Abstract
Acetylcholinesterase (AChE) (EC 3.1.1.7) is an important enzyme that breaks down of acetylcholine in synaptic cleft in neuronal junctions. Inhibition of AChE is associated with treatment of several diseases such as Alzheimer’s disease (AD), myasthenia gravis, and glaucoma as well as the mechanisms of insecticide and anthelmintic drugs. Several AChE inhibitors are available in clinical use currently for the treatment of AD; however, none of them has ability, yet, to seize progress of the disease. Consequently, an extensive research has been going on finding new AChE inhibitors. In this sense, natural inhibitors have gained great attention due to their encouraging effects toward AChE. In this review, promising candidate molecules with marked AChE inhibition from both plant and animal sources will be underlined.
Keywords: Acetylcholinesterase, cholinesterase inhibition, neuroprotection, herbal, animal, microorganism
INTRODUCTION
Acetylcholinesterase (AChE) (EC 3.1.1.7), also known as acetylhydrolase, is a serine protease type of enzyme that catalyzes breaking down of acetylcholine (ACh) into choline and acetic acid and terminates neurotransmission at cholinergic synapses. The enzyme is known to have a fast catalytic activity with an ability to hydrolyze 25.000 ACh molecules per second [1]. Elucidation of the three-dimensional structure of AChE isolated from Torpedo californica (electric eel) (TcAChE) in 1991 by Sussman et al. accelerated the research on this enzyme, which further led to a series of theoretical and experimental studies [2]. AChE-coding sequences have been cloned up to now from a number of simple to complex organisms including insects, nematodes, fish, reptiles, birds, and several mammals including human being [3]. In fact, the crystal structures of AChE from various sources such as mouse, Drosophila, electric eel, and human being have been demonstrated to possess similar homology to each other at high rate [4-7]. The enzyme is present in the cholinergic junctions localized at central nervous system as well as peripheral sites such as erythrocytes, and muscles. The active site of AChE, also called the esteratic subsite lined by 14 conserved amino acid residues, consists of two separate ligand binding subsites: an acylation site at the bottom of the gorge where substrate is hydrolyzed and a peripheral anionic site (PAS) located at mouth of the gorge, both of which are critical to the breakdown of ACh [8,9]. Inhibitors of AChE tend to bind either to the active site or to the PAS, an allosteric site containing the indole of Trp279 as the principal component that serves to bind a molecule of ACh to the enzyme [10].
AChE possesses multifunctional properties. Needless to say, inhibition of AChE is the most popular as the treatment strategy against Alzheimer’s disease (AD), which is characterized by reduction in the activity of the cholinergic neurons according to cholinergic hypothesis [11,12]. Actually, AChE inhibitors constitute the only drug class approved by the U.S. Food and Drug Administration (FDA) for the treatment of AD at the moment. In addition to wide-range use of AChE inhibitors towards AD, certain chemical classes of pesticides, such as organophosphates and carbamates, work by interfering or inhibiting irreversibly this enyzme, which is expressed in all invertebrate and vertebrate animals as a key enzyme of the cholinergic system [13,14]. Another treatment approach for utilization of AChE inhibitors has been stated against myasthenia gravis (MG) through inhibition of peripheral AChE [15,16], while AChE inhibitors are also considered as the third line drugs for the treatment of glaucoma, which have been described to be better agents with fewer local and systemic adverse effects [17].
Due to its unique biochemical properties and physiological significance, AChE is an attractive topic of intensive investigations throughout the world. The research on discovering new inhibitors of AChE has been focused on the compounds of both synthetic and natural origins. In this review, a special focus will be reflected on promising compounds with AChE inhibitory effect from natural sources including plants, animals, and microorganisms along with a brief emphasize about the conventional AChE-inhibiting natural compounds already in use.
AChE Inhibitors of Plant Origin
The first example of a clinically-used natural compound inhibiting AChE was physostigmine (eserine), an alkaloid isolated from the seeds of Physostigma venenosa Balf. (Fabaceae), known as Calabar bean, to treat AD, MG, glaucoma, and delayed gastric emptying. Nonetheless, its short half-life, narrow therapeutic index as well as side effects such as nausea, vomiting, headaches, diarrhea, and dizziness led to its withdrawal from clinical use for this purpose [16,18,19]. Following physostigmine, cholinesterase inhibitors became the first line drugs used to treat the symptoms of AD in early 1990s and galanthamine, isolated from the bulbs of Galanthus woronowii Los. (Amaryllidaceae) (snowdrop), was the second plant-originated alkaloid with AChE inhibitory effect that has found a widespread clinical application after the synthetic drugs tacrine, rivastigmine, and donepezil [19-21]. Later on, huperzine A (HupA), the alkaloid type of AChE-inhibiting compound isolated in 1986 from the Chinese herb Huperzia serrata (Thunb. Ex Murray) Trev. (Lycopodiaceae/Huperziaceae), has been the most recent anti-AChE compound with highly promising clinical use in near future [22]. It is a reversible, potent, and selective inhibitor of this enzyme with a better therapeutic index than physostigmine and tacrine [23]. In fact, HupA (shuangyiping) is already in clinical use for symptomatic treatment of AD in China in a tablet form developed in 1996 and approved as the new drug by the State Administration of Traditional Chinese Medicine [24]. More details can be found on cholinesterase inhibitory activity of the aforementioned compounds in many excellent reviews that have been published up to date [12, 20, 25-28]. In addition to the known ones, the extensive research on plants have been still going on finding new inhibitors from the plant kingdom. Many herbal compounds from different chemical classes were identified with remarkable AChE inhibitory potentials as some examples can be found below:
Withanolides
Withania somnifera Dunal. (ashwaganda or Indian ginseng) (Solanaceae) is a popular plant used for improvement of memory and cognitive enhancement in Indian traditional medicine [29]. Withanolides, firstly isolated from W. somnifera, are the naturally-occurring steroid lactones having ergostane skeleton. Several studies have underlined marked AChE inhibitory activity of the withanolide derivatives. For instance; two new 6a,7a-epoxy-3b,5a,20b-trihydroxy-1-oxowitha-24-enolide (1) and 5b,6b-epoxy-4b,17a,27-trihydroxy-1-oxowitha-2,24-dienolide (2) and four known withanolides withaferin A (3), 2,3-dihydrowithaferin-A (4), 6a,7a-epoxy-5a,20b-dihydroxy-1-oxowitha-2,24-dienolide (5), and 5b,6b-epoxy-4b-hydroxy-1-oxowitha-2,14,24-trienolide (6) isolated from the methanol extract of W. somnifera collected in Karachi (Pakistan) were tested for their possible AChE inhibitory effect and only four of the compounds, whose IC50 values varied between 50±2.0 and 161.5±1.1 µM, were found to display moderate level of inhibition towards AChE as compared to galanthamine (IC50 = 0.50±0.001 µM) employed as the reference [30]. Among them, withaferin A (3) (Fig. 1), a known metabolite, was the most active in this assay (IC50 = 50±2.0 µM). Previously, administration of withaferin A in combination with several sitoindoside derivatives caused an augmentation in AChE activity in the lateral septum and globus pallidus in mice, whereas this combination reduced AChE activity in the vertical diagonal band [31].
Fig. (1).
Withaferin A.
By the same research group, three new withanolide derivatives (bracteosins A-C) were isolated from Ajuga bracteosa Wall ex Benth. (syn. A. remota) (Lamiaceae) of Pakistani origin and subjected to in vitro AChE inhibition assay [32]. The results revealed that bracteosins A-C (IC50 = 25.2±0.4, 35.2±0.5, and 49.2±0.8 µM, respectively) displayed a siginificant inhibitory action in a concentration-dependent manner (Fig. 2).
Fig. (2).
Bracteosins A-C.
In a related study by these researchers on the withanolide derivatives (1-6) isolated in their previous work [31], the active derivatives were further investigated against AChE by in silico methods using molecular docking simulations [33]. Molecular docking study findings indicated that all the ligands (the active compounds) were entirely embedded inside the aromatic gorge of AChE, whilst compounds 1, 3, and 5 extended up to the catalytic triad. Besides, Lineweaver-Burk and Dixon plots and their secondary replots revealed that compounds 1, 3, and 5 are the linear mixed-type inhibitors of AChE. Hence, these studies led to a statement that withanolides appear to be a new class of AChE inhibitors.
In a study published in 2007, Vinutha et al. reported that the root methanol and water extracts of W. somnifera from India exhibited 75.95±0.16% and 24.60±0.38% of inhibition against AChE, respectively, at concentration of 100 µM. The authors concluded that the active principles inhibiting AChE could be withanolides [34]. In a recent study, the crude methanol extract of the whole plant of W. somnifera collected from Khyber tribal area (Pakistan) together with its hexane, chloroform, ethyl acetate, butanol, and water fractions were tested in vitro against AChE and the crude methanol extract was found to have IC50 value at 76±0.16 µg/mL, while IC50 values of the chloroform, ethyl acetate, butanol, and water fractions were calculated as 69±0.20, 78±0.20, 97±0.23, and 111±0.14 µg/mL, respectively. The hexane fraction was inactive, whereas the most active one against AChE was the chloroform extract in comparison to the reference drug, galanthamine (IC50= 22.6±0.09 µg/mL) [35]. In a similar work [36], the aqueous extract of W. somnifera roots from India inhibited AChE in concentration-dependent fashion with maximum inhibition (86.7%) detected at the final assay concentration of 1 mg/mL. The kinetic studies revealed that the extract caused a mixed non-competitive inhibition towards the enzyme.
On the other hand, withanolide A (Fig. 3) showing strong AChE inhibition was applied to molecular docking studies and high-binding affinity of the ligand (withanolide A) to receptor was observed. Furthermore, in this computational study, long de novo simulations exerted interaction of the ligand with the residues Thr78, Trp81, Ser120, and His442 of human AChE [37].
Fig. (3).
Withanolide A.
Curcuminoids
The rhizomes of Curcuma longa L. (Zingiberaceae), known as turmeric, is indigenous to the Southeast Asian countries and used as culinary spice. The principal chemical ingredients in turmeric have been identified as curcuminoids consisting of curcumin (diferuloyl methane) (Fig. 4), bisdemethoxycurcumin (5%), and demethoxycurcumin (15%) and the dominant component in this mixture is curcumin (75-85%) [38].
Fig. (4).
Curcumin.
Curcumin has received a considerable attention from the scientists due to its strong antioxidant, anti-inflammatory, metal-chelator, anticancer, and other biological effects [39-43]. In addition, curcuminoids have been broadly studied for their neuroprotective properties using different experimental methods [44-48]. AChE inhibitory effect of curcuminoids were evaluated by Ahmed and Gilani and IC50 values of the curcuminoids were reported as 67.69 µM for curcumin, 16.84 µM for bisdemethoxycurcumin, and 33.14 µM for demethoxycurcumin [49]. The IC50 value was found as 19.67 µM for curcuminoids, which is the mixture of these three individual compounds. Since bisdemethoxycurcumin appeared to be more effective than the parent curcuminoids, the authors commented that the additional methoxy group (-OCH3) on the structure of bisdemethoxycurcumin may influence the level of AChE inhibitory activity. According to the literature survey, this was the only report on in vitro AChE inhibitory action of curcuminoids up to date. However, it is interesting to note that the ethanol extract of the rhizomes of C. longa of Pakistani origin was found to be totally inactive against AChE in an earlier study [50]. In another report, curcumin was tested against arsenic-induced neurotoxicity in rats, which causes cognitive deficits [51]. According to the results, curcumin reduced AChE activity from 46% to 41% in arsenic-treated rats. It was stated that curcumin significantly modulates arsenic-induced cholinergic dysfunctions in brain that may help to explain neuro-protective effect of this compound. In a similar work [52], curcumin was assessed in rats for its effects on aluminum-induced alterations in ageing-related parameters including AChE activity. The evidence showed that co-administration of curcumin and aluminum lowered the levels of lipid peroxidation, activities of protein kinase C and AChE.
Tanshinones
The roots of Salvia miltiorrhiza Bunge (Lamiaceae) are locally known as ‘Danshen’ in the traditional Chinese herbal medicine. Tanshinones, a mixture of diterpenoids including tanshinone I, dihydrotanshinone I, tanshinone IIA, and cryptotanshinone, are the major active ingredients of S. miltiorrhiza extract (Fig. 5) [53, 54].
Fig. (5).
Tanshinones; tanshinone I (A), Tanshinone IIA (B), dihydrotanshinone I (C), and cryptotanshinone (D).
A wide array of the reports pointed out to neuroprotective effect of S. miltiorrhiza (or danshen) as well as its individual components [55-59]. The acetone extract of danshen was tested in AChE inhibitory activity assay and activity-guided fractionation of the extract led to isolation of tanshinones (tanshinone I, dihydrotanshinone I, tanshinone IIA, and cryptotanshinone) with remarkable AChE inhibitory effects [60]. Among them, dihydrotanshinone I and cryptotanshinone displayed concentration-dependent inhibitory activities with IC50 values of 1.0 and 7.0 µM, respectively. Besides, in the same report, clogP values of tanshinones pointed out to potential of blood-brain barrier passing ability of these compounds. In a later study by Wong et al. [61], cryptotanshinone was also shown to reversibly inhibit AChE of human origin with IC50 value of 4.09 µM. In continuation with this report, the same research group investigated inhibitory characteristics of cryptotanshinone and dihydrotanshinone towards human AChE using molecular docking models and kinetic analysis, which led to the evidence that both of the compounds exerted a mixed non-competitive inhibition for human AChE, while they had mainly hydrophobic interactions with the allosteric site inside the active-site gorge of the enzyme [62]. The danshen ethanol extract and tanshinones were examined in another work for their AChE inhibitory effects using rat brain homo-genates as an enzyme resource [58]. The researchers concluded that the extract, total tanshinones as well as tanshinone I and dihydrotanshinone I exhibited remarkable in vitro inhibition on the enzyme, which was in accordance with the relevant previous reports. On the other hand, cryptotanshinone was again found to inhibit AChE activity for 3 h and 15, 16-dihydrotanshinone I for 6 h in ex vivo conditions [63].
Quercetin
Quercetin (Fig. 6), whose name was derived after its isolation from Quercus sp. (oak), is a flavonoid derivative widely found in plants and ubiquitously present in many foodstuffs such as mainly colored fruits, vegetables, wine, etc. It belongs to flavonol subtype of flavonoid family and also named as “bioflavonoid” due to its broad spectrum health effects [64-69].
Fig. (6).
Quercetin.
Moreover, quercetin has been reported to possess neuroprotective effects approved by different mechanisms and has recently become a more striking target for neurobiological research [70-75]. In our former study [76], we identified quercetin with a moderate in vitro inhibition against AChE (76.2±0.99%) at final concentration of 100 µg/mL and, then, we further investigated AChE inhibitory effect of four flavonoid and xanthone derivatives including quercetin and identified once more inhibition of AChE by quercetin in concentration-dependent fashion (IC50=353.86 µM) [77]. Our findings on enzyme kinetics also revealed that quercetin had a selective inhibition towards the enzyme in competitive manner. The molecular docking simulations indicated that the main phenylchromen part of quercetin binds with several hydrogen bonds to the important amino acid residues of PAS of AChE, while it had hydrophobic interactions with Ser125, His447, and Glu202, which are some of the prominent residues of PAS of AChE.
On the other hand, quercetin has been reported to be active principal of a number of extracts having AChE-inhibiting features. For instance; in a screening of 180 extracts obtained from the medicinal plants from Korea for their AChE inhibitory activity, the ethyl acetate extract of whole plant of Agrimonia pilosa Ledeb. (Rosaceae) yielded tiliroside, 3-methoxy quercetin, quercitrin, and quercetin, among which quercetin in particular showed twice more inhibition as compared to dehydroevodiamine (the reference compound in this assay) [78]. Among the components isolated from Polygonum sachalinensis F. Schmidt ex Maxim (Polygonaceae) collected in Switzerland, quercetin-3-O-β-D-galactopyranoside was revealed to inhibit the enzyme to some extent (55.6%), whereas quercetin-3-O-arabinopyranoside was inactive [79]. In fact, the activity of quercetin-3-O-β-D-galactopyranoside (IC50 = 0.068 mM) was not significantly different from that of its aglycone quercetin (IC50 = 0.060 mM). However, considering IC50 values of quercetin and quercetin-3-O-β-D-galactopyranoside, neither of them was comparable with galanthamine (IC50 = 0.0009 mM) employed as the reference, which was consistent with our previous study [77]. In a similar study [80], quercetin in addition to four other flavonoids isolated from the buds of Cleistocalyx operculatus (Roxb.) Merr. et Perry (Myrtaceae) was tested against AChE and found to inhibit this enzyme with IC50 value of 25.9 µM. A recent report by Sriraksa et al. demonstrated that the high dose of quercetin significantly decreased the AChE activity in the hippocampal homogenate in an animal model of Parkinson's disease [81]. Besides, Lee et al. suggested that quercetin might regulate α3,β4-nicotinic ACh receptor and show the pharmacological actions in nervous systems by this regulation as one of the mechanisms for quercetin [82].
AChE Inhibitors of Animal Origin
Fasciculins
Nature has gifted some animals with AChE-inhibiting components. Doubtlessly, the most interesting animal-based compounds that can potently inhibit reversibly AChE at picomolar level are fasciculins, the two toxins detected in the venom of the snake Dendroaspis angusticeps (green mamba) [83]. 1 g of this venom was found to contain approximately 40 mg of fasciculins, 2/3 of which was fasciculin 2 (FAS-II).
Fasciculins are members of the superfamily of three-fingered peptidic toxins from Elapidae venoms and structural analysis elucidated that fasciculin 1 (FAS-I) and FAS-II are the polypeptides consisting of 61 amino acid residues and four disulfides. The molecular weights were calculated as 6765 Da (FAS-I) and 6735 Da (FAS-II). Sequences of the two toxins were shown to differ only at one position by a replacement of Tyr with Asp/Asn. The aromatic residues, Trp286, Tyr72, and Tyr124, located at the enzyme have been shown to possess the most substantial influence on fasciculin binding, whereas Asp74, a charged residue in the vicinity of the binding site had little impact on binding by fasciculins [84]. After binding of fasciculin, elusive structural rearrangements of AChE were suggested to explain the observed residual catalytic activity of the fasciculin-enzyme complex [85]. Harel et al. demonstrated that the high affinity of FAS for AChE from electric eel was due to a remarkable surface complementarity, involving a large contact area and many residues either unique to FAS or rare in other three-fingered toxins [86]. In another study [87], the crystal structure of FAS-II-mouse AChE complex delineated a large contact area consistent with the low dissociation constant of the complex and only a few of the amino acid residues which make up the overall interactive site of the FAS-II provided strong interactions needed for high binding affinity and inhibition of the enzyme. In more publications, FASs were reported to bind to the gorge entrance of AChE with excellent complementarity through many polar and hydrophobic interactions and to inhibit AChE by combined allosteric and dynamical means [88-92].
Fasciculin 3 (FAS-III), another AChE inhibitor, was also isolated from the venom of D. polylepis (black mamba) and D. viridis (western green mamba), however, its full structure has not been elucidated up to date [93].
Onchidal
Onchidal (Fig. 7) is a natural toxin isolated from the Opisthobranch marine mollusc Onchidella binneyi (Onchidiacea) [94]. Considering its structural similarity to ACh, onchidal was investigated against AChE and explored to inhibit the enzyme in irreversible manner [95]. It was also stated by the authors that a potentially reactive α,β-unsaturated aldehyde group of onchidal may cause an irreversible inhibition of AChE, which result from formation of a novel covalent bond between the toxin and the enzyme and hence, this novel toxin could possibly be exploited as a new class of anticholinesterase agents.
Fig. (7).
Onchidal.
Ant Venom Alkaloids
Many ant species carry various defensive toxins and repellents to survive in the nature. The ant venom alkaloids have been shown to possess diverse biological activities including AChE inhibitory effect [96-100]. For instance; 2,5-dialkylpyrrolidines and pyrroline derivative alkaloids present in the venom of the ant Monomorium minutum were discovered to be strong and competitive inhibitors of AChE [100].
Gilatide
Gilatide is a peptide-type of substance consisting of 9 amino acid residues, which was obtained from a kind of reptile Heloderma suspectum, known as “Gila monster”. It has been discovered by a neuroscientist at Thomas Jefferson University in the year of 2001 [101] and then, developed further by Axonyx Pharmaceutical Inc. (USA), which is under patent protection [102]. Therefore, not much scientific information is available for this compound at the present time.
Sponge Metabolites
Sponges are the primitive marine animals from Porifera with about 5000 species known across the world, although approximately 150 species of them live in fresh waters [103]. Many species have been found to contain toxic substances, probably to discourage their predators. Among them, AChE-inhibiting substances have been reported from a variety of sponges. Several 3-alkylpyridinium oligomers isolated from the Adriatic sponge Reniera sarai were identified as the potent inhibitors of AChE [104]. In continuation of this work, structure of the active oligomer was revealed to compose of repeating subunits of N-butyl(3-butylpyridinium) [105]. The kinetic studies on this active oligomer indicated a reversible inhibition at first, and then a slow binding and irreversible binding finally. The same research group also isolated aplysamine-4 from an unidentified verongid sponge from the Red Sea and reported that aplysamine-4 tested against insect recombinant and electric eel AChE was a non-competitive reversible inhibitor of this enzyme [106].
The sponge Corticium sp. collected from Thailand waters was found to contain a new stigmastane-type steroidal alkaloid, named as 4-acetoxy-plakinamine B with a high AChE inhibitory effect (IC50 = 3.75±1.69 µM) [107]. The compound showed a mixed-competitive mode of inhibition. Another Thai sponge, Petrosia sp., was found to contain petrosamine (Fig. 8), a pyridoacridine alkaloid, with strong AChE inhibitory activity approximately six times higher than that of the reference galanthamine and quaternary ammonium group of petrosamine suggested to display a critical role in this inhibition [108].
Fig. (8).
Petrosamine.
In our recent report on screening of Turkish marine sponges collected from the Aegean and Mediterranean Sea, we identified oroidin (Fig. 9) isolated from Agelas oroides with a moderate level of AChE inhibition (55.21±2.94% at 200 µg/mL) [109].
Fig. (9).
Oroidin.
Arisugacins
Arisugacins A (Fig. 10) and B are the meroterpenoid-type of compounds isolated from the microfungi Penicillium sp. FO-4259, which were revealed to have strong AChE inhibition [110].
Fig. (10).
Arisugacin A.
In addition to arisugacins, territrems B and C (Fig. 11) and cyclopenin isolated from the same microfungus strain were also tested in the same manner. All isolated compounds exerted potent inhibitory activities against AChE with IC50 values in range of 1.0 approximately 25.8 nM. A recent computation docking study suggested that (+)-arisugacin A is a dual binding site covalent inhibitor of AchE with a unique mode of action [111].
Fig. (11).
Territrems B (R=OCH3) and C (R=OH).
CONCLUSION
Nature has great potential to provide bioactive compounds desirable for human health. Discovery of novel inhibitors of AChE is important in developing new drug candidates for the treatment of AD, MG, and glaucoma as well new insecticidal drugs. As emphasized in this review, plants, animals, and microorganisms absolutely have potential to produce natural compounds showing definite AChE-inhibiting features and these sources need to proceed to more advance research. Therefore, AChE inhibitors of natural origin may contribute to the design of new pharmaceuticals and bring in information which might assist to understanding of these processes acknowledging and their potential role in leading new treatments.
ACKNOWLEDGEMENTS
Declared none.
CONFLICT OF INTEREST
The author(s) confirm that this article content has no conflict of interest.
ABBREVIATIONS
- ACh
= Acetylcholine
- AChE
= Acetylcholinesterase
- AD
= Alzheimer’s disease
- FAS
= Fasciculin
- FDA
= U.S. Food and Drug Administration
- HupA
= Huperzine A
- IC
= Inhibitory concentration
- MG
= Myastenia gravis
- PAS
= Peripheric anionic site
- TcAChE
= Torpedo californica (electric eel) acetylcholinesterase
REFERENCES
- 1.Taylor P, Radic Z. The cholinesterases: from genes to proteins. Ann. Rev. Pharmacol. Toxicol . 1994;34: 281–320. doi: 10.1146/annurev.pa.34.040194.001433. [DOI] [PubMed] [Google Scholar]
- 2.Sussman JL, Harel M, Frolow F, Oefner C, Goldman A, Toker L, Silman I. Atomic structure of acetylcholinesterase from Torpedo californica a prototypic acetylcholine-binding protein. Science . 1991; 253:872–879. doi: 10.1126/science.1678899. [DOI] [PubMed] [Google Scholar]
- 3.Soreq H, Seidman S. Acetylcholinesterase - new roles for an old actor. Nature . 2001; 2: 294–302. doi: 10.1038/35067589. [DOI] [PubMed] [Google Scholar]
- 4.Bourne Y, Taylor P, Marchot P. Acetylcholinesterase inhibition by fasciculin: crystal structure of the complex. Cell . 1995;83: 503–512. doi: 10.1016/0092-8674(95)90128-0. [DOI] [PubMed] [Google Scholar]
- 5. Harel M, Kryger G, Rosenberry TL, Mallender WD, Lewis T, Fletcher RJ, Guss JM, Silman I, Sussman JL. Three-dimensional structures of Drosophila melanogaster acetylcholinesterase and of its complexes with two potent inhibitors. Protein Sci . 2000;9: 1063–1072. doi: 10.1110/ps.9.6.1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Kryger G, Harel M, Giles K, Toker L, Velan B, Lazar A, Kronman C, Barak D, Ariel N, Shafferman A, Silman I, Sussman JL. Structures of recombinant native and E202Q mutant human acetylcholinesterase complexed with the snake-venom toxin fasciculin-II. Acta Crystallogr. D Biol. Crystallogr . 2000; 56: 1385–1394. doi: 10.1107/s0907444900010659. [DOI] [PubMed] [Google Scholar]
- 7.Orhan IE, Khan MTH, Erdem SA, Kartal M, Sener B. Selective cholinesterase inhibitors from Buxus sempervirens L.and their molecular docking studies. Curr. Comp. Aided Drug Design . 2011; 7: 276–286. doi: 10.2174/157340911798260296. [DOI] [PubMed] [Google Scholar]
- 8.Silman I, Sussman JL. Acetylcholinesterase ‘classical’ and ‘non-classical’ functions and pharmacology. Curr. Opin. Pharmacol . 2005; 5: 293–302. doi: 10.1016/j.coph.2005.01.014. [DOI] [PubMed] [Google Scholar]
- 9.Johnson JL, Cusack B, Hughes TF, McCullough EH, Fauq A, Romanovski P, Spatola AF, Rosenberry TL. Inhibitors tethered near the acetylcholinesterase active site serve as molecular rulers of the peripheral and acylation sites. J. Biol. Chem . 2003; 278:38948–38955. doi: 10.1074/jbc.M304797200. [DOI] [PubMed] [Google Scholar]
- 10.Bourne Y, Radic Z, Sulzenbacher G, Kim E, Taylor P, Marchot P. Substrate and product trafficking through the active center gorge of acetylcholinesterase analyzed by crystallography and equilibrium binding. J. Biol. Chem . 2006; 281: 29256–29267. doi: 10.1074/jbc.M603018200. [DOI] [PubMed] [Google Scholar]
- 11.Delagarza V. Pharmacologic treatment of Alzheimer's disease: an update. Am. Fam. Phys . 2003; 68: 1365–1373. [PubMed] [Google Scholar]
- 12.Orhan IE. Current concepts on selected plant secondary metabolites with promising inhibitory effects against enzymes linked to Alzheimer’s disease. Curr. Med. Chem . 2012; 19: 2252–2261. doi: 10.2174/092986712800229032. [DOI] [PubMed] [Google Scholar]
- 13.Scholz NL, Truelove NK, Labenia JS, Baldwin DH, Collier TK. Dose-additive inhibition of chinook salmon acetylcholinesterase activity by mixtures of organophosphate and carbamate insecticides. Environ. Toxicol. Chem . 2006; 25: 1200–1207. doi: 10.1897/05-030r1.1. [DOI] [PubMed] [Google Scholar]
- 14.Lang GJ, Zhu KY, Zhang CX. Can acetylcholinesterase serve as a target for developing more selective insecticides? Curr. Drug Targets . 2012; 13: 495–501. doi: 10.2174/138945012799499712. [DOI] [PubMed] [Google Scholar]
- 15.Komloova M, Musilek K, Horova A, Holas O, Dohnal V, Gunn-Moore F, Kuca K. Preparation, in vitro screening and molecular modelling of symmetrical bis-quinolinium cholinesterase inhibitors - implications for early myasthenia gravis treatment. Bioorg. Med. Chem. Lett . 2011; 21: 2505–2509. doi: 10.1016/j.bmcl.2011.02.047. [DOI] [PubMed] [Google Scholar]
- 16.Mehta M, Adem A, Sabbagh M. New acetylcholinesterase inhibitors for Alzheimer's disease. Int. J. Alzheimers Dis. Article ID:728983. . 2012 doi: 10.1155/2012/728983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Hoyng PF, van Beek LM. Pharmacological therapy for glaucoma. a review. Drugs . 2000; 59: 411–434. doi: 10.2165/00003495-200059030-00003. [DOI] [PubMed] [Google Scholar]
- 18.Coelho F, Birks J. Physostigmine for Alzheimer's disease. Cochrane Database Syst. Rev . 2001; 2: 4D001499. doi: 10.1002/14651858.CD001499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Orhan G, Orhan I, Subutay-Oztekin N, Ak F, Sener B. Contemporary anticholinesterase pharmaceuticals of natural origin and their synthetic analogues for the treatment of Alzheimer's disease. Rec. Patents CNS Drug Disc . 2009; 4: 43–51. doi: 10.2174/157488909787002582. [DOI] [PubMed] [Google Scholar]
- 20.Orhan IE, Orhan G, Gurkas E. An overview on natural cholinesterase inhibitors - a multi-targeted drug class and their mass production. Mini Rev. Med. Chem . 2011; 11:836–842. doi: 10.2174/138955711796575434. [DOI] [PubMed] [Google Scholar]
- 21.Heinrich M, Teoh HL. Galanthamine from snowdrop - the development of a modern drug against Alzheimer’s disease from local Caucasian knowledge. J. Ethnopharmacol . 2004;92: 147–162. doi: 10.1016/j.jep.2004.02.012. [DOI] [PubMed] [Google Scholar]
- 22.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: 56–59. doi: 10.1016/j.neulet.2003.12.071. [DOI] [PubMed] [Google Scholar]
- 23.Ma X, Tan C, Zhu D, Gang DR, Xiao P. Huperzine A from Huperzia species - an ethnopharmacological review. J. Ethnopharmacol . 2007; 113: 15–34. doi: 10.1016/j.jep.2007.05.030. [DOI] [PubMed] [Google Scholar]
- 24.Tang XC. Huperzine A shuangyiping a promising drug for Alzheimer’s disease. Zhongguo Yao Li Xue Bao . 1996; 17: 481–484. [PubMed] [Google Scholar]
- 25.Howes MR, Houghton PJ. Plants used in Chinese and Indian traditional medicine for improvement of memory and cognitive function. Pharmacol. Biochem. Behav . 2003; 75: 513–527. doi: 10.1016/s0091-3057(03)00128-x. [DOI] [PubMed] [Google Scholar]
- 26.Mukherjee PK, Kumar V, Mal M, Houghton PJ. Acetylcholinesterase inhibitors from plants. Phytomedicine . 2007; 14: 289–300. doi: 10.1016/j.phymed.2007.02.002. [DOI] [PubMed] [Google Scholar]
- 27.Herrmann N, Chau SA, Kircanski I, Lanctôt KL. Current and emerging drug treatment options for Alzheimer's disease: a systematic review. Drugs . 2011; 71: 2031–2065. doi: 10.2165/11595870-000000000-00000. [DOI] [PubMed] [Google Scholar]
- 28.Perry E, Howes MJ. Medicinal plants and dementia therapy herbal hopes for brain aging? . CNS Neurosci.Ther . 2011; 17: 683–698. doi: 10.1111/j.1755-5949.2010.00202.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kulkarni SK, Dhir A. Withania somnifera an Indian ginseng. Progr. Neuro-Psychopharmacol. Biol. Psych . 2008;32: 1093–1105. doi: 10.1016/j.pnpbp.2007.09.011. [DOI] [PubMed] [Google Scholar]
- 30.Choudhary MI, Yousuf S, Nawaz SA, Ahmed S. Atta-ur-Rahman.Cholinesterase inhibiting withanolides from Withania somnifera. Chem. Pharm. Bull. 2004;52: 1358–1361. doi: 10.1248/cpb.52.1358. [DOI] [PubMed] [Google Scholar]
- 31.Schliebs R, Liebmann A, Bhattacharya SK, Kumar A, Ghosal S, Bigl V. Systemic administration of defined extracts from Withania somnifera (Indian ginseng) and shilajit difference affects cholinergic but not glutamatergic and GABAergic markers in rat brain. Neurochem. Int . 1997;30: 181–190. doi: 10.1016/s0197-0186(96)00025-3. [DOI] [PubMed] [Google Scholar]
- 32.Riaz N, Malik A, Aziz-ur-Rehman NawazSA, Muhammad P, Choudhary MI. Cholinesterase-inhibiting withanolides from Ajuga bracteosa. Chem. Biodivers . 2004; 1: 1289–1295. doi: 10.1002/cbdv.200490091. [DOI] [PubMed] [Google Scholar]
- 33.Choudhary MI, Nawaz SA, ul-Haq Z, Lodhi MA, Ghayur MN, Jalil S, Riaz N, Yousuf S, Malik A, Gilani AH, ur-Rahman A. Withanolides, a new class of natural cholinesterase inhibitors with calcium antagonistic properties. Biochem. Biophys. Res. Commun . 2005;334: 276–287. doi: 10.1016/j.bbrc.2005.06.086. [DOI] [PubMed] [Google Scholar]
- 34.Vinutha B, Prashanth D, Salma K, Sreeja SL, Pratiti D, Padmaja R, Radhika S, Amit A, Venkateshwarlu K, Deepak M. Screening of selected Indian medicinal plants for acetylcholinesterase inhibitory activity. J. Ethnopharmacol . 2007; 109:359–363. doi: 10.1016/j.jep.2006.06.014. [DOI] [PubMed] [Google Scholar]
- 35.Khan H, Tariq SA, Khan MA, ur-Rehman I, Ghaffar R. Saifullah.Cholinesterase and lipoxygenase inhibition of whole plant Withania somnifera. Afr. J. Pharm. Pharmacol . 2011; 5: 2272–2275. [Google Scholar]
- 36.Kumar S, Seal JC, Okello EJ. Kinetics of acetylcholinesterase inhibition by an aqueous extract of Withania somnifera roots. Int. J. Pharm. Sci. Res . 2011; 2: 1188–1192. [Google Scholar]
- 37.Grover A, Shandilya A, Agrawal V, Bisaria VS, Sundar D. Computational evidence to inhibition of human acetylcholinesterase by withanolide a for Alzheimer treatment. J. Biomol. Struct. Dyn . 2012; 29: 651–662. doi: 10.1080/07391102.2012.10507408. [DOI] [PubMed] [Google Scholar]
- 38.Esatbeyoglu T, Huebbe P, Ernst IM, Chin D, Wagner AE, Rimbach G. Curcumin - from molecule to biological function. Angew Chem. Int. Ed. Engl . 2012; 51: 5308–5332. doi: 10.1002/anie.201107724. [DOI] [PubMed] [Google Scholar]
- 39.Thephinlap C, Phisalaphong C, Fucharoen S, Porter JB, Srichairatanakool S. Efficacy of curcuminoids in alleviation of iron overload and lipid peroxidation in thalassemic mice. Med. Chem . 2009; 5: 474–482. doi: 10.2174/157340609789117912. [DOI] [PubMed] [Google Scholar]
- 40.Yu H, Li J, Shi K, Huang Q. Structure of modified e-polylysine micelles and their application in improving cellular antioxidant activity of curcuminoids. Food Funct . 2011; 2:373–380. doi: 10.1039/c1fo10053j. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Ramadan G, Al-Kahtani MA, El-Sayed WM. Anti-inflammatory and anti-oxidant properties of Curcuma longa (turmeric) versus Zingiber officinale (ginger) rhizomes in rat adjuvant-induced arthritis. Inflammation . 2011;34: 291–301. doi: 10.1007/s10753-010-9278-0. [DOI] [PubMed] [Google Scholar]
- 42.Rao KV, Samikkannu T, Dakshayani KB, Zhang X, Sathaye SS, Indap MA, Nair MP. Chemopreventive potential of an ethyl acetate fraction from Curcuma longa is associated with upregulation of p57(kip2) and Rad9 in the PC-3M prostate cancer cell line. Asian Pac. J. Cancer Prev . 2012; 13: 1031–1038. doi: 10.7314/apjcp.2012.13.3.1031. [DOI] [PubMed] [Google Scholar]
- 43.Ali BH, Marrif H, Noureldayem SA, Bakheit AO, Blunden G. Some biological properties of curcumin: a review. Nat. Prod. Commun . 2006; 1: 509–521. [Google Scholar]
- 44.Cole GM, Teter B, Frautschy FA. Neuroprotective effects of curcumin. Adv. Exp. Med. Biol . 2007; 595: 197–212. doi: 10.1007/978-0-387-46401-5_8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Dairam A, Limson JL, Watkins GM, Antunes E, Daya S. Curcuminoids, curcumin, and demethoxycurcumin reduce lead-induced memory deficits in male Wistar rats. J. Agric. Food Chem . 2007; 55: 1039–1044. doi: 10.1021/jf063446t. [DOI] [PubMed] [Google Scholar]
- 46.Ahmed T, Enam SA, Gilani AH. Curcuminoids enhance memory in an amyloid-infused rat model of Alzheimer's disease. Neurosci . 2010; 169: 1296–1306. doi: 10.1016/j.neuroscience.2010.05.078. [DOI] [PubMed] [Google Scholar]
- 47.Xu Y, Li S, Vernon MM, Pan J, Chen L, Barish PA, Zhang Y, Acharya AP, Yu J, Govindarajan SS, Boykin E, Pan X, O'Donnell JM, Ogle WO. Curcumin prevents corticosterone-induced neurotoxicity and abnormalities of neuroplasticity via 5-HT receptor pathway. J. Neurochem . 2011; 118: 784–795. doi: 10.1111/j.1471-4159.2011.07356.x. [DOI] [PubMed] [Google Scholar]
- 48.Matteucci A, Cammarota R, Paradisi S, Varano M, Balduzzi M, Leo L, Bellenchi GC, De Nuccio C, Carnovale-Scalzo G, Scorcia G, Frank C, Mallozzi C, Di Stasi AM, Visentin S, Malchiodi-Albedi F. Curcumin protects against NMDA-induced toxicity: a possible role for NR2A subunit. Invest. Ophthalmol. Vis. Sci . 2011; 52: 1070–1077. doi: 10.1167/iovs.10-5966. [DOI] [PubMed] [Google Scholar]
- 49.Ahmed T, Gilani AH. Inhibitory effect of curcuminoids on acetylcholinesterase activity and attenuation of scopolamine-induced amnesia may explain medicinal use of turmeric in Alzheimer's disease. Pharmacol. Biochem. Behav . 2009;91: 554–559. doi: 10.1016/j.pbb.2008.09.010. [DOI] [PubMed] [Google Scholar]
- 50.Khattak S, ur-Rehman S, Shah UK, Khan T, Ahmad M. In vitro enzyme inhibition activities of crude ethanolic extracts derived from medicinal plants of Pakistan. Nat. Prod. Res . 2005; 19: 567–571. doi: 10.1080/14786410410001721986. [DOI] [PubMed] [Google Scholar]
- 51.Yadav RS, Chandravanshi LP, Shukla RK, Sankhwar ML, Ansari RW, Shukla PK, Pant AB, Khanna VK. Neuroprotective efficacy of curcumin in arsenic induced cholinergic dysfunctions in rats. Neurotoxicol . 2011;32: 760–768. doi: 10.1016/j.neuro.2011.07.004. [DOI] [PubMed] [Google Scholar]
- 52.Sharma D, Sethi P, Hussain E, Singh R. Curcumin counteracts the aluminum-induced ageing-related alterations in oxidative stress, Na+, K+ ATPase and protein kinase C in adult and old rat brain regions. Biogerontol . 2009; 10:489–502. doi: 10.1007/s10522-008-9195-x. [DOI] [PubMed] [Google Scholar]
- 53.Zhou L, Zuo Z, Chow MS. Danshen an overview of its chemistry, pharmacology, pharmacokinetics, and clinical use. J. Clin. Pharmacol . 2005; 45: 1345–1359. doi: 10.1177/0091270005282630. [DOI] [PubMed] [Google Scholar]
- 54.Wang XH, Morris-Natschke SL, Lee KH. New developments in the chemistry and biology of the bioactive constituents of Tanshen. Med. Res. Rev . 2007; 27: 133–148. doi: 10.1002/med.20077. [DOI] [PubMed] [Google Scholar]
- 55.Liu J, Kuang P, Wu W, Zhang F, Liu J, Wan F, Huang Y, Ding A. Radix Salviae miltiorrhizae protects rat hippocampal neuron in culture from anoxic damage. J. Trad. Chin. Med . 1998; 18: 49–54. [PubMed] [Google Scholar]
- 56.Li LX, Dai JP, Ru LQ, Yin GF, Zhao B. Effects of tanshinone on neuropathological changes induced by amyloid ß-peptide1-40 injection in rat hippocampus. Acta Pharmacol. Sin . 2004; 25: 861–868. [PubMed] [Google Scholar]
- 57.Kim DH, Jeon SJ, Jung JW, Lee S, Yoon BH, Shin BY, Son KH, Cheong JH, Kim YS, Kang SS, Ko KH, Ryu JH. Tanshinone congeners improve memory impairments induced by scopolamine on passive avoidance tasks in mice. Eur. J. Pharmacol . 2007; 574: 140–147. doi: 10.1016/j.ejphar.2007.07.042. [DOI] [PubMed] [Google Scholar]
- 58.Zhou Y, Li W, Xu L, Chen L. In Salvia miltiorrhiza, phenolic acids possess protective properties against amyloid ß-induced cytotoxicity, and tanshinones act as acetylcholinesterase inhibitors. Environ. Toxicol. Pharmacol . 2011;31: 443–452. doi: 10.1016/j.etap.2011.02.006. [DOI] [PubMed] [Google Scholar]
- 59.Shi LL, Yang WN, Chen XL, Zhang JS, Yang PB, Hu XD, Han H, Qian YH, Liu Y. The protective effects of tanshinone IIA on neurotoxicity induced by ß-amyloid protein through calpain and the p35/Cdk5 pathway in primary cortical neurons. Neurochem. Int . 2012; 61: 227–235. doi: 10.1016/j.neuint.2012.04.019. [DOI] [PubMed] [Google Scholar]
- 60.Ren Y, Houghton PJ, Hider RC, Howes MJ. Novel diterpenoid acetylcholinesterase inhibitors from Salvia miltiorhiza. Planta Med . 2004; 70: 201–204. doi: 10.1055/s-2004-815535. [DOI] [PubMed] [Google Scholar]
- 61.Wong KK, Ho MT, Lin HQ, Lau KF, Rudd JA, Chung RC, Fung KP, Shaw PC, Wan DC. Cryptotanshinone, an acetylcholinesterase inhibitor from Salvia miltiorrhiza, ameliorates scopolamine-induced amnesia in Morris water maze task. Planta Med . 2010; 76: 228–234. doi: 10.1055/s-0029-1186084. [DOI] [PubMed] [Google Scholar]
- 62.Wong KK, Ngo JC, Liu S, Lin HQ, Hu C, Shaw PC, Wan DC. Interaction study of two diterpenes, cryptotanshinone and dihydrotanshinone, to human acetylcholinesterase and butyrylcholinesterase by molecular docking and kinetic analysis. Chem. Biol. Interact . 2010; 187:335–339. doi: 10.1016/j.cbi.2010.03.026. [DOI] [PubMed] [Google Scholar]
- 63.Kim DH, Jeon SJ, Jung JW, Lee S, Yoon BH, Shin BY, Son KH, Cheong JH, Kim YS, Kang SS, Ko KH, Ryu JH. Tanshinone congeners improve memory impairments induced by scopolamine on passive avoidance tasks in mice. Eur. J. Pharmacol . 2007; 574: 140–147. doi: 10.1016/j.ejphar.2007.07.042. [DOI] [PubMed] [Google Scholar]
- 64.Bischoff SC. Quercetin potentials in the prevention and therapy of disease. Curr. Opin. Clin. Nutr. Metab. Care . 2008; 11: 733–740. doi: 10.1097/MCO.0b013e32831394b8. [DOI] [PubMed] [Google Scholar]
- 65.Murakami A, Ashida H, Terao J. Multitargeted cancer prevention by quercetin. Cancer Lett . 2008; 269:315–325. doi: 10.1016/j.canlet.2008.03.046. [DOI] [PubMed] [Google Scholar]
- 66.Perez-Vizcaino F, Duarte J, Jimenez R, Santos-Buelga C, Osuna A. Antihypertensive effects of the flavonoid quercetin. Pharmacol. Rep . 2009; 61: 67–75. doi: 10.1016/s1734-1140(09)70008-8. [DOI] [PubMed] [Google Scholar]
- 67.Davis JM, Murphy EA, Carmichael MD. Effects of the dietary flavonoid quercetin upon performance and health. Curr. Sports Med. Rep . 2009; 8: 206–213. doi: 10.1249/JSR.0b013e3181ae8959. [DOI] [PubMed] [Google Scholar]
- 68.Chirumbolo S. The role of quercetin, flavonols and flavones in modulating inflammatory cell function. Inflamm. Allergy Drug Targets . 2010;9: 263–285. doi: 10.2174/187152810793358741. [DOI] [PubMed] [Google Scholar]
- 69.Mendoza EE, Burd R. Quercetin as a systemic chemopreventative agent: structural and functional mechanisms. Mini Rev. Med. Chem . 2011; 11: 1216–1221. doi: 10.2174/13895575111091216. [DOI] [PubMed] [Google Scholar]
- 70.Dajas F, Rivera-Megret F, Blasina F, Arredondo F, Abin-Carriquiry JA, Costa G, Echeverry C, Lafon L, Heizen H, Ferreira M, Morquio A. Neuroprotection by flavonoids. Braz. J. Med. Biol. Res . 2003;36: 1613–1620. doi: 10.1590/s0100-879x2003001200002. [DOI] [PubMed] [Google Scholar]
- 71.Schültke E, Kamencic H, Zhao M, Tian GF, Baker AJ, Griebel RW, Juurlink BH. Neuroprotection following fluid percussion brain trauma: a pilot study using quercetin. J. Neurotrauma . 2005; 22: 1475–1484. doi: 10.1089/neu.2005.22.1475. [DOI] [PubMed] [Google Scholar]
- 72.Silva B, Oliveira PJ, Dias A, Malva JO. Quercetin, kaempferol and biapigenin from Hypericum perforatum are neuroprotective against excitotoxic insults. Neurotox. Res . 2008; 13: 265–279. doi: 10.1007/BF03033510. [DOI] [PubMed] [Google Scholar]
- 73.Ossola B, Kääriäinen TM, Männistö PT. The multiple faces of quercetin in neuroprotection. Expert Opin. Drug Saf . 2009; 8:397–409. doi: 10.1517/14740330903026944. [DOI] [PubMed] [Google Scholar]
- 74.Zhang ZJ, Cheang LC, Wang MW, Lee SM. Quercetin exerts a neuroprotective effect through inhibition of the iNOS/NO system and pro-inflammation gene expression in PC12 cells and in zebrafish. Int. J. Mol. Med . 2011; 27: 195–203. doi: 10.3892/ijmm.2010.571. [DOI] [PubMed] [Google Scholar]
- 75.Pandey AK, Patnaik R, Muresanu DF, Sharma A, Sharma HS. Quercetin in hypoxia-induced oxidative stress: novel target for neuroprotection. Int. Rev. Neurobiol . 2012; 102: 107–146. doi: 10.1016/B978-0-12-386986-9.00005-3. [DOI] [PubMed] [Google Scholar]
- 76.Orhan I, Kartal M, Tosun F, Sener B. Screening of various phenolic acids and flavonoid derivatives for their anticholinesterase potential. Z. Naturforsch . 2007; 62: 829–832. doi: 10.1515/znc-2007-11-1210. [DOI] [PubMed] [Google Scholar]
- 77.Khan MT, Orhan I, Senol FS, Kartal M, Sener B, Dvorská M, Smejkal K, Slapetová T. Cholinesterase inhibitory activities of some flavonoid derivatives and chosen xanthone and their molecular docking studies. Chem. Biol. Interact . 2009; 181:383–389. doi: 10.1016/j.cbi.2009.06.024. [DOI] [PubMed] [Google Scholar]
- 78.Jung M, Park M. Acetylcholinesterase inhibition by flavonoids from Agrimonia pilosa. Molecules. 2007; 12: 2130–2139. doi: 10.3390/12092130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Fan P, Terrier L, Hay AE, Marston A, Hostettmann K. Antioxidant and enzyme inhibition activities and chemical profiles of Polygonum sachalinensis F. Schmidt ex Maxim (Polygonaceae). Fitoterapia . 2010; 81: 124–131. doi: 10.1016/j.fitote.2009.08.019. [DOI] [PubMed] [Google Scholar]
- 80.Min BS, Cuong TD, Lee JS, Shin BS, Woo MH, Hung TM. Cholinesterase inhibitors from Cleistocalyx operculatus buds. Arch. Pharm. Res . 2010;33: 1665–1670. doi: 10.1007/s12272-010-1016-5. [DOI] [PubMed] [Google Scholar]
- 81.Sriraksa N, Wattanathorn J, Muchimapura S, Tiamkao S, Brown K, Chaisiwamongkol K. Cognitive-enhancing effect of quercetin in a rat model of Parkinson's disease induced by 6-hydroxydopamine. Evid. Based Complement. Alternat. Med . 2012;Article ID:823206. doi: 10.1155/2012/823206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Lee BH, Hwang SH, Choi SH, Shin TJ, Kang J, Lee SM, Nah SY. Quercetin inhibits ?3,?4 nicotinic acetylcholine receptor-mediated ion currents expressed in Xenopus oocytes. Korean J. Physiol. Pharmacol . 2011; 15: 17–22. doi: 10.4196/kjpp.2011.15.1.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Karlsson E, Mbugua PM, Rodriguez-Ithurralde D. Fasciculins, anticholinesterase toxins from the venom of the green mamba Dendroaspis angusticeps. J. Physiol. (Paris) 1984; 79: 232–240. [PubMed] [Google Scholar]
- 84.Radic Z, Duran R, Vellom DC, Li Y, Cervenansky C, Taylor P. Site of fasciculin interaction with acetylcholinesterase. J. Biol. Chem . 1994; 269: 11233–11239. [PubMed] [Google Scholar]
- 85.Bourne Y, Taylor P, Marchot P. Acetylcholinesterase inhibition by fasciculin: crystal structure of the complex. Cell . 1995; 83: 503–512. doi: 10.1016/0092-8674(95)90128-0. [DOI] [PubMed] [Google Scholar]
- 86.Harel M, Kleywegt GJ, Ravelli RBG, Silman I, Sussman JL. Crystal structure of an acetylcholinesterase-fasciculin complex interaction of a three-fingered toxin from snake venom with its target. Structure Source . 1995;3: 1355–1366. doi: 10.1016/s0969-2126(01)00273-8. [DOI] [PubMed] [Google Scholar]
- 87.Marchot P, Bourne Y, Prowse CN, Bougis PE, Taylor P. Inhibition of mouse acetylcholinesterase by fasciculin crystal structure of the complex and mutagenesis of fasciculin. Toxicon Source. 1998;36: 1613–1622. doi: 10.1016/s0041-0101(98)00154-8. [DOI] [PubMed] [Google Scholar]
- 88.Le Du MH, Housset D, Marchot P, Bougis PE, Navaza J, Fontecilla-Camps JC. Structure of fasciculin 2 from green mamba snake venom: evidence for unusual loop flexibility. Acta Crystallogr. D Biol. Crystallogr . 1996; 52: 87–92. doi: 10.1107/S0907444995007517. [DOI] [PubMed] [Google Scholar]
- 89.Rodríguez-Ithurralde D, Vincent O. Fasciculins, enzyme site-directed polypeptides, as motor neurone research tools. J. Neurol. Sci . 1994; 124: 54–55. doi: 10.1016/0022-510x(94)90177-5. [DOI] [PubMed] [Google Scholar]
- 90.Cerveñanský C, Engström A, Karlsson E. Study of structure-activity relationship of fasciculin by acetylation of amino groups. Biochim. Biophys. Acta . 1994; 1199: 1–5. doi: 10.1016/0304-4165(94)90088-4. [DOI] [PubMed] [Google Scholar]
- 91.Cervenanský C, Engström A, Karlsson E. Role of arginine residues for the activity of fasciculin. Eur. J. Biochem . 1995; 229: 270–275. doi: 10.1111/j.1432-1033.1995.0270l.x. [DOI] [PubMed] [Google Scholar]
- 92.Tai K, Shen T, Henchman RH, Bourne Y, Marchot P, McCammon JA. Mechanism of acetylcholinesterase inhibition by fasciculin: a 5-ns molecular dynamics simulation. J. Am. Chem. Soc . 2002; 124: 6153–6161. doi: 10.1021/ja017310h. [DOI] [PubMed] [Google Scholar]
- 93.Harvey AL, Mckessy SP. Fasciculins In Handbook of Venoms and Toxins of Reptiles. CRC Press . 2009;pp:317–324. [Google Scholar]
- 94.Ireland C, Faulkner DJ. The defensive secretion of the opisthobranch mollusc. Onchidella binneyi. Bioorg. Chem . 1978; 7: 125–131. [Google Scholar]
- 95.Abramson SN, Radic Z, Manker D, Faulkner DJ, Taylor P. Onchidal a naturally occurring irreversible inhibitor of acetylcholinesterase with a novel mechanism of action. Mol. Pharmacol . 1989;36:349–354. [PubMed] [Google Scholar]
- 96.Jouvenaz DP, Blum MS, MacConnell JC. Antibacterial activity of venom alkaloids from the imported fire ant, Solenopsis invicta Buren. Antimicrob. Agents Chemother . 1972; 2: 291–293. doi: 10.1128/aac.2.4.291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Javors MA, Zhou W, Maas JW Jr, Han S, Keenan RW. Effects of fire ant venom alkaloids on platelet and neutrophil function. Life Sci . 1993; 53: 1105–1112. doi: 10.1016/0024-3205(93)90546-f. [DOI] [PubMed] [Google Scholar]
- 98.Kem WR, Wildeboer K, Le Francois S, Raja M, Marszalec W, Braekman JC. Nicotinic receptor inhibition by Tetraponera ant alkaloids. Cell Mol. Neurobiol . 2004; 24: 535–551. doi: 10.1023/B:CEMN.0000023628.65936.a7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Howell G, Butler J, Deshazo RD, Farley JM, Liu HL, Nanayakkara NP, Yates A, Yi GB, Rockhold RW. Cardiodepressant and neurologic actions of Solenopsis invicta (imported fire ant) venom alkaloids. Ann. Allergy Asthma Immunol . 2005;94:380–386. doi: 10.1016/S1081-1206(10)60991-X. [DOI] [PubMed] [Google Scholar]
- 100.Ronzani N, Lajat M. Acetylcholinesterase inhibition by alkaloids of the ant's venom Monomorium minutum. Bioorg. Med. Chem. Lett . 1995; 5: 1131–1132. [Google Scholar]
- 101.Badger D. Lizards A Natural History of Some Uncommon Creatures. Voyageur Press. USA . 2006:118. [Google Scholar]
- 102.http://www.google.com/patents/US20040092432. Access date 05.August. 2012.
- 103.Bond C, Harris AK. Locomotion of sponges and its physical mechanism. J. Exp. Zool . 1988; 246: 271–284. doi: 10.1002/jez.1402460307. [DOI] [PubMed] [Google Scholar]
- 104.Sepcic K, Guella G, Mancini I, Pietra F, Serra MD, Menestrina G, Tubbs K, Macek P, Turk T. Characterization of anticholinesterase-active 3-alkylpyridinium polymers from the marine sponge Reniera sarai in aqueous solutions. J. Nat. Prod . 1997; 60:991–996. doi: 10.1021/np970292q. [DOI] [PubMed] [Google Scholar]
- 105.Sepcic K, Marcel V, Klaebe A, Turk T, Suput D, Fournier D. Inhibition of acetylcholinesterase by an alkylpyridinium polymer from the marine sponge, Reniera sarai. Biochim. Biophys. Acta . 1998; 1387: 217–225. doi: 10.1016/s0167-4838(98)00125-3. [DOI] [PubMed] [Google Scholar]
- 106.Sepcic K, Mancini I, Vidic I, Franssanito R, Pietra F, Macek P, Turk T. Antibacterial and anticholinesterase activities of aplysamine-4, a bromotyrosine-derived metabolite of a Red Sea marine sponge. J. Nat. Toxins . 2001; 10: 181–91. [PubMed] [Google Scholar]
- 107.Langjae R, Bussarawit S, Yuenyongsawad S, Ingkaninan K, Plubrukarn A. Acetylcholinesterase-inhibiting steroidal alkaloid from the sponge Corticium sp. Steroids . 2007; 72: 682–685. doi: 10.1016/j.steroids.2007.05.005. [DOI] [PubMed] [Google Scholar]
- 108.Nukoolkarn VS, Saen-oon S, Rungrotmongkol T, Hannongbua S, Ingkaninan K, Suwanborirux K. Petrosamine, a potent anticholinesterase pyridoacridine alkaloid from a Thai marine sponge Petrosia n.sp. Bioorg. Med. Chem . 2008; 16: 6560–6567. doi: 10.1016/j.bmc.2008.05.027. [DOI] [PubMed] [Google Scholar]
- 109.Orhan IE, Ozcelik B, Konuklugil B, Putz A, Kaban UG, Proksch P. Bioactivity screening of the selected Turkish marine sponges and three compounds from Agelas oroides. Rec. Nat.Prod . 2012; 6:356–367. [Google Scholar]
- 110.Kuno F, Otoguro K, Shiomi K, Iwai Y, Omura S. Arisugacins A and B, novel and selective acetylcholinesterase inhibitors from Penicillium sp.FO-4259. I. Screning taxonomy, fermentation, isolation and biological activity. J. Antibiot.(Tokyo). 1996; 49: 742–747. doi: 10.7164/antibiotics.49.742. [DOI] [PubMed] [Google Scholar]
- 111.Al Rashid ZF, Hsung RP. (+)-Arisugacin A - Computational evidence of a dual binding site covalent inhibitor of acetylcholinesterase. Bioorg. Med. Chem. Lett . 2011; 21: 2687–2691. doi: 10.1016/j.bmcl.2010.12.041. [DOI] [PMC free article] [PubMed] [Google Scholar]











