Sunteți pe pagina 1din 11

e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 3 3 ( 2 0 0 8 ) 109–119

available at www.sciencedirect.com

journal homepage: www.elsevier.com/locate/ejps

In vitro screening assays to identify natural or synthetic


acetylcholinesterase inhibitors: Thin layer
chromatography versus microplate methods

Saviana Di Giovanni a , Aline Borloz b , Aurélie Urbain b , Andrew Marston b ,


Kurt Hostettmann b , Pierre-Alain Carrupt a , Marianne Reist a,∗
a LCT-Pharmacochimie, Ecole de Pharmacie Genève-Lausanne, Section des Sciences Pharmaceutiques, Quai Ernest-Ansermet 30,
CH-1211 Genève 4, Switzerland
b Laboratoire de Pharmacognosie et Phytochimie, Ecole de Pharmacie Genève-Lausanne, Section des Sciences Pharmaceutiques,

Quai Ernest-Ansermet 30, CH-1211 Genève 4, Switzerland

a r t i c l e i n f o a b s t r a c t

Article history: Acetylcholinesterase inhibitors (AChEI) are currently still the best available pharmacother-
Received 28 August 2007 apy for Alzheimer patients. Successful screening for new AChEI relies on effective and fast
Received in revised form assays. Two colorimetric screening assays frequently used to search for new AChEI, namely
17 October 2007 a thin layer chromatography (TLC) assay with Fast Blue B salt as reagent and a 96-well plate
Accepted 23 October 2007 assay based on Ellman’s method, were compared. For the majority (83%) of the 138 test
Published on line 30 October 2007 compounds of natural and synthetic origin, the results obtained with the two assays con-
verged and both screening assays were considered suitable for the generation of new hits.
Keywords: Fifteen percent of investigated compounds were classified as active with the microplate
Acetylcholinesterase inhibitors assay but were shown to be inactive by TLC and about 2% were measured active by TLC but
Thin layer chromatography showed to be inactive with the microplate assay. These divergences were not due to the
Microplate method main differences between the experimental protocols of the two screening assays, namely
Screening the different colorimetric methods and pre-incubation of test compounds with acetyl-
Alzheimer’s disease cholinesterase (AChE). They might be explained by the interaction of either AChE or test
Ellman’s method compounds with the silica of the TLC plates, resulting in an altered affinity of the enzyme
Fast blue B salt for the compounds.
© 2007 Elsevier B.V. All rights reserved.

1. Introduction (ACh) by nearly 90%. On the behavioral side, the impor-


tant decrease of ACh levels causes impairment in cognitive
Alzheimer’s disease (AD) is the leading cause of dementia function (Cummings, 2004; Selkoe, 1996). This “cholinergic
among older people. An estimated 10% of the world’s popu- hypothesis of AD”, formulated for the first time by Whitehouse
lation over the age of 65 years is afflicted by AD (Racchi et et al. (1982), was postulated on the basis of the negative cog-
al., 2004). The most important changes observed in the brain nitive effect of cholinergic antagonists and the positive effect
of AD patients are the appearance of neuritic plaques and of muscarinic agonists on memory in humans (Guillou et al.,
neurofibrillary tangles, as well as a decrease in hippocam- 2000) and became the neurobiologic incentive for a treatment
pal and cortical levels of the neurotransmitter acetylcholine aiming at the improvement of cholinergic function in AD.


Corresponding author. Tel.: +41 22 379 33 65; fax: +41 22 379 33 60.
E-mail address: marianne.reistoechslin@pharm.unige.ch (M. Reist).
0928-0987/$ – see front matter © 2007 Elsevier B.V. All rights reserved.
doi:10.1016/j.ejps.2007.10.004
110 e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 3 3 ( 2 0 0 8 ) 109–119

Acetylcholinesterase (AChE) is the enzyme involved in the nation of AChE activity have been described. These include
metabolic hydrolysis of ACh at cholinergic synapses in the colorimetric methods using Ellman’s reagent (Ellman et al.,
central and peripheral nervous system. Thus, inhibitors of 1961) or Fast Blue B salt reagent, fluorometric assays with flu-
AChE activity (AChEIs) promote an increase in the concentra- orogenic substrates, and pH-metric or electrochemical activity
tion and duration of action of synaptic ACh (Rollinger et al., detection of AChE (Rhee et al., 2003). They have been used to
2004). Currently, AChEIs represent the therapy of choice for quantify AChE in tissue (Ren et al., 2002), to study anticholin-
AD (Martinez and Castro, 2006). ergic agents for use as insecticides and pesticides (Hawkins
About 50% of the drugs introduced on the market dur- and Knittle, 1972; Motoyama et al., 1980; van Asperen, 1962),
ing the last 20 years have been derived directly or indirectly and to screen for AChE inhibitors of interest for the treatment
from small molecules of natural origin (Newman and Cragg, of AD (Ingkaninan et al., 2003; Rhee et al., 2001; Rhee et al.,
2007). Their therapeutic potential has also been success- 2003).
fully demonstrated in the field of AD (Hostettmann et al., The choice of an appropriate screening assay is cru-
2006; Howes and Houghton, 2003). Hence, nature can be con- cial. Indeed, biological activities of natural and synthetic
sidered as an important source of new chemical entities compounds obtained with different tests and/or by dif-
potentially interesting for the treatment of various diseases, ferent research groups can be quite variable (Buenger et
including AD. In addition, several interesting compounds were al., 2006; Di et al., 2007; Lam, 2007). Hence, the aim of
recently synthesized and tested for their ability to inhibit AChE this study was the comparison of two colorimetric screen-
(Brühlmann et al., 2001). ing assays frequently used to detect acetylcholinesterase
In order to speed up the screening for AChE inhibitors inhibitory activity, namely a TLC assay using Fast Blue B salt
of natural and synthetic origin, effective and fast assays are as reagent, previously reported for the detection of acetyl-
needed. In the past years, several methods for the determi- cholinesterase inhibitors in plants (Marston et al., 2002), and

Fig. 1 – Chemical structures of reference AChEIs tested.


e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 3 3 ( 2 0 0 8 ) 109–119 111

a microplate assay using Ellman’s method (Brühlmann et al., microplate assay resulted inactive in the TLC test and about
2004). 2% of investigated compounds measured active in the TLC
The test set of 138 compounds consisted of seven refer- assay were shown to be inactive with the microplate assay.
ence acetylcholinesterase inhibitors (Fig. 1) and compounds Hence, this divergence in the identification of AChE inhibitors
chosen from a focused screening library for the identification observed between the two screening assays was further inves-
of dual inhibitors of acetylcholinesterase and monoamine oxi- tigated and the advantages and inconveniences of the two
dase B composed of compound classes reported to have either tests were discussed.
one or both of these activities (Brühlmann et al., 2001, 2004;
Gnerre et al., 2001; Kneubühler et al., 1995). These are 7 com-
pounds of natural origin (4 coumarins, 1 xanthone, 1 terpene, 2. Experimental
and 1 alkaloid), 77 compounds based on scaffolds of natu-
ral origin (47 coumarins, 13 aurones, 12 azaaurones, and 5 2.1. Materials
chromones), and 47 synthetic compounds (45 5H-indeno-[1,2-
c]-pyridazin-5-ones, 5-(benzyloxy)benzo[d][1,3]dioxole, and Acetylcholinesterase from Electrophorus electricus (E.C.3.1.1.7,
5-(benzyloxy)isoindoline-1,3-dione) (Fig. 2). Sigma C 2888), acetylthiocholine iodide (ATCI), 5,5 -dithiobis-
For the majority of compounds tested, the results obtained (2-nitrobenzoic acid) (DNTB) and the reference compounds
with the two screening assays were convergent. However, galanthamine hydrobromide, physostigmine hemisulfate,
about 15% of investigated compounds testing positive in the propidium iodide, (±)-huperzine A and tacrine hydrochlo-

Fig. 2 – Chemical structures of compound classes examined.


112 e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 3 3 ( 2 0 0 8 ) 109–119

ride were purchased from Sigma–Aldrich Chemical (St. Louis, compound solutions. The enzymatic reaction was initiated by
MA, USA). DMSO (microselect for molecular biology), phos- addition of 1.6 ␮l of ␣-naphthyl acetate solution (final concen-
phate salts, Fast Blue B salt (O-dianisidine-bis (diazotized) tration equal to the Km value) in DMSO. The final concentration
hydrochloride zinc double salt), sodium dodecylsulphate of DMSO was 0.6 or 1.6% and the final assay volume was 260 ␮l.
(SDS), and ␣-naphthyl acetate came from Fluka AG (Buchs, After mixing for 90 s and incubation at 25 ◦ C for 90 s, the reac-
CH). (−)-Huperzine-A was from Sequoia Research Products tion was stopped with 20 ␮l of a 5% solution of sodium dodecyl
Ltd. (Oxford, UK). Donepezil hydrochloride was from Interchim sulphate (SDS) in water. The color was developed with 20 ␮l of
(Montluçon, France). Fast Blue B salt solution (final concentration 0.17 mM in water).
The four coumarins of natural origin (C-1 to C-4) were iso- Enzyme activity and inhibition were quantified by determina-
lated from Peucedanum ostruthium (Apiaceae) (Urbain et al., tion of the absorbance in end point lecture at 600 nm after
2005), the xanthone derivative from Gentiana campestris (Gen- formation of the purple-colored diazonium dye, which turns
tianaceae) (Urbain et al., 2004), the diterpene derivative from to stable blue in presence of SDS.
Detarium microcarpum (Caesalpinaceae) (Cavin et al., 2006), and
the alkaloid lycopodine from Huperzia selago (Lycopodiaceae) 2.3. Microplate assay using Ellman’s method
(Borloz, 2007).
The synthesis of coumarin derivatives (C-5 to C-51, For the microplate assay carried out following Ellman et al.
n = 47), 5H-indeno [1,2-c] pyridazin-5-one derivatives (n = 45), (1961), wells were filled with 227.3 ␮l or 210 ␮l of Ellman’s
chromones (n = 5), 5-(benzyloxy)benzo[d][1,3]dioxole, and 5- reagent (0.15 mM final concentration of 5,5 -dithiobis-(2-
(benzyloxy)isoindoline-1,3-dione was performed in the Dipar- nitrobenzoic acid) in 0.1 M phosphate buffer pH 7.4), 20 ␮l
timento Farmaco-Chimico, Università di Bari, I-70125, Bari, of a solution of acetylcholinesterase (final concentration
Italy, as described previously (Gnerre et al., 2000; Kneubühler 0.037 U/ml in 0.1 M phosphate buffer pH 7.4), and 2.7 ␮l of test
et al., 1995). Aurone and azaaurone derivatives (n = 25) were compound solutions in DMSO or 20 ␮l of test compound solu-
synthesized in the Département de Pharmacochimie Molec- tions in buffer. For controls, test compound solutions were
ulaire, UMR CNRS 5063, Faculté de Pharmacie de Grenoble, replaced by the corresponding volume of DMSO or buffer.
France (Boumendjel et al., 2002; Okombi et al., 2006). The enzymatic reaction was initiated by addition of 20 ␮l
All buffers and solutions were prepared in demineralized of acetylthiocholine iodide (ATCI) solution in demineralised
and purified water obtained with the Elix 3 Millipore water water (final concentration equal to the Km value). The final
purifying system, except test compound solutions that were assay volume was 270 ␮l. The plate was shaken for 2 s and
prepared in DMSO. All assays in solution were performed in the increase in absorbance at 412 nm was monitored at 25 ◦ C
conventional, flat-bottomed polypropylene 96-well microtitre for 180 s using a microplate spectrophotometer (PowerWaveX ,
plates (Evergreen Scientific, Los Angeles, USA). BioTek Instrument, Winooski, Vermont, USA). The kinetic
parameters Km and Vmax and inhibitory potencies of seven ref-
2.2. TLC and microplate assays using Fast Blue B salt erence compounds were measured in presence of 0 (control),
as reagent 0.6%, 1.6% and 2% of DMSO (Table 1).

Using Fast Blue B salt as reagent, the inhibitory activity of 2.4. Determination of kinetic parameters Km and Vmax
compounds was determined either with a TLC assay or a
microplate assay. The detailed procedure of the TLC assay To obtain the kinetic parameters Km and Vmax , hydrolysis rates
has been described previously by Marston et al. (2002). Briefly, for duplicates of six concentrations of substrate were deter-
compounds dissolved either in ethyl acetate or in methanol, mined. When using Ellman’s method, ATCI solutions (final
according to their solubility, were spotted first at 1 ␮g, then at concentrations in the reaction mixture ranging from 0.09 mM
decreasing quantities in the case of active compounds, onto to 1.11 mM) were prepared in water. For the microplate assay
the TLC plate. (±)-Huperzine-A (0.001 ␮g) was added to each using Fast Blue B salt as reagent, ␣-naphthyl acetate solutions
plate as positive control. Migration was conducted with hex- (final concentrations of 0.06–1.02 mM) were prepared in DMSO.
ane:ethyl acetate (1:1; v/v). Then the plate was sprayed with The kinetic parameters Km and Vmax were obtained by curve
an enzyme stock solution (concentration 6.67 U/ml) and pre- fitting according to the classical Michaelis–Menten equation
incubated at 37 ◦ C for 20 min in a humid atmosphere. Ten (Michaelis and Menten, 1913). Data are means of two inde-
millilitres of the ␣-naphthyl acetate solution and 40 ml of the pendents experiments, each performed in duplicate.
Fast Blue B salt solution were mixed and sprayed onto the plate
to give a purple coloration after 1–2 min. The minimum quan- 2.5. Expression of inhibitory activities
tity of compound was noted for which an inhibition spot was
still visible. For the TLC assay, AChE inhibitory activities were expressed
For the microplate assay with Fast Blue B salt as reagent, as pMIQ, which represents the negative logarithm of the min-
the method published by van Asperen (1962) was adapted for imal inhibitory quantity in mole that produced the spot with
96-well plates. The reaction mixture consisted of 235.7 ␮l or the least observable whiteness. For the two microplate assays,
188.4 ␮l of 0.1 M phosphate buffer pH 7.4, 20 ␮l of a solution of the activity of a compound was expressed either as percent-
AChE (final concentration 0.1 U/ml in 0.1 M phosphate buffer age of inhibition at a fixed concentration of 10−5 M or as pIC50 .
pH 7.4), 2.7 ␮l of test compound solutions in DMSO or 50 ␮l The percentage of inhibition was calculated relative to a con-
of test compound solutions in buffer. The controls contained trol sample, for which the AChE activity was assessed under
the corresponding volume of DMSO or buffer instead of test identical conditions but in absence of test compound (con-
e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 3 3 ( 2 0 0 8 ) 109–119 113

Table 1 – Kinetic parameters (Km and Vmax ) determined in solution in presence of different percentages of DMSO
Percent of DMSO Microtitre plate method with Ellman’s reagent Microtitre plate method with Fast Blue B salt as reagent

Km (mM)a Vmax (mM min−1 U−1 )a Km (mM)a Vmax (mM min−1 U−1 )a

0 0.12 ± 0.02 1.39 ± 0.06 – –


0.6 0.21 ± 0.04 0.86 ± 0.06 0.23 ± 0.01 0.27 ± 0.02
1.6 0.25 ± 0.09 0.84 ± 0.11 0.25 ± 0.03 0.26 ± 0.02
2 0.36 ± 0.04 0.52 ± 0.02 0.41 ± 0.04 0.20 ± 0.02

a
Means ± standard errors (n = 4).

sidered to be 100%). Data are means of n = 3. The inhibitory hyperbola and reported in Table 1. Since ␣-naphthyl acetate is
potencies, expressed as pIC50 values, represent the negative not soluble in water, its AChE-catalyzed hydrolysis in absence
logarithms of the molar concentrations of inhibitor required of co-solvent could not be measured; a minimal final DMSO
to decrease AChE activity by 50% and were calculated by curve concentration of 0.6% was necessary to solubilize this sub-
fitting according to the classical sigmoidal dose–response strate.
equation. All pIC50 values were determined in duplicate from Interestingly, AChE showed the same affinity for the
at least six compound concentrations, with the exception of two substrates acetylthiocholine and ␣-naphthyl acetate.
inhibitory potencies of seven reference AChEIs, determined Moreover, the Km values obtained, i.e. 0.12 ± 0.02 mM for
using Ellman’s method, that are means of two independent acetylthiocholine in absence of co-solvent, and 0.23 ± 0.01 mM
experiments, each performed in duplicate. for ␣-naphthyl acetate in presence of 0.6% of DMSO, were
in good agreement with those reported in the literature, i.e.
2.6. Data analysis 0.11 ± 0.01 mM for acetylthiocholine (Selwood et al., 1993) and
0.1 mM for ␣-naphthyl acetate (van Asperen, 1962). The lat-
Statistical significance was determined by the use of Stu- ter was determined with acetone as co-solvent for housefly
dent’s t-test and differences were considered significant when esterases, which can explain the slight difference between the
p < 0.05. Statistical tests, kinetic parameters, percents of inhi- Km value determined for ␣-naphthyl acetate in the present
bition, inhibitory potencies and correlation coefficients were study and the one reported by van Asperen (1962).
calculated using Prism V4.0 (GraphPad Software Inc.). As previously described (Plummer et al., 1983), it was shown
that DMSO inhibits acetylcholinesterase. Indeed, the Km val-
ues for both substrates increased and the turnover numbers
3. Results and discussion decreased with increasing percentages of DMSO, indicating a
mixed type inhibition by DMSO of both enzymatic reactions
3.1. Effect of dimethylsulfoxide on screening tests in (Table 1). Inhibition constants for the free enzyme (Ki ) and the
solution enzyme–substrate complex (␣Ki ) determined with acetylthio-
choline as substrate were 0.42 mM and 0.14 mM, respectively.
DMSO is usually the solvent of choice for preparing stock It has to be noted that the inhibition type reported by Plummer
solutions of molecule libraries for biological and pharmaco- et al. (1983) was competitive inhibition with a Ki of 0.11 mM.
logical studies, as well as for activity screening, and was thus Hence, the inhibition constants were in the same order of mag-
also used to solubilize ␣-naphthyl acetate and the 138 tested nitude although the inhibition type differed between the two
compounds in the present study. However, previous work studies.
has shown that not all enzymes are organic solvent tolerant As even low percentages of DMSO considerably interfered
(Jagota, 1992; Salleh et al., 2002). Thus, the influence of DMSO with AChE kinetics (Table 1), the influence of this co-solvent
on the kinetic parameters of AChE and the inhibitory potencies on the determination of the inhibitory activity of test com-
of seven reference AChE inhibitors was investigated for both pounds had to be evaluated. A substrate concentration close
colorimetric methods used, namely detection of the hydroly- to the apparent Km value obtained with the corresponding per-
sis of acetylthiocholine with Ellman’s reagent and detection centage of DMSO was chosen to assess the inhibitory activity
of the hydrolysis of ␣-naphthyl acetate with Fast Blue B salt of compounds for all subsequent experiments.
reagent.
3.1.2. Effect of DMSO on inhibitory potencies of reference
3.1.1. Influence of DMSO on the enzymatic activity of AChE inhibitors
AChE using acetylthiocholine and ˛-naphthyl acetate as To assess the influence of DMSO on the determination of
substrates inhibitory activities, pIC50 values of seven well known AChE
To assess the influence of DMSO on the enzymatic activ- inhibitors (Fig. 1) were determined in presence of 0.6% and
ity of AChE for the two substrates used (acetylthiocholine 1.6% of DMSO for both substrates and also in absence of
and ␣-naphthyl acetate) different percentages of co-solvent co-solvent when acetylthiocholine was used as substrate.
(0.6%, 1.6% and 2% final concentrations) were added to the Inhibitory potencies (pIC50 values) obtained with Ellman’s
reaction mixtures. The kinetic parameters Km and Vmax were method and with Fast Blue B salt as reagent are reported in
determined by means of the Michaelis–Menten rectangular Table 2 together with literature values. With the exception of
114 e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 3 3 ( 2 0 0 8 ) 109–119

Table 2 – Influence of DMSO at different final concentrations (0%, 0.6%, and 1.6%) on pIC50 values of reference AChE
inhibitors determined with microtitre plate methods using Ellman’s reagent and Fast Blue B salt
Compounds Ellman’s reagenta Fast Blue B salt as reagentb Literature pMIQ

0%c 0.6%c 1.6%c 0.6%c 1.6%c 0%c

Propidium (1) 4.66 ± 0.02 4.66 ± 0.17 4.37 ± 0.12 4.75 ± 0.17 4.71 ± 0.15 4.49 (Bolognesi et al., 2005) 8.00
Physostigmine (2) 6.07 ± 0.08 5.93 ± 0.10 5.61 ± 0.09 5.93 ± 0.07 5.86 ± 0.03 6.90 (Ishihara et al., 2000) 11.81
Galanthamine (3) 6.60 ± 0.03 6.46 ± 0.04 6.12 ± 0.04 5.88 ± 0.07 5.09 ± 0.08 6.44 (Guillou et al., 2000) 10.57
(±)-Huperzine A (4) 6.64 ± 0.06 6.45 ± 0.12 5.68 ± 0.06 6.42 ± 0.08 6.14 ± 0.05 12.08
(−)-Huperzine A (5) 7.43 ± 0.12 7.34 ± 0.11 6.21 ± 0.20 6.61 ± 0.10 5.34 ± 0.12 7.37 (Feng et al., 2005) 13.38
Tacrine (6) 7.53 ± 0.14 7.42 ± 0.13 7.34 ± 0.11 7.78 ± 0.05 7.77 ± 0.05 7.30 (Guillou et al., 2000) 12.07
Donepezil (7) 7.61 ± 0.10 7.50 ± 0.10 7.16 ± 0.08 7.55 ± 0.05 7.46 ± 0.07 7.40 (Martinez et al., 2000) 11.56

Comparison with literature data and pMIQ values obtained by TLC.


a
Means ± standard errors (n = 4).
b
Means ± standard errors (n = 2).
c
Percent of DMSO used.

physostigmine, which showed a lower activity in the present equal to 5) were considered to be inactive or not sufficiently
study than reported by Ishihara et al., pIC50 values determined active to be of interest as hit compounds for the development
in absence of DMSO were in good agreement with litera- of a novel AChE inhibitor. These activity limits were chosen
ture values. Moreover, for all known AChEIs, the influence of according to the pMIQ and pIC50 values of the reference AChE
DMSO was found to be significant. Indeed, all compounds were inhibitors currently in clinical use that were all above 10.5 and
slightly less active in presence of 0.6% of DMSO and the differ- 5, respectively (Table 2).
ences in pIC50 values became more pronounced in presence Fig. 3 shows the comparison of the results obtained with
of 1.6% of co-solvent. Nevertheless, the ranking of activities in the two screening tests for the 138 compounds investigated,
absence and in presence of DMSO is very similar, especially for i.e. pMIQ values determined by TLC versus percentages of inhi-
pIC50 values determined by Ellman’s method. Indeed, Spear- bition at a test compound concentration of 10−5 M determined
man rank order correlation coefficients were very close to 1 with Ellman’s method. For the majority of compounds (83% of
(rs = 0.96 between pIC50 values determined with 0% and 0.6% tested compounds), located in the white areas in Fig. 3, the
or 1.6% of DMSO using acetylthiocholine as substrate). It was results obtained with the two screening assays were conver-
thus concluded that the order of activities could be correctly gent. Indeed, the classification of these compounds, including
evaluated in presence of 1.6% of DMSO as co-solvent. Since all seven reference AChE inhibitors tested (full diamonds
␣-naphthyl acetate and test compounds were not soluble in
water, DMSO at a final concentration of 1.6% was therefore
selected as co-solvent for all further assays in solution, unless
stated otherwise.

3.2. Comparison of two screening tests: TLC assay


with Fast Blue B salt as reagent and microplate assay
using Ellman’s method

3.2.1. Comparison of results obtained with the two


screening tests
A priori, the two screening tests under investigation, i.e. the
TLC assay with Fast Blue B salt as reagent and the microplate
assay using Ellman’s method, should result in the identifi-
cation of the same compounds as active AChE inhibitors. To
evaluate whether this is actually the case, the first step was Fig. 3 – Comparison of pMIQ values obtained by TLC with
to define for each assay, when a compound was considered as percentages of inhibition at 10−5 M of test compound
active. With respect to the TLC assay, the activity limit was set determined using the microplate assay with Ellman’s
at a pMIQ (negative logarithm of the minimal inhibitory quan- reagent in presence of 1.6% of DMSO for 138 compounds
tity in mole that produced the spot with the least observable including 7 reference acetylcholinesterase inhibitors (). For
whiteness) of 10.5 and all compounds presenting a pMIQ above compounds located in the white areas ( and 䊉) the two
this cut-off value were considered as active. In the microplate assays gave convergent results; compounds located in the
assays, a compound that inhibited AChE to more than 50% at light grey area () resulted active by TLC and inactive with
a fixed test compound concentration of 10−5 M (corresponding the microplate assay; compounds in dark grey areas ( and
to a pIC50 value above 5) was classified as active. On the other ) showed to be active in solution and inactive by TLC; full
hand, compounds with inhibition percentages at 10−5 M less symbols represent compounds for which also pIC50 values
than or equal to 50 (corresponding to pIC50 values below or were established; data are means of n = 3.
e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 3 3 ( 2 0 0 8 ) 109–119 115

in white areas in Fig. 3) was the same by TLC as by Ell-


man’s method in solution. However, 20 compounds, i.e. about
15% of investigated compounds, classified as active with the
microplate assay, presented pMIQ values below 10.5 and were
considered as inactive by TLC (triangles in dark grey area in
Fig. 3). These compounds comprise 9 coumarins, 9 indenopy-
ridazines, 1 terpene and 1 chromone, and represent 63% of
compounds classified as active inhibitors by the microplate
assay. Hence, more than half of the hit compounds identi-
fied by the microplate assay were missed by TLC. Moreover, 3
compounds, 2 indenopyridazines and 1 aurone derivative, rep-
resenting about 2% of investigated compounds, were active in
the TLC assay but showed percentages of inhibition at a test
compound concentration of 10−5 M below 50 and were thus
considered as inactive with the microplate assay (inverted tri- Fig. 4 – Relationship between percentages of inhibition at
angles in light grey area in Fig. 3). It can be noted that the 10−5 M and pIC50 values determined with Ellman’s method
differences observed between the two assays involve almost in presence of 1.6% of DMSO for 20 selected compounds
all chemical compound classes investigated. Hence, these including 7 reference acetylcholinesterase inhibitors (), 6
method-dependent differences seem not related to a specific compounds for which the two assays gave convergent
compound class. results (䊉) and 7 compounds that showed to be active in the
The divergence in the identification of AChE inhibitors microplate assay and inactive by TLC (); data are means of
observed between the two screening assays was further inves- n = 2.
tigated. First, it was checked whether the percentage of
inhibition at a fixed test compound concentration of 10−5 M
allowed a correct prediction of the AChE inhibitory activ- 3.2.3. Correlation between the two colorimetric methods
ity, by comparing inhibition percentages with pIC50 values. in solution
Then, the influence of the two main differences between An investigation was then made to see if the two colorimet-
the experimental protocols of the two screening assays was ric methods used, namely detection of the hydrolysis of either
assessed: on the one hand the impact of the colorimet- ␣-naphthyl acetate with Fast Blue B salt as reagent or acetylth-
ric methods used, i.e. Fast Blue B salt as reagent versus iocholine with Ellman’s reagent, allow the identification of the
Ellman’s method, and on the other hand the effect of pre- same compounds as active AChE inhibitors when the tests
incubation of the test compound with/without AChE at 37 ◦ C were assayed in 96-well plates. For the seven reference AChEIs
for 20 min. Indeed, in the TLC assay, Fast Blue B salt is used (Fig. 1) this was the case. Indeed, with both methods, all ref-
as reagent and compounds are pre-incubated at 37 ◦ C for erence AChEIs except propidium showed pIC50 values above 5
20 min with AChE before substrate and reagent solutions are and were classified as active (Table 2), despite the slight dif-
sprayed onto the plate. In contrast, the microplate assay ferences in pIC50 values obtained with the two colorimetric
is based on Ellman’s method and no pre-incubation of test methods.
compounds with enzyme was performed before activity deter- In addition, inhibition percentages at a fixed concentration
minations. of 10−5 M determined by the two colorimetric methods were

3.2.2. Evaluation of the inhibition percentage at 10−5 M as


indicator of AChE inhibitory activity
The test set for the comparison between inhibition per-
centages at a fixed test compound concentration of 10−5 M
and pIC50 values consisted of the seven reference AChE
inhibitors, six compounds for which results obtained in the
two screening assays were convergent, seven compounds
active by Ellman’s method in solution but inactive by TLC
and the three compounds identified as active inhibitors by
TLC but inactive in the microplate assay. In Figs. 3–5, these
compounds are represented by full diamonds (), full cir-
cles (䊉), full triangles (), and full inverted triangles (),
respectively. For the three latter (), no pIC50 values could
be determined due to their low activity in solution. Fig. 4
shows the relationship between percentages of inhibition at a Fig. 5 – Correlation between percentages of inhibition at
fixed concentration of 10−5 M and pIC50 values. Within exper- 10−5 M determined in solution using Ellman’s reagent and
imental errors, both parameters allow the same classification Fast Blue B salt as reagent for 138 compounds including 7
of compounds and it can be concluded that inhibition per- reference acetylcholinesterase inhibitors (). Symbols and
centages allow a reliable prediction of the AChE inhibitory color code of areas are the same as in Fig. 3; data are means
activity. of n = 3, obtained in presence of 1.6% of DMSO.
116 e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 3 3 ( 2 0 0 8 ) 109–119

compared for the entire test set of 138 compounds. As shown IP derivatives (IP-42 to IP-45), for which convergent results
in Fig. 5, a reasonably good correlation was observed between were obtained in the two screening assays (located in the
values achieved using 96-well microtitre assays with Fast Blue white area in Fig. 3 and Table 3), were not or only to
B salt as reagent and Ellman’s reagent (r2 = 0.84). Indeed, for a certain extent affected by pre-incubation. In fact, these
94% of investigated compounds the separation of active hits compounds retained inhibition percentages above 50% after
from compounds not sufficiently active to be of interest was pre-incubation with/without AChE. On the other hand, for the
the same with both colorimetric methods. And in particular, ten compounds found active by Ellman’s method in solution
both methods in solution gave the same classification for all but inactive by TLC (C-45 to C-47, C-51, IP-34, IP-36 to IP-38, IP-
compounds that showed deviant results in the comparison 40 and IP-41, located in dark grey area) an important decrease
between the TLC assay with Fast Blue B salt as reagent and in activity was observed after pre-incubation, with result-
the microplate assay using Ellman’s method, represented in ing inhibition percentages below 50% (Table 3). Moreover, the
Fig. 5 by full triangles () for compounds that resulted active presence of AChE during pre-incubation did not influence
by Ellman’s method in solution but inactive by TLC, and by the loss of AChE inhibitory activity observed for these com-
full inverted triangles () for the three compounds identified pounds. Indeed, the same results were obtained after a 20 min
as active inhibitors by TLC but which were inactive in the pre-incubation period of compounds with AChE as after pre-
microplate assay. Hence, the divergences in the identification incubation for 20 min without enzyme. Hence, their loss of
of active AChE inhibitors observed between the TLC assay and activity during pre-incubation seems to be due to degrada-
the microplate assay cannot be explained by the different col- tion in aqueous buffer solutions of pH 7.4 and might to some
orimetric methods used. degree explain the divergences found between the TLC and
the microplate assays. Indeed, loss of activity for these ten
3.2.4. Effect of pre-incubation of test compounds compounds might also happen during the TLC procedure.
with/without AChE on their AChE inhibitory activity
Finally, pre-incubation of test compounds for 20 min at 37 ◦ C 3.2.4.2. Influence of pre-incubation of test compounds on their
either in presence or absence of AChE was investigated to see activity determined by the TLC assay. The influence of pre-
if this could explain the divergences in the identification of incubation was also investigated with the TLC assay, by
AChE inhibitors observed between the TLC and the microplate omitting the 20 min pre-incubation step at 37 ◦ C of the TLC
assays. Indeed, compounds were pre-incubated with AChE in standard protocol. However, the same results were found with
the TLC test before the addition of substrate whereas in solu- and without pre-incubation. Further, it has to be noted that for
tion no pre-incubation step was performed. The influence of all compounds only one spot was visible on the TLC plate after
pre-incubation of test compounds was first investigated in migration with hexane:ethyl acetate (1:1; v/v) as detected by
solution using Ellman’s method, then by TLC using Fast Blue UV and by means of a permanganate solution, indicating that
B salt as reagent. Experiments were performed for 20 com- potential degradation products could not be observed. Hence,
pounds representative of the entire test set, namely three the influence of pre-incubation of test compounds on their
reference AChEIs ((±) huperzine A (4), tacrine (6) and donepezil AChE activity observed in solution could not be reproduced by
(7)), four 5H-indeno [1,2-c] pyridazin-5-one (IP) derivatives TLC.
found active in both assays and located in the white area in
Fig. 3, 10 compounds (6 IP and 4 coumarin (C) derivatives) that 3.2.5. Hypotheses postulated to explain the differences
showed to be active in solution but inactive by TLC (located in observed
the dark grey area in Fig. 3), as well as the three compounds The 10 compounds found active in the microplate assay but
(2 IP and 1 aurone (AU) derivative) that resulted active by TLC inactive by TLC were found to lose their AChE inhibitory activ-
but inactive in the microplate assay (located in the light grey ity in aqueous buffer solutions at pH 7.4. Hence, lower stability
area in Fig. 3). might all the same be a possible reason for their inactivity
by TLC, even if loss of activity seems not to occur during the
3.2.4.1. Influence of pre-incubation of test compounds on their pre-incubation step. Inactivation after interactions of the com-
activity determined in solution with the microplate assay. The pounds with silica of the TLC plate may occur.
inhibitory activities (inhibition percentages at 10−5 M) of the Another, although less probable hypothesis, is that dif-
20 selected compounds, determined in solution with Ellman’s ferences between pMIQ values determined by TLC and
method either without or after a pre-incubation time of 20 min percentages of inhibition at 10−5 M obtained by Ellman’s
at 37 ◦ C in presence and in absence of enzyme, are reported in method in solution might be due to subtle differences in the 3D
Table 3 together with their pMIQ values determined by TLC. structures of the enzyme in the two assays. In fact, adsorption
First, it can be noted that the tested compounds were either of AChE onto silica plates might result in a change of con-
not affected or gave lower activities after pre-incubation. formation of the enzyme, and as a consequence, in altered
None of the compounds showed a higher activity after pre- accessibility of the enzyme for inhibitors. Indeed, a differ-
incubation with the enzyme. Hence, pre-incubation is not the ent kinetic behaviour of AChE between immobilized and free
reason for the high activity detected by TLC for the three states has been reported by several authors (Bartolini et al.,
compounds that were positive by TLC but inactive with the 2005; Sahin et al., 2005) and structural changes at the sec-
microplate assay (IP-11, IP-12 and AU-13, located in the light ondary and tertiary level upon adsorption onto silica surfaces
grey area in Fig. 3 and Table 3). were also reported for other enzymes (Koutsopoulos et al.,
A closer look at the results presented in Table 3 shows 2007). In the present case, adsorption of AChE onto silica would
that the activities of the three reference AChEIs and four result in a less favourable conformation for interaction with
e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 3 3 ( 2 0 0 8 ) 109–119 117

Table 3 – Effect of pre-incubation of test compounds with/without AChE at 37 ◦ C for 20 min on their AChE inhibitory
activity (color code of areas is the same as in Fig. 3)

compounds that showed to be inactive by TLC although they use of a co-solvent such as DMSO is avoided. A drawback of the
were identified as active hits in solution, and on the other TLC assay might be the possible interaction of either AChE or
hand, in a more favourable conformation for interaction with test compounds with the silica support, resulting in an alter-
the three compounds that were more active by TLC than with ation of the affinity of the enzyme for the compounds. Another
the microplate assay. point of concern is a potential loss of AChE inhibitory activity
of test compounds under the conditions used to develop the
3.2.6. Advantages and inconveniences of screening by TLC plate.
and microplate assays The general advantages of screening performed in a
As previously reported, the TLC assay is simple to use microplate are the easy handling of large amounts of sam-
and gives quick access to information concerning the AChE ples simultaneously, the possibility of automation and the
inhibitory activity in particular of plant extracts (Marston et need for little material. Indeed, both microtitre plate meth-
al., 2002). No expensive equipment is required and several ods used were shown to be reliable, rapid and accurate. To
plant extracts can be screened at the same time. Moreover, the screen libraries of synthetic or isolated natural compounds,
118 e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 3 3 ( 2 0 0 8 ) 109–119

microplate assays are therefore generally preferred over TLC solution has to be performed to confirm results obtained with
assays. A difficulty of enzyme assays performed in solution both screening assays.
might be the choice of a suitable co-solvent to solubilize test
compounds.
Regarding the colorimetric methods, Fast Blue B salt is
Supporting information available
without doubt a better choice than Ellman’s reagent for the
TLC assay, as it results in a strong purple coloration easily A table with molecular structures and detailed numerical
observable on the plate (Marston et al., 2002). However, for results (inhibition percentage at 10−5 M, pIC50 , pMIQ) of all
the microplate assay, Ellman’s method has several advantages tested compounds is available as supplementary information.
over the method using Fast Blue B salt as reagent and it can
be considered the colorimetric method of choice for medium-
Acknowledgments
and high-throughput inhibitor screening in solution. First, the
good solubility of the substrate acetylthiocholine in buffer, in
The authors express their gratitude to Prof. Angelo Carotti,
contrast to ␣-naphthyl acetate, allows a significant reduction
Dipartimento Farmaco-Chimico, Università di Bari, for the
of the final amount of DMSO in the reaction mixture. Moreover,
kind donation of the coumarin and indenopyridazine deriva-
stock solutions of substrate and reagent have a higher stability.
tives, and to Dr. Ahcène Boumendjel, Département de
Indeed, ␣-naphthyl acetate solutions have to be prepared just
Pharmacochimie Moleculaire, Faculté de Pharmacie de Greno-
before use whereas acetylthiocholine solutions can be kept for
ble, for having us kindly provided with the aurone and
several days. Finally, with Ellman’s method the formation of
azaaurone derivatives.
product of the enzymatic reaction can be monitored in a con-
tinuous manner, and the concentration of enzyme required is
lower. Appendix A. Supplementary data

Supplementary data associated with this article can be found,


4. Conclusion in the online version, at doi:10.1016/j.ejps.2007.10.004.

Results obtained in this work have shown that the percentages references
of inhibition at a fixed concentration of 10−5 M of test com-
pound using Ellman’s method are convergent with the pMIQ
values determined by TLC using Fast Blue B salt as reagent for
Bartolini, M., Cavrini, V., Andrisano, V., 2005. Batchwise covalent
the majority of the 138 compounds tested. However, about 15% immobilization of human acetylcholinesterase: kinetic and
of investigated compounds identified as hits in the microplate inhibition spectrophotometric studies. Anal. Biochem. 342,
assay were inactive by TLC, and on the other hand, about 2% of 163–166.
investigated compounds were active by TLC were inactive in Bolognesi, M.L., Andrisano, V., Bartolini, M., Banzi, R., Melchiorre,
the microplate assay. The differences observed between the C., 2005. Propidium-based polyamine ligands as potent
inhibitors of acetylcholinesterase and
two assays were not accredited to the different colorimetric
acetylcholinesterase-induced amyloid-beta aggregation. J.
methods used. In fact, both methods in solution allowed the
Med. Chem. 48, 24–27.
same classification for almost all compounds tested. More- Borloz, A., 2007. PhD Thesis, University of Geneva, 2007.
over, the inhibition percentages allowed a reliable prediction Boumendjel, A., Beney, C., Deka, N., Mariotte, A.-M., Lawson,
of the AChE inhibitory activity in solution. However, com- M.A., Trompier, D., Baubichon-Cortay, H., Di Pietro, A., 2002.
pounds identified as active inhibitors in the microplate assay 4-Hydroxy-6-methoxyaurones with high-affinity binding to
but inactive in the TLC test were shown to lose activity dur- cytosolic domain of P-glycoprotein. Chem. Pharm. Bull. 50,
854–856.
ing pre-incubation for 20 min at 37 ◦ C with/without AChE. This
Brühlmann, C., Ooms, F., Carrupt, P.A., Testa, B., Catto, M.,
loss of activity might also occur in the TLC assay during the Leonetti, F., Altomare, C., Carotti, A., 2001. Coumarins
development of the plate. Another hypothesis to explain the derivatives as dual inhibitors of acetylcholinesterase and
observed differences between the two screening assays is the monoamine oxidase. J. Med. Chem. 44, 3195–3198.
interaction of either AChE or test compounds with the silica Brühlmann, C., Marston, A., Hostettmann, K., Carrupt, P.A., Testa,
of the TLC plates, which might result in an altered affinity of B., 2004. Screening of non-alkaloidal natural compounds as
the enzyme for the compounds. acetylcholinesterase inhibitors. Chem. Biodivers. 1, 819–829.
Buenger, J., Ackermann, H., Jentzsch, A., Mehling, A., Pfitzner, I.,
In conclusion, both screening assays for AChE inhibitory
Reiffen, K.-A., Schroeder, K.-R., Wollenweber, U., 2006. An
activity investigated are considered suitable for the generation interlaboratory comparison of methods used to assess
of new hits. The TLC assay is advantageous for the screen- antioxidant potentials. Int. J. Cosmetic Sci. 28, 135–146.
ing of crude extracts of plants as it allows separation and Cavin, A.-L., Hay, A.-E., Marston, A., Stoeckli-Evans, H., Scopolliti,
detection of active compounds at the same time. This allows R., Diallo, D., Hostettmann, K., 2006. Bioactive diterpenes from
an easy follow up of the isolation and purification process. the fruits of Detarium microcarpum. J. Nat. Prod. 69,
768–773.
The microplate assay is more appropriate for the screening of
Cummings, J.L., 2004. Alzheimer’s disease. N. Eng. J. Med. 351,
libraries of synthetic and/or isolated natural compounds as it
56–67.
allows the possibility of automation and the easy handling of Di, L., Kerns, E.H., Li, S.Q., Carter, G.T., 2007. Comparison of
large amounts of samples. Finally, determination of IC50 val- cytochrome P450 inhibition assays for drug discovery using
ues and mode of inhibition with the microplate method in human liver microsomes with LC-MS, rhCYP450 isozymes
e u r o p e a n j o u r n a l o f p h a r m a c e u t i c a l s c i e n c e s 3 3 ( 2 0 0 8 ) 109–119 119

with fluorescence, and double cocktail with LC-MS. Int. J. Martinez, A., Fernandez, E., Castro, A., Conde, S.,
Pharm. 335, 1–11. Rodriguez-Franco, I., Banos, J.E., Badia, A., 2000.
Ellman, G.L., Courtney, D., Andres Jr., V., Featherstone, R.M., 1961. N-benzylpiperidine derivatives of 1,2,4-thiadiazolidinone as
A new and rapid colorimetric determination of new acetylcholinesterase inhibitors. Eur. J. Med. Chem. 35,
acetylcholinesterase activity. Biochem. Pharmacol. 7, 88–95. 913–922.
Feng, S., Wang, Z., He, X., Zheng, S., Xia, Y., Jiang, H., Tang, X., Bai, Michaelis, L., Menten, M.L., 1913. Die Kinetik der
D., 2005. Bis-huperzine B: highly potent and selective Invertinwirkung. Biochem. Z. 49, 333–369.
acetylcholinesterase inhibitors. J. Med. Chem. 48, 655–657. Motoyama, N., Hayaoka, T., Nomura, K., Dauterman, W.C., 1980.
Gnerre, C., Catto, M., Leonetti, F., Weber, P., Carrupt, P.A., Multiple factors for organophosphorus resistance in the
Altomare, C., Carotti, A., Testa, B., 2000. Inhibition of housefly, Musca domestica L. J. Pestic. Sci. 5, 393–402.
monoamine oxidases by functionalized coumarin derivatives: Newman, D.J., Cragg, G.M., 2007. Natural products as sources of
biological activities, QSARs, and 3D-QSARs. J. Med. Chem. 43, new drugs over the last 25 years. J. Nat. Prod. 70, 461–477.
4747–4758. Okombi, S., Rival, D., Bonnet, S., Mariotte, A.-M., Perrier, E.,
Gnerre, C., Thull, U., Gaillard, P., Carrupt, P.A., Testa, B., Boumendjel, A., 2006. Discovery of
Fernandes, E., Silva, F., Pito, M., Pinto, M.M.M., Wolfender, J.L., benzylidenebenzofuran-3(2H)-one (aurones) as inhibitors of
Hostettmann, K., Cruciani, G., 2001. Natural and synthetic tyrosinase derived from human melanocytes. J. Med. Chem.
xanthones as monoamine oxidase inhibitors: biological assay 49, 329–333.
and 3D-QSAR. Helv. Chim. Acta 84, Plummer, J.M., Greenberg, M.J., Lehman, H.K., Watts, J.A., 1983.
552–570. Competitive inhibition by dimethylsulfoxide of molluscan and
Guillou, C., Mary, A., Zafiarisoa Renko, D., Gras, E., Thal, C., 2000. vertebrate acetylcholinesterase. Biochem. Pharmacol. 32,
Potent acetylcholinesterase inhibitors: design, synthesis and 151–158.
structure–activity relationships of alkylene linked Racchi, M., Mazzucchelli, M., Porrello, E., Lanni, C., Govoni, S.,
bis-galanthamine and galanthamine–galanthaminium salts. 2004. Acetylcholinesterase inhibitors: novel activities of old
Bioorg. Med. Chem. Lett. 10, 637–639. molecules. Pharmacol. Res. 50, 441–451.
Hawkins, K.I., Knittle, C.E., 1972. Comparison of Ren, X., Han, Z., Wang, M., 2002. Mechanisms of monocrotophos
acetylcholinesterase determinations by the Michel and resistance in cotton bollworm Helicoverpa armigera (Hübner).
Ellman methods. Anal. Chem. 44, 416–417. Arch. Insect Biochem. Physiol. 51, 103–110.
Hostettmann, K., Borloz, A., Urbain, A., Marston, A., 2006. Natural Rhee, I.K., van de Meent, M., Ingkaninan, K., Verpoorte, R., 2001.
product inhibitors of acetylcholinesterase. Curr. Org. Chem. Screening for acetylcholinesterase inhibitors from
10, 825–847. Amaryllidaceae using silica gel thin-layer chromatography in
Howes, M.J.R., Houghton, P.J., 2003. Plants used in Chinese and combination with bioactivity staining. J. Chromatogr. A. 915,
Indian traditional medicine for improvement of memory and 217–223.
cognitive function. Pharmacol. Biochem. Behavior. 75, Rhee, I.K., Appels, N., Luijendijk, T., Irth, H., Verpoorte, R., 2003.
513–527. Determining acetylcholinesterase inhibitory activity in plant
Ingkaninan, K., Temkitthawon, P., Chuenchom, K., Yuyaem, T., extracts using a fluorimetric flow assay. Phytochem. Anal. 14,
Thongnoi, W., 2003. Screening for acetylcholinesterase 145–149.
inhibitory activity in plants used in Thai traditional Rollinger, J.M., Hornick, A., Langer, T., Stuppner, H., Prast, H.,
rejuvenating and neurotonic remedies. J. Ethnopharmacol. 89, 2004. Acetylcholinesterase inhibitory activity of scopolin and
261–264. scopoletin discovered by virtual screening of natural
Ishihara, Y., Goto, G., Miyamoto, M., 2000. Central selective products. J. Med. Chem. 47, 6248–6254.
acetylcholinesterase inhibitor with neurotrophic activity: Sahin, F., Demirel, G., Tümtürk, H., 2005. A novel matrix for the
structure-activity relationships of TAK-147 and related immobilization of acetylcholinesterase. Int. J. Biol. Macromol.
compounds. Curr. Med. Chem. 7, 341–354. 37, 148–153.
Jagota, S.K., 1992. Inhibition of acetylcholinesterase of mice Salleh, A.B., Basri, M., Taib, M., Jasmani, H., Rahman, R.N.Z.A.,
erythrocytes & synaptosomes by dimethylsulfoxide. Indian J. Rahman, M.B.A., Razak, C.N.A., 2002. Modified enzymes for
Med. Res. 96, 275–278. reactions in organic solvents. App. Biochem. Biotech. 102–103,
Kneubühler, S., Thull, U., Altomare, C., Carta, V., Gaillard, P., 349–357.
Carrupt, P.A., Carotti, A., Testa, B., 1995. Inhibition of Selkoe, D.J., 1996. Amyloid ␤-protein and the genetics of
monoamine oxidase-B by 5H-indeno[1,2-c]pyridazine Alzheimer’s disease. J. Biol. Chem. 271, 18295–18298.
derivatives: biological activities, quantitative Selwood, T., Feaster, S.R., States, M.J., Pryor, A.N., Quinn, D.M.,
structure–activity relationships (QSARs) and 3D-QSARs. J. 1993. Parallel mechanisms in acetylcholinesterase-catalyzed
Med. Chem. 38, 3874–3883. hydrolysis of choline esters. J. Am. Chem. Soc. 115,
Koutsopoulos, S., Patzsch, K., Bosker, W.T.E., Norde, W., 2007. 10477–10482.
Adsorption of trypsin on hydrophilic and hydrophobic Urbain, A., Marston, A., Queiroz, E.F., Ndjoko, K., Hostettmann, K.,
surfaces. Langmuir 23, 2000–2006. 2004. Xanthones from Gentiana campestris as new
Lam, K.S., 2007. New aspects of natural products in drug acetylcholinesterase inhibitors. Planta Med. 70, 1011–1014.
discovery. Trends Microbiol. 15, 279–289. Urbain, A., Marston, A., Hostettmann, K., 2005. Coumarins from
Marston, A., Kissling, J., Hostettmann, K., 2002. A rapid TLC Peucedanum ostruthium as inhibitors of acetylcholinesterase.
bioautographic method for the detection of Pharm. Biol. 43, 647–650.
acetylcholinesterase inhibitors in plants. Phytochem. Anal. van Asperen, K., 1962. A study of housefly esterases by means of
13, 51–54. a sensitive colorimetric method. J. Insect Physiol. 8, 401–416.
Martinez, A., Castro, A., 2006. Novel cholinesterase inhibitors as Whitehouse, P.J., Price, D.L., Struble, R.G., Clark, A.W., Coyle, J.T.,
future effective drugs for the treatment of Alzheimer’s Delon, M.R., 1982. Alzheimer’s disease and senile dementia:
disease. Expert Opin. Investig. Drugs 15, 1–12. loss of neurons in the basal forebrain. Science 215, 1237–1239.

S-ar putea să vă placă și