChE displaying an IC50 of 0.35 mm.[122] The higher potency of this
compound for EeAChE can be assigned to the presence of
the ring C aromatic together with the quaternary nitrogen
atom, which is important for the inhibitory activity displayed by other isoquinoline alkaloids, such as berberine.
Furthermore, the methyl and epoxy groups present in
incartine (EeAChE IC50 = 107 mm) and incartine N-oxide
(EeAChEIC50 = 34.5 mm) also favour AChE inhibition.[123]
Amaryllidaceae alkaloids possessing the other skeleton seem
to behave as weak cholinesterase inhibitors with IC50 values
close to 10-4 to 10-3 m.[53,124,125]
Other isoquinoline alkaloids
The activity of protoberberine alkaloids on cholinesterases
has been evaluated by numerous authors, mainly regarding
the quaternary compounds berberine and palmatine, as
seen in Figure 5. Ingkaninan et al.,[126] in a study with proto-

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

Eduardo Luis Konrath et al.

Anticholinesterase alkaloids for AD

OH

OH

O
O

HO

OH
N

11-Hydroxygalantamine

Sanguinine

2
3

OH
O

10

O
9

11

10a

6a
7

NH

12
6

Assoanine

Epinorgalantamine

OH
HO

O
H
HO

N
O

H
N

Pseudolycorine

Oxoassoanine

OAc

OH

AcO

O
O
H

1-O-Acetyllycorine

Chlidanthine
Figure 5

Isoquinoline-type alkaloids with anticholinesterase activity isolated from natural sources.

berberine alkaloids from Stephania venosa, verified that


the quaternary compounds stepharanine (EeAChE IC50 =
14.1 mm), cyclanoline (EeAChE IC50 = 9.23 mm) and Nmethyl stepholidine (EeAChE IC50 = 31.3 mm) were about

five-fold more potent for EeAChE inhibition than the


tertiary compounds isolated from the same plant
(IC50 > 100 mm). Additionally, the effects of the alkaloids
from S. venosa on EeAChE were compared with those

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

1711

Anticholinesterase alkaloids for AD

Eduardo Luis Konrath et al.

OH
N+

O
O

N+

O
Ungeremine

Berberine

O
HO

HO
H

N+

O
Incartine

Incartine N-oxide

O
+

N+
HO

O
O
OH
Stepharanine

Palmatine

OH
O

O
H

OH
H
HO

N+

O
Cyclanoline
Figure 5

1712

N+
HO

N-methyl stepholidine

Continued.

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

Eduardo Luis Konrath et al.

Anticholinesterase alkaloids for AD

HO
O

O
Jatrorrhizine

O
Chelerythrine

HO

N
H
NO2

OH

HO

NO2

NO2
O

()2,9-Dihydroxyl-3,11-Dimethoxy1,10-Dinitrotetrahydroprotoberberine

(+)Nitroapocavidine

O
O

O
O

Sanguinarine

Dehydrocavidine
O
+

HO

Dehydrocorydaline
Figure 5

Pseudocolumbamine

Continued.

displayed by the well-known quaternary alkaloids berberine


(EeAChE IC50 = 0.58 mm), palmatine (EeAChE IC50 =
0.26 mm) and jatrorrhizine (IC50 = 0.93 mm). Besides
EeAChE, berberine and palmatine were also tested in
human, mice and rat enzymes displaying about the same

potency.[127132] Regarding BChE, both compounds inhibited


equine serum enzymes, displaying IC50 values corresponding to 3.44 and 6.84 mm, respectively.[132]
Berberine has gained considerable attention because of
its wide spectrum of biochemical and pharmacological

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

1713

Anticholinesterase alkaloids for AD

Eduardo Luis Konrath et al.

O
O

O
N+

N+
O
O

OH

Coptisine

Pseudopalmatine

O
O

OH

O
H

Corydaline

Bulbocapnine

OH

O
HO
O
O

Corydine

2-Hydroxy-9-Methoxyaporphine

O
HN

H
OH

HO

O
O

(R,S)-2-N-Norberbamunine
O

O
N

H
O
O

O
HO

H
N

H
N

HO

(R,R)-Isochondodendrine
Figure 5

1714

(S-S)-O4-Methyl,O6-Demethyl(+)-Curine

Continued.

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

Eduardo Luis Konrath et al.

potentials. Its effects on AChE, BChE, MAO and other key


enzymes have been reported.[133,134] Additionally, berberine
can reduce Ab levels by altering APP processing in human
neuroglioma H4 cells, which stably express Swedish-type
APP in concentrations ranging from 0.1 to 100 mm.[135]
Molecular modelling studies have revealed that this molecule interacts with eight hydrophobic residues in AChE
and with six hydrophic residues in BChE. Also, there is the
possibility of a hydrogen bond between berberine and Tyr121 of AChE.[134]
The benzophenanthridine alkaloid chelerythrine has
been shown to inhibit AChE (from Electrophorus electricus
and humans) and BChE (from equine serum and humans)
in an equipotent way, with IC50 values ranging from 1.54
to 10.3 mm.[128] Kinetic studies indicated that it acts as a
mixed inhibitor of EeAChE and human AChE (hAChE)
with a competitive behaviour. Considering this mode of
inhibition, Brunhofer et al.[128] suggested that chelerythrine
could bind to the peripheral anionic site (PAS). Docking
simulations indicated that this molecule is able to bind in
the Torpedo californica AChE (TcAChE) (PDB ID 1FSS)
active site establishing a hydrogen bond with Tyr-130 and
p-stacking interactions with Tyr-121 and Tyr-334 PAS residues. This binding mode represents an interesting feature,
since it is possible to assume an additional capacity to
target the AChE-induced Ab aggregation process. Further
experiments demonstrated that chelerythrine at 5, 10 and
100 mm is able to inhibit the AChE-induced Ab fibrillogenesis in a concentration-dependent way reaching almost
90% of inhibition at the highest concentration.[128]
Beyond the previously cited berberine and palmatine,
another ten isoquinoline alkaloids from Corydalis saxicola
were tested on AChE.[127] The most active compounds, displaying IC50 values between 1.70 and 9.92 mm, were (-)-2,9dihydroxyl-3,11-dimethoxy-1,10-dinitrotetrahydroprotober
berine, (+)-nitroapocavidine, dehydrocavidine and sanguinarine. SAR studies indicated that all of the tested compounds were active at similar concentrations (IC50 values
close to 10 mm). AChE inhibition displaying similar IC50
values (110 mm) was also observed for other protoberberine alkaloids, such as jatrorrhizine, dehydrocorydaline,
pseudocolumbamine, pseudopalmatine
and
coptisine.[132,136,137] The activity-guided fractionation of Corydalis
cava led to the identification of three benzylisoquinoline
alkaloids inhibiting EeAChE and equine BChE (eqBChE):
bulbocapnine (EeAChE IC50 = 40 mm; eqBChE IC50 =
83 mm); corydaline (EeAChE IC50 = 40 mm; eqBChE
IC50 = 83 mm); corydine (EeAChE IC50 = 15 mm; eqBChE
IC50 > 100 mm).[138] These differences in potency and selectivity may be explained by the skeleton of the evaluated
alkaloids. Corydaline, which is most potent and selective for
EeAChE inhibition, is a protoberberine alkaloid, while the
other compounds possess the aporphine skeleton.[138]

Anticholinesterase alkaloids for AD

Aporphine alkaloids from species belonging to Beilschmiedia displayed a narrow range of AChE inhibitory
activity with IC50 values in the range of 10-5 to 10-6 m,[139]
comparable with the reference compound huperzine A.
2-Hydroxy-9-methoxyaporphine, an alkaloid obtained
from B. alloiophylla, inhibited AChE activity with an IC50
in the region of 2.0 mm. On the other hand, compounds
possessing the aporphine skeleton isolated from Corydalis
turtschaninovii demonstrated lower potency, with
IC50 > 25 mm.[130]
The
bisbenzylisoquinoline
alkaloids
(R,S)-2-Nnorberbamunine, (R,R)-isochondodendrine and (S,S)-O4methyl,O6-demethyl-(+)-curine, isolated from Abuta
grandifolia (Menispermaceae), were able to inhibit AChE
from mouse brain and BChE from mouse serum, showing a
slight selectivity for the serum cholinesterase (about fivefold), with IC50 values ranging from 34.7 to 78.2 mm for
mouse AChE (mAChE) inhibition, and from 1.00 to
9.46 mm for mouse BChE (mBChE) inhibition.[140] This
inhibition profile is in agreement with that found for
other bisbenzylisoquinoline alkaloids from Cocculus
pendulus, another species belonging to the Menispermaceae
family.[141]

Indole alkaloids
Monoterpene indole alkaloids
The indole alkaloids nitrarine, hirsutine, rauwolscine and
catharanthine (Figure 6) display BChE selective inhibition,
with no significant difference between eqBChE and hBChE.
The IC50 values for eqBChE inhibition range from 4.97 to
10.6 mm, while the IC50 values for hBChE inhibition ranged
from 1.97 to 13.7 mm.[128] These results suggest that the
monoterpene side chain fused to the indole ring does not
critically influence the potential for BChE inhibition. In
agreement with these results, Passos et al.[142] verified that
the monoterpene indole alkaloids (MIAs) angustine, vallesiachotamine lactone and E/Z-vallesiachotamine also selectively inhibit eqBChE with IC50 values ranging from 3.47 to
14 mm (Table S3). Docking simulations indicated that vallesiachotamine lactone and E/Z-vallesiachotamine bind to the
BChE active site maintaining the same orientation of their
scaffold: the indole ring faces to Ser-198 and His-438 residues of the enzyme catalytic site, enabling the establishment
of hydrogen bonds. Moreover, the Trp-82, Trp-231, Leu286, Phe-329 and Ile-442 residues aid in stabilizing the
complexes through van der Waals contacts with the carbon
atoms characterizing the ring systems. Angustine, on the
other hand, binds to the BChE active site in a different orientation than vallesiachotamine structures. The complex
proteinligand seems to be stabilized mainly by hydrophobic interactions involving Trp-82, Trp-231, Leu-286 and
Phe-329 and the aromatic moieties of the molecule.[142]

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

1715

Anticholinesterase alkaloids for AD

Eduardo Luis Konrath et al.

N
H

N
H

N
H

O
O

Nitrarine

Hirsutine
H

N
N
H

N
H

H
H
O

N
H

OH

O O

Catharanthine

Rauwolscine
O

N
N
H

N
H

H
O

Angustine

Vallesiachotamine lactone

N H
H

N
N
H

E-vallesiachotamine

Z-vallesiachotamine
N+

N+
N
H

N
O

Fascaplysin

Infractopicrin
N+

HO

H
N+

N
N
H

10-Hydroxy-infractopicrin
H
N+
N
H

1716

Prunifoleine

O
N+

Cl
N
H

14-Oxoprunifoleine
Figure 6

Nostocarboline

Indole alkaloids with anticholinesterase activity isolated from natural sources.


2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

Eduardo Luis Konrath et al.

Quaternary b-carboline alkaloids


b-Carboline
alkaloids,
mainly
the
N3-alkylated
b-carbolinium salts, are well-known cholinesterase inhibitors. The marine compound fascaplysin was evaluated using
EeAChE and eqBChE and displayed IC50 values of 1.49 and
90.5 mm, respectively.[143] This inhibition profile suggests a
60-fold selectivity for AChE inhibition. In addition, kinetic
experiments demonstrated that fascaplysin acts as a
non-competitive EeAChE (Ki = 2.28 mm) and eqBChE
(Ki = 48.5 mm) inhibitor, indicating that this ligand binds
with high affinity to a site different from the catalytic
one.[143] Further docking studies revealed details regarding
the binding mode of this compound in AChE and BChE
active sites. Fascaplysin, which possesses a planar structure,
seems to occupy the AChE active site near to the gorge
entrance. The main interactions between the molecule and
AChE are pp interactions established between the terminal
aromatic ring of fascaplysin and the Trp-279 of the PAS.
Moreover, the carbonyl and NH groups may establish
hydrogen bonds with water molecules, which are conserved
in the enzyme active site. However, no direct hydrogen
bonds were observed to occur between ligand and amino
acids residues of the enzyme. Furthermore, hydrophobic
interactions can be observed between fascaplysin and the
residues Phe-330, Phe-331, Tyr-334 and Tyr-121.[143]
Two quaternary b-carboline alkaloids isolated from
the toadstool Cortinarius infractus, infractopicrin and
10-hydroxy-infractopicrin, were shown to selectively inhibit
bovine erythrocytes AChE (bAChE, IC50 of 9.72 and
12.7 mm, respectively). Neither compound showed significant inhibition on eqBChE when tested at 100 mm.[144] Based
on the chemical features of these two compounds, together
with findings of Schott et al.,[145] the authors assign the
AChE selectivity displayed by infractopicrin and 10hydroxy-infractopicrin to the indole N-alkylation (N1alkylation). Complementary docking studies were carried
out to investigate the selectivity for AChE inhibition. The
results revealed that both compounds may establish pp
interactions with the aromatic residues Trp-84 and Phe330. Furthermore, the carboxamide oxygen of the ligands is
connected with the hydroxyl oxigen of Tyr-121 through a
hydrogen bond, fixing their positions. On the contrary it
has been verified for AChE that the docking solution for
infractopicrin and 10-hydroxy-infractopicrin in BChE
shows no pp interactions between ligands and aromatic
residues and the lack of hydrogen bonds with Tyr-121.[144]
Prunifoleine and 14-oxoprunifoleine, two quaternary
b-carboline alkaloids found in Psychotria prunifolia, are able
to inhibit EeAChE with IC50 values of 10.0 and 3.39 mm,
respectively. Additionally, both alkaloids also inhibit
eqBChE with IC50 values of 100 and 11.0 mm, respectively.[142] As well as described for other quaternary

Anticholinesterase alkaloids for AD

b-carbolines, they act as non-competitive inhibitors for


EeAChE (Ki of 15.8 and 8.93 mm, respectively). Further
docking experiments indicated that both molecules bind to
the TcAChE active site in the same orientation, insofar as
the endocyclic oxygen of both ligands is involved in a
hydrogen bond with the hydroxyl group of Tyr-121 from
the PAS.[142] Hydrophobic interactions occur between
ligands and Phe-290, Phe-330 and Phe-331 residues.
Moreover, stacking interactions between Trp-84 and the terminal aromatic ring of the two compounds are also established. These interactions are similar to those described in
earlier sections for Cortinarius infractus alkaloids[144] and for
some synthetic quaternary b-carbolines.[146] Docking simulations of prunifoleine and 14-oxoprunifoleine in hBChE
revealed that these compounds bind to the enzyme with different orientation, which can help to explain their different
potencies for eqBChE inhibition. In addition to hydrophobic interactions, prunifoleine, which is about 10-fold more
potent than 14-oxoprunifoleine, may establish polar contacts with Ser-198 and His-438 residues of the hBChE catalytic site.[142]
The cyanobacterial alkaloid nostocarboline is a nonselective AChE and BChE inhibitor displaying IC50 corresponding to 5.3 and 13.0 mm for EeAChE and eqBChE
inhibition, respectively.[147,148] The mode of inhibition, demonstrated by kinetic experiments, was non-competitive.[148]

Miscellaneous
Several other alkaloids have demonstrated anticholinesterase activity in addition to those mentioned above
(Figure 7). However, as they have a reduced number of representatives of their classes, these compounds have been
grouped together as miscellaneous alkaloids (Table S4).
Juliflorine is a piperidinium alkaloid isolated from the
leaves of Prosopis juliflora, and consists of two piperidines
connected with a dihydroindilizine moiety through two
aliphatic chains. This compound inhibited AChE with an
IC50 of 0.42 mm and BChE with an IC50 of 0.12 mm. Through
analysis by LineweaverBurk and Dixon plots, a noncompetitive type of inhibition was indicated. Docking
studies performed with TcAChE, suggested that C/D rings
enter deeper into the aromatic gorge, probably due to their
hydrophobicity, and that the rings A and B remain at the
top. The alkaloid is able to span the entire aromatic gorge,
with multiple binding sites and several intermolecular interactions identified, mainly hydrogen bonding, hydrophobic
contacts, pp interactions and hydrophilichydrophobic
interactions.[149] Some other piperidine alkaloids also inhibited AChE activity, although with lower potencies.[150]
From the Thai marine sponge Petrosia n. sp., petrosamine, a pyridoacridine alkaloid was isolated. This compound exists in both enolate and keto forms and showed a

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

1717

Anticholinesterase alkaloids for AD

Eduardo Luis Konrath et al.

HO
B
N
H

HO
C
A

D
N

N
H
Juliflorine
Br

Br

2 3
4a

12b

11
N 9

8a
8

12c

4b

7a

OH

O
5
6

N+

14

13

12a
12

15

16

N+
O

Petrosamine

OH
N

N
N

N
Peganole
Figure 7

HO
Vasicine

Miscellaneous alkaloids with anticholinesterase activity isolated from natural sources.

potent inhibitory profile for AChE, with an IC50 of


0.091 mm. To understand the binding mode of petrosamine
to AChE, a docking study using TcAChE complexed with
galantamine and with donepezil was performed. It was
observed that the quaternary ammonium atom at ring D is
responsible for the major and strong interactions, mainly
pp interactions with Trp-84 and Phe-330 residues, and
by a salt bridge with Glu-199. Also, Ser-200 and His-440
residues from the catalytic site interact directly with
petrosamine.[151]
Three quinazoline alkaloids evaluated in a study by Brunhofer et al.[128] were shown to be selective BChE inhibitors.
1718

Desoxypeganine

Among them, peganole seems to be more selective for


eqBChE (IC50 = 11.4 mm) than for hBChE (IC50 > 20 mm).
On the other hand, vasicine showed similar potencies for
eqBChE (IC50 = 2.53 mm) and hBChE (IC50 = 3.13 mm) inhibition. Finally, unlike the other evaluated quinazoline alkaloids, desoxypeganine was a non-selective inhibitor, being
equipotent for eqBChE (IC50 = 11.8 mm) and eqAChE
(IC50 = 11.9 mm). The quinazoline scaffold has been used as
a model for the synthesis of hybrid compounds, which are
able to inhibit BChE in a range of nanomolar concentration, being about 80-fold more potent for serum cholinesterase inhibition than for AChE.[128]

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

Eduardo Luis Konrath et al.

Some other alkaloids with different skeletons, such as


leptomerine (IC50 = 2.5 mm)[152] and 8-demethoxy-10-Omethylhostasine (IC50 = 2.32 mm),[153] also may serve as good
scaffolds for AD treatment due to their high potencies.

Final remarks
Among the different alkaloid classes, most of the members
may be considered interesting anticholinesterase compounds. Steroidal and triterpenoidal alkaloids have, in
general, a slightly more selective potential as BChE inhibitors. Some of them exhibit an IC50 value lower than 1 mm
(e.g. buxakashmiramine, chonemorphine and chuanbeinone), all of them representing good scaffolds for the development of new BChE inhibitors. Some indole alkaloids,
such as angustine, vallesiachotamine and deoxyvobtusine,
are also more selective for BChE. However, several alkaloids
from other classes have only been evaluated for AChE activity, and when their inhibitory potential for BChE was
assayed, less selectivity was achieved. Although the role of
AChE in improving memory and cognition in patients with
AD is well established, the search for inhibitors of BChE,
acting in the CNS, should not be neglected, since this
enzyme is also responsible for inactivating acetylcholine in
the brain tissue, and its expression is increased in patients
suffering from AD.[154] Therefore, the evaluation of the
effects displayed by alkaloids on both cholinesterases could
constitute a valuable strategy for discovering new templates
for brain selective AChE/BChE inhibitors possessing less
undesired peripheral effects.
Considering the quinolizidine-type/Lycopodium alkaloids, except for huperzine A and B and sieboldine A, most
of the compounds from this class do not behave as potent
AChE inhibitors. Some of them, such as acrifoline, annotine
and annotinine, exhibit an IC50 value higher than 1000 mm,
which is a hindrance to their applicability as a source of
promising templates for cholinesterases inhibitors. On the
other hand, isoquinoline alkaloids remain good scaffolds
mainly as AChE inhibitors. The fact that the alkaloid galantamine, obtained from plants belonging to the Amaryllidaceae family, is one of the AChE inhibitors approved for
the treatment of AD has motivated the search of different
species within this family, aiming to identify other potent
cholinesterase inhibitors. Among these compounds, 1-Oacetyllycorine and sanguinine are potent AChE inhibitors
with IC50 values of 0.96 and 0.10 mm, respectively. Despite
the approximately 10-fold lower IC50 for AChE inhibition in
comparison with the value reported for galantamine, the
low natural abundance of sanguinine has precluded its
further development. Regarding the bioavailability, the
study of the interactions of Amaryllidaceae alkaloids with
the bloodbrain barrier efflux transporter P-glycoprotein
(P-gp)[155] indicated little or no interaction with P-gp for the

Anticholinesterase alkaloids for AD

nine tested compounds, including galantamine. Moreover,


Amaryllidaceae alkaloids from the crinane and galantamine
groups did not show inhibitory activity against cytochrome
P450 3A4 (CYP3A4). The CYP3A subfamily comprises
about 30% of the total liver cytochrome P450 enzyme pool
in humans, and the isoenzyme CYP3A4 accounts for
approximately 60% of drugs metabolized. In the lycorane
series, only narciclasine inhibited CYP3A4 at low micromolar concentration.[156]
A number of the most active isoquinoline compounds
are quaternary alkaloids, such as berberine, palmatine, chelerythrine, coptsine and dehydrocorydaline. As such, these
compounds could have difficulty in crossing the blood
brain barrier under physiological conditions in a salt form.
Moreover, their oral bioavailability is usually poor, due to
their physicochemical properties.
It is worth noting that most of these studies only demonstrate the in-vitro activity of the isolated alkaloids, and it is
well known that other aspects, mainly pharmacokinetics
and toxicity, are related to the clinical potential of bioactive
molecules. Therefore, studies aiming to evaluate these
promising alkaloids in cell cultures and in in-vivo models
are indispensable. All these efforts should culminate in the
identification of good scaffolds and the subsequent development of more effective and safer anticholinesterase
drugs.

Conclusions and future perspectives


AD has a multifactorial pathology and although many
efforts are currently being devoted to achieving new
therapies aimed at reducing the amyloid and tauopathy
processes, the main therapeutic approach approved for
symptomatic treatment of AD is centred around the reduction of cholinergic deficit by the use of cholinesterase
inhibitors. Among these, the majority of studies have
focused on alkaloids, such as the prototype inhibitor of
AChE, physostigmine, obtained from Physostigma venenosum, which inspired the synthesis of the analogue rivastigmine. Another alkaloid currently marketed to treat AD
symptoms is galantamine, isolated from bulbs of several
Amaryllidaceae species. The major classes of molecules
reported to have significant anti-enzymatic activity are the
steroidal/triterpenoidal (most of them selectively inhibiting
BChE), quinolizidine (with focus on huperzine A, obtained
from the Chinese lycopod Huperzia serrata, currently
undergoing clinical trials), isoquinoline (especially galantamine type, protoberberine, aporphine and bisbenzylisoquinoline classes) and indole alkaloids, as summarized in
Table 1. Several active compounds have been discovered and
further investigated through molecular modelling studies,
to verify the sites of interaction between the chemical scaffold and the enzymatic amino acid residues, resulting in the

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

1719

Anticholinesterase alkaloids for AD

Eduardo Luis Konrath et al.

Table 1 Main classes of alkaloids reported to have anticholinesterase activity, including their main enzymatic selectivity, mode of inhibition and the
IC50 range of concentrations for most of the representatives belonging to each class
Alkaloid class

AChE/BChE selectivity

Mode of inhibition

IC50

References

Steroids

BChE

Non-competitive[84]

[8597,107110]

Triterpenes

BChE

Non-competitive[105]

Quinolizidine

AChE IC50 ~10-710-3 M


BChE IC50 ~10-710-3 M
AChE IC50 ~10-610-3 M
BChE IC50 ~10-710-3 M
AChE IC50 ~10-610-3 M
BChE IC50 ~10-410-3 M

Isoquinoline
Amaryllidaceae
Benzylisoquinoline, Aporphine

AChE

AChE
AChE

Competitive[73]

[99104,106]
[73,81,111114,116,117]

Competitive[73]
Non-competitive/mixed
(chelerythrine)[128]

AChE IC50 ~10-710-3 M


AChE IC50 ~10-610-3 M
BChE IC50 ~10-710-3 M

[53,74,119125]
[126134,136141]

Indole
Physostigmine

AChE

Competitive[73]

[64,65]

MIAS
Quaternary b-carbolines

BChE
AChE

Non-competitive[143,146,148]

AChE IC50 ~10-7 M


BChE IC50 ~10-6 M
f
BChE IC50 ~10-610-4 M
AChE IC50 ~10-610-5 M
BChE IC50 ~10-510-4 M

[128,142]
[142148]

Despite some steroid and triterpene alkaloids displaying selectivity for AChE inhibition, most of compounds belonging to these classes present some
capacity for BChE inhibition. bResults refer to Huperzine A. cResults referd to galantamine. dThe Amaryllidaceae alkaloids possessing the galantamine
skeleton inhibit AChE with IC50 values close to 10-6 M. eIn general, the quaternary compounds are about five-fold more active for AChE inhibition
than the tertiary benzylisoquinoline alkaloids. fThe MIAs seem to inhibit both eqBChE and hBChE, displaying similar potencies (IC50 values ranging
from 1 to 15 mM).[128]

achievement of highly active and less toxic products. Currently, a great deal of work involving the development of
new anti-Alzheimer therapeutics is underway in order to
provide more efficient lead compounds for future use.

Funding
The review received no specific grant from any funding
agency in the public, commercial or not-for-profit sectors.

Declarations
Conflict of interest
The Author(s) declare(s) that they have no conflicts of
interest to disclose.

References
1. Cummings JL. Alzheimers disease. N
Engl J Med 2004; 351: 5667.
2. Brookmeyer R et al. Forecasting the
global burden of Alzheimers disease.
Alzheimers Dem 2007; 3: 186191.
3. Ballard C et al. Alzheimers disease.
Lancet 2011; 377: 10191031.
4. Ferri CP et al. Global prevalence of
dementia: a Delphi consensus study.
Lancet 2005; 366: 21122117.
5. Kalaria RN et al. Alzheimers disease
and vascular dementia in developing
countries: prevalence, management,
and risk factors. Lancet Neurol 2008;
7: 812826.
6. Portelius E et al. An Alzheimers
disease-specific b-amyloid fragment
1720

7.

8.

9.

10.

11.

signature in cerebrospinal fluid. Neurosci Lett 2006; 409: 215219.


Kril JJ. Alzheimer disease: Alzheimer
disease neuropathology in the oldest
old. Nat Rev Neurol 2009; 5: 411412.
Schliebs R, Arendt T. The significance of the cholinergic system in the
brain during aging and in Alzheimers disease. J Neural Transm 2006;
113: 16251644.
Sauvaitre T et al. New potent acetylcholinesterase inhibitors in the tetracyclic triterpene series. J Med Chem
2007; 50: 53115323.
Lleo A et al. Current pharmacotherapy for Alzheimers disease.
Annu Rev Med 2006; 57: 513533.
Mangialasche F et al. Alzheimers
disease: clinical trials and drug devel-

12.

13.

14.

15.

16.

opment. Lancet Neurol 2010; 9: 702


716.
Buckholtz NS. Perspective: in search
of biomarkers. Nature 2011; 475:
S8.
Yamada K, Nabeshima T. Animal
models of Alzheimers disease and
evaluation of anti-dementia drugs.
Pharmacol Ther 2000; 88: 93113.
Van Dam D, De Deyn PP. Animal
models in the drug discovery pipeline for Alzheimers disease. Br J
Pharmacol 2011; 164: 12851300.
Mattson MP. Pathways towards and
away from Alzheimers disease.
Nature 2004; 430: 631639.
Benson A. Alzheimers disease: a
tangled issue. Drug Discov Today
2005; 10: 749751.

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

Eduardo Luis Konrath et al.

17. Hardy J, Allsop D. Amyloid deposition as the central event in the aetiology of Alzheimers disease. Trends
Pharmacol Sci 1991; 12: 383388.
18. Wang YP et al. Endogenous overproduction of b-amyloid induces tau
hyperphosphorylation and decreases
the solubility of tau in N2a cells.
J Neural Transm 2006; 113: 1723
1732.
19. Conway KA et al. Emerging betaamyloid therapies for the treatment
of Alzheimers disease. Curr Pharm
Des 2003; 9: 427447.
20. Morgan C et al. Structure and function of amyloid in Alzheimers
disease. Prog Neurobiol 2004; 74:
323349.
21. Bekris LM et al. Genetics of Alzheimer disease. J Geriatr Psychiatry
Neurol 2010; 23: 213227.
22. Hoyer S. Memory function and brain
glucose metabolism. Pharmacopsychiatry 2003; 36: S62S67.
23. Sun XT et al. Review of drugs for
Alzheimers disease. Drug Discov
Ther 2012; 6: 285290.
24. Squier TC. Oxidative stress and
protein aggregation during biological
aging. Exp Gerontol 2001; 36: 1539
1550.
25. He H et al. Anti-Amyloidogenic and
anti-apoptotic role of melatonin in
Alzheimer disease. Curr Neuropharmacol 2010; 8: 211217.
26. Cummings JL. Treatment of Alzheimers disease: current and future
therapeutic approaches. Rev Neurol
Dis 2004; 1: 6069.
27. Schliebs R. Basal forebrain cholinergic dysfunction in Alzheimers
disease-interrelationship with betaamyloid, inflammation and neurotrophin signaling. Neurochem Res
2005; 30: 895908.
28. Churcher I. Tau therapeutic strategies for the treatment of Alzheimers
disease. Curr Top Med Chem 2006; 6:
579595.
29. Selkoe DJ. Alzheimers disease: genes,
proteins, and therapy. Physiol Rev
2001; 81: 741766.
30. Cummings JL. The role of cholinergic agents in the management of
behavioural disturbances in Alzhe-

Anticholinesterase alkaloids for AD

31.

32.

33.

34.

35.

36.

37.

38.

39.

40.

41.

42.

imers disease. Int J Neuropsychopharmacol 2000; 3: S21S29.


Lahiri DK et al. Current drug targets
for Alzheimers disease treatment.
Drug Dev Res 2002; 56: 267281.
Giacobini E. Cholinesterases: new
roles in brain function and in Alzheimers disease. Neurochem Res 2003;
28: 515522.
Pakaski M, Kalman J. Interactions
between the amyloid and cholinergic
mechanisms in Alzheimers disease.
Neurochem Int 2008; 53: 103111.
Greenblatt HM et al. The complex of
a bivalent derivative of galanthamine
with torpedo acetylcholinesterase
displays drastic deformation of the
active-site gorge: implications for
structure-based drug design. J Am
Chem Soc 2004; 126: 1540515411.
Francis PT et al. Neurochemical basis
for symptomatic treatment of Alzheimers disease. Neuropharmacology
2010; 59: 221229.
Greig NH et al. Butyrylcholinesterase: an important new target in
Alzheimers disease therapy. Int Psychogeriatr 2002; 14: 7791.
Beard CM et al. The prevalence of
dementia is changing over time in
Rochester, Minnesota. Neurology
1995; 45: 7579.
Munoz-Torrero D. Acetylcholinesterase inhibitors as disease-modifying
therapies for Alzheimers disease.
Curr Med Chem 2008; 15: 2433
2455.
Ballard CG et al. Cholinesterases:
roles in the brain during health and
disease. Curr Alzheimer Res 2005; 2:
307318.
Loizzo MR et al. Natural products
and their derivatives as cholinesterase inhibitors in the treatment of
neurodegenerative disorders: an
update. Curr Med Chem 2008; 15:
12091228.
Martinez A, Castro A. Novel
cholinesterase inhibitors as future
effective drugs for the treatment of
Alzheimers disease. Expert Opin
Investig Drugs 2006; 15: 112.
Orhan IE et al. An overview on
natural cholinesterase inhibitors a
multi-targeted drug class and their

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

43.

44.

45.
46.

47.

48.

49.

50.

51.

52.

53.

54.

55.

mass production. Mini-Rev Med


Chem 2011; 11: 836842.
Eagger SA et al. Tacrine in Alzheimers disease. Lancet 1991; 337: 989
992.
Schneider LS. Clinical pharmacology
of aminoacridines in Alzheimers
disease. Neurology 1993; 43: S64
S79.
Davis KL, Powchick P. Tacrine.
Lancet 1995; 345: 625630.
Siddiqui MF, Levey AI. Cholinergic
therapies in Alzheimers disease.
Drugs Future 1999; 24: 417424.
Geldmacher DS. Donepezil (Aricept)
for treatment of Alzheimers disease
and other dementing conditions.
Expert Rev Neurother 2004; 4: 516.
Rogers SL et al. E2020 produces both
clinical global and cognitive test
improvement in patients with mild
to moderately severe Alzheimerrs
diseases: results of 30-week phase III
trial. Neurology 1996; 46: A217.
Scarpini E et al. Treatment of Alzheimers disease: current status and
new perspectives. Lancet Neurol
2003; 2: 539547.
Bores GM et al. Pharmacological
evaluation of novel Alzheimers
disease therapeutics: acetylcholinesterase inhibitors related to
galanthamine. J Pharmacol Exp Ther
1996; 277: 728738.
Harvey AL. The pharmacology of
galanthamine and its analogues.
Pharmacol Ther 1995; 68: 113128.
Wilkinson DG et al. Cholinesterase
inhibitors used in the treatment of
Alzheimers disease: the relationship
between pharmacological effects and
clinical efficacy. Drugs Aging 2004;
21: 453478.
Elgorashi EE et al. Acetylcholinesterase enzyme inhibitory effects of
Amaryllidaceae alkaloids. Planta Med
2004; 70: 260262.
Tariot PN, Federoff HJ. Current
treatment for Alzheimer disease and
future prospects. Alzheimer Dis Assoc
Disord 2003; 17: S105S113.
Perry EK et al. Medicinal plants and
Alzheimers disease: from ethnobotany to phytotherapy. J Pharm
Pharmacol 1999; 51: 527534.
1721

Anticholinesterase alkaloids for AD

56. Hostettmann K et al. Natural


product inhibitors of acetylcholinesterase. Curr Org Chem 2006; 10:
825847.
57. Houghton PJ et al. Acetylcholinesterase inhibitors from plants and fungi.
Nat Prod Rep 2006; 23: 181199.
58. Mukherjee PK et al. Acetylcholinesterase inhibitors from plants. Phytomedicine 2007; 14: 289300.
59. Mukherjee PK et al. Lead findinding
for acetyl cholinesterase inhibitors
from natural origin: structure activity relationship and scope. Mini Rev
Med Chem 2011; 11: 247262.
60. Orhan IE. Current concepts on
selected plant secondary metabolites
with promising inhibitory effects
against enzymes linked to Alzheimers disease. Curr Med Chem 2012;
19: 22522261.
61. Pereira DM et al. Pharmacological
effects of Catharanthus roseus root
alkaloids in acetylcholinesterase inhibition and cholinergic neurotransmission. Phytomedicine 2010; 17:
646652.
62. Bruhlmann C et al. Screening of
non-alkaloidal natural compounds as
acetylcholinesterase inhibitors. Chem
Biodivers 2004; 1: 819829.
63. Bruneton J. Pharmacognosie Phytochimie Plantes Mdicinale, 3me edn.
Paris: Tec & Doc Lavoisier, 1999.
64. Asthana S et al. Treatment of Alzheimer disease by continuous intravenous infusion of physostigmine.
Alzheimer Dis Assoc Disord 1995; 9:
223232.
65. Thal LJ et al. A multicenter doubleblind study of controlled-release
physostigmine for the treatment of
symptoms secondary to Alzheimers
disease. Physostigmine Study Group.
Neurology 1996; 47: 13891395.
66. Guo B et al. Research advances of
Huperzia serrata (Thunb.) Trev. J
Chin Mat Med 2009; 34: 2018
2023.
67. Ma X et al. Huperzine A from Huperzia speciesan ethnopharmacolgical
review. J Ethnopharmacol 2007; 113:
1534.
68. Zhang HY, Tang XC. Neuroprotective effects of huperzine A: new
1722

Eduardo Luis Konrath et al.

69.

70.

71.

72.

73.

74.

75.

76.

77.

78.

79.

therapeutic targets for neurodegenerative disease. Trends Pharmacol Sci


2006; 27: 619625.
Liang YQ, Tang XC. Comparative
effects of huperzine A, donepezil and
rivastigmine on cortical acetylcholine
level and acetylcholinesterase activity
in rats. Neurosci Lett 2004; 361:
5659.
Xu SS et al. Huperzine-A in capsules
and tablets for treating patients with
Alzheimer disease. Zhongguo Yao Li
Xue Bao 1999; 20: 486490.
Zhang Z et al. Clinical efficacy and
safety of huperzine Alpha in
treatment of mild to moderate
Alzheimer disease, a placebocontrolled, double-blind, randomized trial. Zhonghua Yi Xue Za Zhi
2002; 82: 941944.
Rafii MS et al. A phase II trial of
huperzine A in mild to moderate
Alzheimer disease. Neurology 2011;
76: 13891394.
Ma X, Gang DR. The Lycopodium
alkaloids. Nat Prod Rep 2004; 21:
752772.
Lopez S et al. Acetylcholinesterase
inhibitory activity of some Amaryllidaceae alkaloids and Narcissus
extracts. Life Sci 2002; 71: 2521
2529.
Pagliosa LB et al. Effect of isoquinoline alkaloids from two Hippeastrum
species on in vitro acetylcholinesterase activity. Phytomedicine 2010; 17:
698701.
Kim DK. Inhibitory effect of corynoline isolated from the aerial parts of
Corydalis incisa on the acetylcholinesterase. Arch Pharm Res 2002; 25:
817819.
Kim DK et al. Acetylcholinesterase
inhibitors from the aerial parts of
Corydalis speciosa. Arch Pharm Res
2004; 27: 11271131.
Rahman AU, Choudhary MI. Bioactive natural products as a potential
source of new pharmacophores. A
theory of memory. Pure Appl Chem
2001; 73: 555560.
Ortega MG et al. Anticholinesterase
activity in an alkaloid extract of
Huperzia saururus. Phytomedicine
2004; 11: 539543.

80. Konrath EL et al. Investigation of the


in vitro and ex vivo acetylcholinesterase and antioxidant activities of traditionally used Lycopodium species
from South America on alkaloid
extracts. J Ethnopharmacol 2012; 139:
5867.
81. Konrath EL et al. Alkaloid profiling
and anticholinesterase activity of
South American Lycopodiaceae
species. J Enzyme Inhib Med Chem
2013; 28: 218222.
82. Andrade MT et al. Indole alkaloids
from Tabernaemontana australis
(Muell. Arg) Miers that inhibit
acetylcholinesterase enzyme. Bioorg
Med Chem 2005; 13: 40924095.
83. Rahman AU et al. Diterpenoid and
steroidal alkaloids. Nat Prod Rep
1997; 14: 191203.
84. Khalid A et al. Kinetics and
structure-activity relationship studies
on pregnane-type steroidal alkaloids
that inhibit cholinesterases. Bioorg
Med Chem 2004; 12: 19952003.
85. Rahman AU et al. New pregnanetype steroidal alkaloids from Sarcocca
saligna and their cholinesterase
inhibitory activity. Steroids 2004; 69:
735741.
86. Devkota KP et al. Cholinesterase
inhibiting and antiplasmodial steroidal alkaloids from Sarcococca hookeriana. Chem Pharm Bull 2007; 55:
13971401.
87. Kalauni SK et al. New cholinesterase
inhibiting steroidal alkaloids from
the leaves of Sarcococca coriacea of
Nepalese origin. Chem Pharm Bull
2002; 50: 14231426.
88. Choudhary MI et al. Cholinesteraseinhibiting new steroidal alkaloids
from Sarcococca hookeriana of Nepalese Origin. Helv Chim Acta 2004; 87:
10991108.
89. Choudhary MI et al. Cholinesterase
inhibitory pregnane-type steroidal
alkaloids from Sarcococca hookeriana.
Steroids 2005; 70: 295303.
90. Devkota
KP
et al.
Bioactive
5a-Pregnane-Type Steroidal Alkaloids from Sarcococca hookeriana.
J Nat Prod 2008; 71: 14811484.
91. Rahman
AU
et al.
New
Cholinesterase-Inhibiting Steroidal

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

Eduardo Luis Konrath et al.

92.

93.

94.

95.

96.

97.

98.

99.

100.

101.

102.

Alkaloids from Sarcococca saligna.


Helv Chim Acta 2004; 87: 439448.
Khalid A et al. Cholinesterase inhibitory and spasmolytic potential of
steroidal alkaloids. J Steroid Biochem
Mol Biol 2004; 92: 477484.
Kalauni SK et al. Steroidal alkaloids
from the leaves of Sarcococca coriacea
of Nepalese origin. J Nat Prod 2001;
64: 842844.
Rahman AU et al. Pregnane-type
steroidal alkaloids of Sarcococca
saligna: a new class of cholinesterase
inhibitors. Helv Chim Acta 2002; 85:
678688.
Giliani AU et al. Presence of antispasmodic, antidiarrheal, antisecretory, calcium antagonist and
acetylcholinesterase inhibitory steroidal alkaloids in Sarcococca saligna.
Planta Med 2005; 71: 120125.
Zaheer ul H et al. 3D-QSAR studies
on
natural
acetylcholinesterase
inhibitors of Sarcococca saligna by
comparative molecular field analysis
(CoMFA). Bioorg Med Chem Lett
2003; 13: 43754380.
Zaheer-Ul-Haq ZU et al. Molecular
docking
studies
of
natural
cholinesterase-inhibiting
steroidal
alkaloids from Sarcococca saligna.
J Med Chem 2003; 46: 50875090.
Musharraf SG et al. Structurefragmentation relationship and rapid
dereplication of Buxus steroidal alkaloids by electrospray ionizationquadrupole time-of-flight mass
spectrometry. Rapid Commun Mass
Spectrom 2013; 27: 169178.
Babar ZU et al. New bioactive steroidal alkaloids from Buxus hyrcana.
Steroids 2006; 71: 10451051.
Choudhary MI et al. New triterpenoid alkaloid cholinesterase inhibitors from Buxus hyrcana. J Nat Prod
2003; 66: 739742.
Rahman AU et al. Acetyl and
butyrylcholinesterase-inhibiting
triterpenoid alkaloids from Buxus
papillosa. Phytochemistry 2001; 58:
963968.
Choudhary
MI
et al.
New
cholinesterase-inhibiting
triterpenoid alkaloids from Buxus hyrcana.
Chem Biodivers 2006; 3: 10391052.

Anticholinesterase alkaloids for AD

103. Ata A et al. Triterpenoidal alkaloids


from Buxus hyrcana and their
enzyme inhibitory, anti-fungal and
anti-leishmanial activities. Phytochemistry 2010; 71: 17801786.
104. Matochko WL et al. Triterpenoidal
alkaloids from Buxus natalensis and
their acetylcholinesterase inhibitory
activity. J Nat Prod 2010; 73: 1858
1862.
105. Khalid A et al. Structural basis of
acetylcholinesterase inhibition by
triterpenoidal alkaloids. Biochem
Biophys Res Commun 2005; 331:
15281532.
106. Orhan IE et al. Selective cholinesterase inhibitors from Buxus sempervirens L. and their molecular docking
studies. Curr Comp Aided Drug Des
2011; 7: 276286.
107. Zhou JL et al. Characterization and
identification of steroidal alkaloids
in Fritillaria species using liquid
chromatography coupled with electrospray ionization quadrupole timeof-flight tandem mass spectrometry.
J Chromatogr A 2010; 1217: 7109
7122.
108. Lin BQ et al. Inhibitors of acetylcholine esterase in vitro screening of
steroidal alkaloids from Fritillaria
species. Planta Med 2006; 72: 814
818.
109. Rahman AU et al. New steroidal alkaloids from Fritillaria imperialis and
their cholinesterase inhibiting activities. Chem Pharm Bull 2002; 50:
10131016.
110. Yang ZD et al. Steroidal alkaloids
from Holarrhena antidysenterica as
acetylcholinesterase inhibitors and
the investigation for structureactivity relationships. Life Sci 2012;
90: 929933.
111. Halldorsdottir ES et al. Acetylcholinesterase inhibitory activity of
lycopodane-type alkaloids from the
Icelandic Lycopodium annotinum ssp.
alpestre. Phytochemistry 2010; 71:
149157.
112. Koyama K et al. Cryptadines A and
B, novel C27N3-type pentacyclic
alkaloids from Lycopodium cryptomerinum. Bioorg Med Chem 2007;
15: 78037808.

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

113. Hirasawa Y et al. Lycoperine A, A


novel C27N3-type pentacyclic alkaloid from Lycopodium hamiltonii,
inhibiting acetylcholinesterase. Org
Lett 2005; 8: 123126.
114. Choo CY et al. Carinatumins A-C,
new alkaloids from Lycopodium carinatum inhibiting acetylcholinesterase. Bioorg Med Chem 2007; 15:
17031707.
115. Hirasawa Y et al. Lycoparins A-C,
new alkaloids from Lycopodium casuarinoides inhibiting acetylcholinesterase. Bioorg Med Chem 2008;
16: 61676171.
116. Hirasawa Y et al. Sieboldine A, a
novel tetracyclic alkaloid from Lycopodium sieboldii, inhibiting acetylcholinesterase. Org Lett 2003; 5:
39913993.
117. Konrath EL. Qumica, atividades
antioxidante e anticolinestersicas de
espcies da famlia Lycopodiaceae.
PhD Thesis. Faculty of Pharmacy.
Federal University of Rio Grande
do Sul (UFRGS): Porto Alegre,
2011.
118. Ghosal S et al. Crinum alkaloids:
their chemistry and biology. Phytochemistry 1985; 24: 21412156.
119. Greenblatt HM et al. Structure
of acetylcholinesterase complexed
with (-)-galanthamine at 2.3 A
resolution. FEBS Lett 1999; 463:
321326.
120. Reyes-Chilpa
R
et al.
Acetylcholinesterase-inhibiting alkaloids from Zephyranthes concolor.
Molecules 2011; 16: 95209533.
121. Bartolucci C et al. Three-dimensional
structure of a complex of galanthamine (Nivalin) with acetylcholinesterase from Torpedo californica:
implications for the design of new
anti-Alzheimer drugs. Proteins 2001;
42: 182191.
122. Rhee IK et al. Isolation of the acetylcholinesterase inhibitor ungeremine
from Nerine bowdenii by preparative
HPLC coupled on-line to a flow assay
system. Biol Pharm Bull 2004; 27:
18041809.
123. Sarikaya BB et al. Alkaloids from
Galanthus rizehensis. Phytochem Lett
2012; 5: 367370.
1723

Anticholinesterase alkaloids for AD

124. Houghton PJ et al. Choline esterase


inhibitory properties of alkaloids
from two Nigerian Crinum species.
Phytochemistry 2004; 65: 28932896.
125. Adewusi EA et al. Cytotoxicity and
acetylcholinesterase inhibitory activity of an isolated crinine alkaloid
from Boophane disticha (Amaryllidaceae). J Ethnopharmacol 2012; 143:
572578.
126. Ingkaninan K et al. Acetylcholinesterase inhibitors from Stephania venosa
tuber. J Pharm Pharmacol 2006; 58:
695700.
127. Huang QQ et al. Bioactive isoquinoline alkaloids from Corydalis saxicola.
Planta Med 2011; 78: 6570.
128. Brunhofer G et al. Exploration of
natural compounds as sources of
new bifunctional scaffolds targeting
cholinesterases and beta amyloid
aggregation: the case of chelerythrine. Bioorg Med Chem 2012; 20:
66696679.
129. Hung TM et al. Cholinesterase
inhibitory activities of alkaloids from
Corydalis tuber. Nat Prod Sci 2011;
17: 108112.
130. Hung TM et al. Cholinesterase
inhibitory and anti-amnesic activity
of alkaloids from Corydalis turtschaninovii. J Ethnopharmacol 2008;
119: 7480.
131. Hung TM et al. Alkaloids from roots
of Stephania rotunda and their
cholinesterase inhibitory activity.
Planta Med 2010; 76: 17621764.
132. Jung HA et al. Anti-Alzheimer and
antioxidant activities of Coptidis
Rhizoma alkaloids. Biol Pharm Bull
2009; 32: 14331438.
133. Ji HF, Shen L. Berberine: a potential
multipotent natural product to
combat Alzheimers disease. Molecules 2011; 16: 67326740.
134. Ji HF, Shen L. Molecular basis of
inhibitory activities of berberine
against pathogenic enzymes in
Alzheimers disease. Sci World J
2012; 2012: 823201.
135. Asai M et al. Berberine alters the
processing of Alzheimers amyloid
precursor protein to decrease Abeta
secretion. Biochem Biophys Res
Commun 2007; 352: 498502.
1724

Eduardo Luis Konrath et al.

136. Xiao HT et al. Acetylcholinesterase


inhibitors from Corydalis yanhusuo.
Nat Prod Res 2011; 25: 1418
1422.
137. Tsai SF, Lee SS. Characterization
of acetylcholinesterase inhibitory
constituents from Annona glabra
assisted by HPLC microfractionation. J Nat Prod 2010; 73: 1632
1635.
138. Adsersen A et al. Acetylcholinesterase
and butyrylcholinesterase inhibitory
compounds from Corydalis cava Schweigg. & Kort. J Ethnopharmacol
2007; 113: 179182.
139. Mollataghi
A
et al.
Antiacetylcholinesterase,
anti-alphaglucosidase, anti-leishmanial and
anti-fungal activities of chemical
constituents of Beilschmiedia species.
Fitoterapia 2012; 83: 298302.
140. Cometa MF et al. New cholinesterase
inhibiting
bisbenzylisoquinoline
alkaloids from Abuta grandifolia. Fitoterapia 2012; 83: 476480.
141. Rahman AU et al. New cholinesterase
inhibiting
bisbenzylisoquinoline
alkaloids from Cocculus pendulus.
Chem Pharm Bull 2004; 52: 802
806.
142. Passos CS et al. Indole alkaloids
of Psychotria as multifunctional
cholinesterases and monoamine oxidases inhibitors. Phytochemistry
2013; 86: 820.
143. Bharate SB et al. Total synthesis
and anti-cholinesterase activity of
marine-derived bis-indole alkaloid
fascaplysin. MedChemComm 2012; 3:
10981103.
144. Geissler T et al. Acetylcholinesterase
inhibitors from the toadstool Cortinarius infractus. Bioorg Med Chem
2010; 18: 21732177.
145. Schott Y et al. 6-Hydroxy- and
6-methoxy-beta-carbolines as acetyland butyrylcholinesterase inhibitors.
Bioorg Med Chem Lett 2006; 16:
58405843.
146. Torres JM et al. Structural insights
into cholinesterases inhibition by
harmane b-carbolinium derivatives:
a kinetics Molecular modeling
approach. Phytochemistry 2012; 81:
2430.

147. Becher PG et al. Nostocarboline:


isolation and synthesis of a new
cholinesterase inhibitor from Nostoc
78-12A. J Nat Prod 2005; 68: 1793
1795.
148. Becher PG et al. The cyanobacterial alkaloid nostocarboline: an
inhibitor of acetylcholinesterase and
trypsin. J Appl Phycol 2009; 21: 103
110.
149. Choudhary MI et al. Juliflorine: a
potent natural peripheral anionicsite-binding inhibitor of acetylcholinesterase with calcium-channel
blocking potential, a leading candidate for Alzheimers disease therapy.
Biochem Biophys Res Commun 2005;
332: 11711177.
150. Ferheen S et al. Haloxylines A and
B, antifungal and cholinesterase
inhibiting piperidine alkaloids from
Haloxylon salicornicum. Chem Pharm
Bull 2005; 53: 570572.
151. Nukoolkarn VS et al. Petrosamine, a
potent anticholinesterase pyridoacridine alkaloid from a Thai marine
sponge Petrosia n. sp. Bioorg Med
Chem 2008; 16: 65606567.
152. Cardoso-Lopes EM et al. Alkaloids
from stems of Esenbeckia leiocarpa
Engl. (Rutaceae) as potential treatment for Alzheimer disease. Molecules 2010; 15: 92059213.
153. Wang YH et al. Benzylphenethylamine alkaloids from Hosta plantaginea with inhibitory activity against
tobacco mosaic virus and acetylcholinesterase. J Nat Prod 2007; 70:
14581461.
154. Francis PT et al. The cholinergic
hipothesis of Alzheimers disease:
a review of progress. J Neurol
Neurosurg Psychiatry 1999; 66: 137
147.
155. Eriksson AH et al. In vitro evaluation of the P-glycoprotein interactions of a series of potentially CNS
active Amaryllidaceae alkaloids. J
Pharm Pharmacol 2012; 64: 1667
1677.
156. McNulty J et al. Selective cytochrome
P450 3A4 inhibitory activity of
Amaryllidaceae alkaloids. Bioorg
Med Chem Lett 2009; 19: 3233
3237.

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

Eduardo Luis Konrath et al.

Supporting Information
Additional
Supporting
Information
may be found in the online version of
this article at the publishers website:

Anticholinesterase alkaloids for AD

Table S1 Structures of steroidal and triterpenoidal alkaloids with anticholinesterase activity


Table S2 Structures of quinolizidine
(Lycopodium) alkaloids with anticholinesterase activity

2013 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 65, pp. 17011725

Table S3 Structures of isoquinoline and


indole alkaloids with anticholinesterase
activity
Table S4 Structures of miscellaneoustype alkaloids with anticholinesterase
activity

1725

Anda mungkin juga menyukai