ArticlePDF AvailableLiterature Review

Cholinesterase, a target of pharmacology and toxicology

Authors:

Abstract and Figures

Cholinesterases are a group of serine hydrolases that split the neurotransmitter acetylcholine (ACh) and terminate its action. Of the two types, butyrylcholinesterase and acetylcholinesterase (AChE), AChE plays the key role in ending cholinergic neurotransmission. Cholinesterase inhibitors are substances, either natural or man-made that interfere with the break-down of ACh and prolong its action. Hence their relevance to toxicology and pharmacology. The present review summarizes current knowledge of the cholinesterases and their inhibition. Particular attention is paid to the toxicology and pharmacology of cholinesterase-related inhibitors such as nerve agents (e.g. sarin, soman, tabun, VX), pesticides (e.g. paraoxon, parathion, malathion, malaoxon, carbofuran), selected plants and fungal secondary metabolites (e.g. aflatoxins), drugs for Alzheimer's disease (e.g. huperzine, metrifonate, tacrine, donepezil) and Myasthenia gravis (e.g. pyridostigmine) treatment and other compounds (propidium, ethidium, decamethonium). The crucial role of the cholinesterases in neural transmission makes them a primary target of a large number of cholinesterase-inhibiting drugs and toxins. In pharmacology, this has relevance to the treatment of neurodegenerative disorders.
Content may be subject to copyright.
219
Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 2011 Sep; 155(3):219–230. DOI 10.5507/bp.2011.036
© M. Pohanka
CHOLINESTERASES, A TARGET OF PHARMACOLOGY AND TOXICOLOGY
Miroslav Pohanka
Faculty of Military Health Sciences, University of Defense, Trebesska 1575, Hradec Kralove, Czech Republic
University Hospital Hradec Kralove, Sokolska 581, Hradec Kralove
E-mail: miroslav.pohanka@gmail.com
Received: February 24, 2011; Accepted with revision: June 3, 2011
Key words: Acetylcholinesterase/Butyrylcholinesterase/Alzheimer’s disease/Myasthenia gravis/Huperzine/Donepezil/
Rivastigmine/Galantamine
Background. Cholinesterases are a group of serine hydrolases that split the neurotransmitter acetylcholine (ACh)
and terminate its action. Of the two types, butyrylcholinesterase and acetylcholinesterase (AChE), AChE plays the key
role in ending cholinergic neurotransmission. Cholinesterase inhibitors are substances, either natural or man-made that
interfere with the break-down of ACh and prolong its action. Hence their relevance to toxicology and pharmacology.
Methods and Results. The present review summarizes current knowledge of the cholinesterases and their inhibi-
tion. Particular attention is paid to the toxicology and pharmacology of cholinesterase-related inhibitors such as nerve
agents (e.g. sarin, soman, tabun, VX), pesticides (e.g. paraoxon, parathion, malathion, malaoxon, carbofuran), selected
plants and fungal secondary metabolites (e.g. aflatoxins), drugs for Alzheimer’s disease (e.g. huperzine, metrifonate,
tacrine, donepezil) and Myasthenia gravis (e.g. pyridostigmine) treatment and other compounds (propidium, ethidium,
decamethonium).
Conclusions. The crucial role of the cholinesterases in neural transmission makes them a primary target of a large
number of cholinesterase-inhibiting drugs and toxins. In pharmacology, this has relevance to the treatment of neuro-
degenerative disorders.
INTRODUCTION
The cholinergic system is based on the neurotransmit-
ter acetylcholine (ACh), firstly recognized by Loewi in
1920s (ref.1) and found widely distributed in both cen-
tral and peripheral nervous systems. The two basic types
of acetylcholine receptors in the nervous system and at
neuromuscular junctions are: muscarinic acetylcholine
receptors (mAChR) and nicotinic acetylcholine recep-
tors (nAChR). Acetylcholine receptors are also found
expressed in multiple cells including endothelial and im-
mune system cells2.
Cholinesterases are a family of enzymes that kata-
lyse the hydrolysis of ACh into choline and acetic acid,
an essential process allowing for the restoration of the
cholinergic neuron. Cholinesterases are divided into two:
acetylcholinesterase (AChE; EC 3.1.1.7.) and butyrylcho-
linesterase (BuChE; EC 3.1.1.8). AChE participates in
cholinergic neurotransmission by hydrolyzing acetylcho-
line. It is expressed in nerve and blood cells. Compared to
AChE, the importance of BuChE is not well understood.
BuChE was known as plasmatic cholinesterase or pseu-
docholinesterase. Similarly, AChE was called blood, also
erythrocytal cholinesterase as its activity remains in the
cell mass after blood centrifugation. The name AChE de-
rives from the natural substrate acetylcholine as opposed
to BuChE that has no natural substrate. An absence or
mutation of BuChE leads to a medical condition (see
below) that shows itself only in the presence of some
drugs (e.g. succinylcholine) and toxins (e.g. cocaine)
(ref.3), due to its ability to split artificial substrates. This
review’s main focus is on cholinesterases as targets of
toxins and drugs. The biochemistry of AChE and BuChE
is also discussed. Drugs and toxins are divided in chapters
according to target sites.
BUTYRYLCHOLINESTERASE
Though BuChE activity is prevalent in the human
body, its’ physiological function is not completely under-
stood. BuChE deficient individuals are generally healthy
with no manifest signs of disease4. The case is similar for
mice with a damaged BuChE gene5,6. BuChE deficient
individuals have increased sensitivity to muscle relaxants
such as succinylcholine, resulting in lasting breath insuf-
ficiency7. Peoples with the deficient, K type BuChE have
lower plasma activity as well as lower affinity for succi-
nylcholine8. The K allele is widely-spread especially in
the Caucasus area. In recent publications, a link between
K type BuChE and lower incidence of Alzheimer’s disease
has been described9 However, more research is needed to
examine this connection. Regular BuChE is sensitive to
inhibition caused by dibucaine (or cinchocaine in some
sources) whereas AChE and K type BuChE are relatively
resistant to dibucaine. Biochemical examination of K type
BuChE is based on serum/plasma BuChE assessment
with and without dibucaine. The output is called a dibu-
caine number (DN). This represents the percentage of
inhibited BuChE. People with regular BuChE have a high
dibucaine number (DN ≥ 75), heterozygotes have medial
220 M. Pohanka
inhibition of DN ~ 40–70, and K type homozygotes have
nearly non-inhibited BuChE (DN < 20) (ref.10).
In comparison with AChE, BuChE is not constituted
in situ but in different organs, mainly in the liver11. BuChE
reaches serum levels of 5 mg/ml with a half time of 12
days12. Assay of BuChE activity in plasma can also serve
as a liver function test. BuChE activity decreases until
complex liver necrosis occurs. However, the importance
of BuChE as a liver function marker is limited by low
sensitivity. Genetic aspects (see above) or intoxication
with some compounds such as organophosphate pesti-
cides and/or organophosphonate nerve agents (see below)
are sources of false positive findings. BuChE is capable of
detoxifying a large number of exogenous substances: pro-
caine13, succinylcholine14, cocaine15, heroin, acetylsalicylic
acid16, and it can also protect the body from the impact
of organophosphorus AChE inhibitors17. However the pri-
mary reason for the existence of BuChE is still unknown.
As mentioned, BuChE, is named according to its pref-
erence for the artificial substrate butyrylcholine. BuChE
can split butyrylcholine with higher turnover number than
AChE. BuChE is also able to hydrolyze much slower than
AChE, indole derivatives18, adipoylcholine19, benzoylcho-
line20, acetylcholine/acetylthiocholine21,22 , butyrylcholine/
butyrylthiocholine23,24 and propionylcholine/propionylthi-
ocholine25,26. On the other hand, BuChE is not able to split
acetyl-β-methyl-thiocholine oracetyl-β-methyl-choline27,28
whereas AChE can. A summary of substrates and reaction
products is depicted below (Table 1).
The fact that BuChE has wider substrate specificity
than AChE is structurally determined. BuChE is a te-
trameric glycoprotein composed of four subunits. Both
dimeric and monomeric forms are stable and ubiquitous
in the body29. All subunits are identical and composed of
574 amino acids with an overall molecular weight close to
85 kDa. As described by Lockridge et al.30 the structural
similarity to AChE is 54% and to bovine thyroglobuline
28%, which leaves ample differences: BuChE is not in-
hibited by substrate excess as is typical for AChE (ref.31),
the active site is wider for BuChE (ref.32), BuChE is sen-
sitive to inhibition by tetraisopropyl pyrophosphoramide
(iso-OMPA; see text about AChE), and the inhibition is
considered to be a fast proof whether a sample contains
BuChE or AChE.
ACETYLCHOLINESTERASE
AChE and BuChE are similar, resembling each other
by more than 50% but their significance and localiza-
tion in the body are very different. AChE is expressed in
cholinergic neurons. Relatively high AChE activity can
also be found in blood cells responsible for the degrada-
tion of plasma acetylcholine33. The primary function of
AChE is rapid splitting of acetylcholine and terminating
cholinergic neurotransmission. Individuals with inhibited
AChE or knock out AChE mice have over-stimulated ace-
tylcholine receptors34. Although, AChE deficient mice are
viable, they have reduced musculature with changed mor-
phology35 and levels of extracellular acetylcholine nearly
sixty times higher than normal. It seems that BuChE is
able to partially recover the AChE missing activity in the
deficient animals36.
The structure of AChE has been extensively investigat-
ed since the 1990s. The first experiments were conducted
on AChE in the electric eel (Torpedo californica) due to
its availability37. This was also considered an informal
model until the commercialization of human recombinant
AChEs. The AChE active site as well as the whole AChE
structure is evolutionary conservative and it contains
common regions similar to the other serine hydrolases.
Cholinesterases are a type α/β hydrolase folded with an
α helix bound with β sheet that contains a catalytic do-
main38 with catalytic triad Ser – His – Glu, the same as
in AChE, BuChE, and lipases. A similar structure can
be also found in carboxyesterases where glutamate is re-
Table 1. Selected substrates and products of AChE respectivelly BuChE catalyzed hydrolysis.
Substrate Product Enzyme Reference
Succinylcholine + water succinate + choline BuChE 14
Adipoylcholine + water adipoate + choline BuChE 19
Benzoylcholine + water benzoiate + choline BuChE 20
Acetylcholine + water acetate + choline AChE > BuChE 21
Acetylthiocholine + water acetate + thiocholine AChE > BuChE 22, 67
Butyrylcholine + water butyrate + choline BuChE > AChE 23
Butyrylthiocholine + water butyrate + thiocholine BuChE > AChE 24, 61, 67
Propionylcholine + water propionate + choline BuChE, AChE 25
Propionylthiocholine + water propionate + thiocholine BuChE, AChE 26, 67
Acetyl-β-methyl-thiocholine + water β-metyl-thiocholine + acetate AChE 27
Acetyl-β-methyl-choline + water β-methyl-choline + acetate AChE 28
221
Cholinesterases, a target of pharmacology and toxicology
placed by aspartate39. Serine is a part of a stable sequence
Gly-Glu(His)-Ser-Gly-Ala/Gly (ref.40).
The electric eel AChE’s active site lies on a bottom of
long and narrow cavity 20 Å deep. The active site con-
tains a catalytic triad within an esteratic site with amino
acid positions for the electric eel AChE: Ser 200, His
440 and Glu 327 (ref.41). The anionic site (also α-anionic
site) is another part of the active site and it is close to the
esteratic site. The anionic site is composed of the amino
acids Trp 86, Tyr 337 and Phe 338 for murine AChE
(ref.42) or Trp 84, Tyr 121 and Phe 330 for the electric eel
AChE (ref.43,44). While the esteratic site hydrolyzes the
ester bond, the anionic site interacts with the acetylcho-
line quaternary ammonium atom and is responsible for
its correct orientation. Entry into the active site through
the cavity composed of aromatic amino acids, i.e. aro-
matic gorge, enables higher selectivity for acetylcholine.
Substrate penetration is allowed by cation – π interactions
between acetylcholine quaternary ammonium atom and
π electrons of phenylalanine, tryptophan and tyrosine
aromatic cores45,46.
The peripheral anionic site (also β-anionic site) is lo-
calized on the AChE surface around the cavity entrance.
This site was recognized as a target for multiple AChE
activity modulators and the first experiments began in the
1960s (ref.47). The aromatic site contains loops and it has
good conformational flexibility. Tyr 70, Asp 72, Tyr 121,
Trp 279 and Tyr 334 amino acids residues are the most
significant residues in the peripheral anionic site48. As
described in the following chapters, the peripheral anionic
site is a target for a number of toxins and also promis-
ing drugs49,50. It probably plays an important role in the
development of Alzheimer’s disease. Amyloid β peptide
interacts with the peripheral anionic site resulting in the
formation of amyloid plaques and consequent damage to
cholinergic neurons51.
Both AChE and BuChE form mainly tetramer G4 but
they can also form dimmer G2 that can be secreted as
a water soluble molecule52. Monomeric AChE has mo-
lecular weight of 69 kDa (ref.53). The predominant part
of AChE localized in the central nervous system con-
tains botgh hydrophilic and hydrophobic regions i.e. it is
amphiphilic. There are differences between amphiphilic
and non-amphiphilic cholinesterases. The amphiphiphilic
cholinesterase contains G4 catalytic tetramer and one
non-catalytic subunit P (ref.54). The P subunit has a mo-
lecular weight 20 kDa and it is asymmetrically bound to
two G subunits. From a chemical point of view, it is glyco-
phosphatidylinositol (GPI) called GPI anchor (ref.55,56).
In the human AChE, the liphophilic part of the GPI an-
chor is palmitate57. The biological role of individual AChE
forms can be ascertained from an experiment done on
monkey brains58. 85% of AChE are tetramers with a sedi-
mentation constant 9.7 S, 10% is dimeric (5.7 S) and 5%
monomeric (3.2 S). In total, 83% AChE molecules are
amphiphilic and only 17% hydrophilic. Besides free and
membrane bound AChE, there is also collagen bound
AChE. From the symbols introduced by Massoulie and
Bon, bound AChE is abbreviated AChET and one collagen
oligomer is connected with one (A4), two (A8) or three
(A12) tetrameric AChE molecules59,60.
AChE and BuChE have different abilities to split sub-
strates. Compared to BuChE, AChE is not able to hydro-
lyze high molecular weight esters butAChE has higher
affinity for acetylcholine and BuChE for butyrylcholine.
The differences in affinity to substrate are probably caused
by changes in the aromatic gorge disposition. Substitution
of two phenylalanines to leucine and valine in the elec-
tric eel AChE aromatic gorge possessed butyrylcholine
turnover rate at a similar level to BuChE. Further, mu-
tated AChE was sensitive to inhibition by iso-OMPA and
was not inhibited by propidium, a peripheral anionic site
inhibitor61. Differences in AChE and BuChE structures
are revealed by huperzine A. This Alzheimer’s disease
drug is a strong AChE inhibitor binding to the peripheral
anionic site; however, BuChE intacts in the presence of
huperzine62. Another Alzheimer’s disease drug, tacrine,
binds into the α-anionic site. It inhibits AChE as well as
BuChE to a comparable degree63. A similar situation to
tacrin is common for other drugs containing quaternary
ammonium, nerve agents, and neurotoxic pesticides64-67.
The basic parameters for AChE and BuChE are summa-
rized below (Table 2).
Table 2. Basic parameters of AChE and BuChE.
AChE BuChE References
Subunit 69 kDa 85 kDa 29, 30, 53
Quaternary structure Tetramer predominate, mono-, di-
and trimeric forms can also occur 29, 54, 55, 66, 68, 69
Conversion of acetyl-β-methyl-(thio)choline High low 27, 28, 71, 72
Inhibition by excess of substrate Yes no 68, 69, 70
Inhibition by iso-OMPA* No yes 61, 75
Inhibition by nerve agents Yes yes 65, 66
Inhibition by huperzine Yes no 62
*Iso-OMPA: tetraisopropyl pyrophosphoramide
222 M. Pohanka
Apropos AChE enzymology, there are differences be-
tween AChE and BuChE. Inhibition by excess of substrate
is probably the most important fact68,69. The biological
significance of AChE inhibition by substrate excess has
not yet been proven. One theory however looks feasible70.
After acetylcholine vesicles are released into the neuro-
synaptic cleft, AChE is inhibited and the receptors are
stimulated. The termination signal, which has a square
plot in cholinergic nerves, is fast once the ACh level drops
below threshold concentration. The basic biochemical
test for estimating whether a sample contains AChE and
not BuChE is based on assessment of acetylthiocholine
and acetyl-β-methyl-thiocholine as fast conversion in low
concentrations of substrate and slow conversion in high
concentration71,72 Specific inhibitors introduced later are
also applicable. Non-enzymatic functions of AChE are
probable and the research is on-going. Interestingly, body
growth and cell adhesion are probably partially connected
with AChE (ref.73,74).
CHOLINESTERASE INHIBITORS
Cholinesterase inhibitors (e.g. drugs, natural toxins,
pesticides, chemical warfare agents) are a wide group of
chemical compounds with different physico-chemical
properties. AChE inhibitors play a significant role in
the biochemical processes of the human body due to
the physiological importance of AChE. Specific BuChE
inhibitors, such as iso-OMPA (Fig. 1), have mainly diag-
nostic importance75. Lower interest in BuChE inhibitors
can be explained by probable BuChE physiological redun-
dancy. For example, drugs suppressing the manifestation
of Alzheimer’s disease through impact on the cholinergic
system are predominantly selective inhibitors of AChE
(ref.76). Selective inhibitors of BuChE have also been in-
vestigated as potential drugs for Alzheimer’s disease77;
but to a lesser degree than AChE. Inhibition of AChE
also plays an important role in nerve agent toxicology.
Intact BuChE however can temporarily substitute inhib-
ited AChE and is able to slowly hydrolyze accumulated
acetylcholine78.
3. Compounds bound at the peripheral (β) anionic site
(e.g. huperzine, propidium).
Inhibitors binding at the esteratic part of active site
Inhibitors of the esteratic part found on the active
site are compounds with the chemical structure of or-
ganophosphorus or carbamate derivatives. Inhibitors of
esteratic subsite of the active centre are mainly toxins,
chemical warfare agents or pesticides. Of course some
are used as drugs. These compounds interact with ser-
ine in the catalytical triad of active site, providing sta-
ble esters. Organophosphorus compounds create stable,
covalently bound adducts with spontaneous dissociation
once covalently connected with serine hydroxyl. Some
drugs containing the oxime group are able to split or-
ganophoshorus moiety from the active site resulting in
liberation and enzyme reactivation. Obidoxime, trime-
doxime, pralidoxime (2-PAM) and asoxime (HI-6) can
be mentioned as commercially available drugs3. After
a specific time interval (from minutes up hours) for each
organophosphorus inhibitor, the bound inhibitor under-
goes dealkylation called “aging” (ref. 80). The aging has
no beneficial effect on the enzyme as it remains inactive.
In comparison with organosphosphorus inhibitors, the
carbamate moiety is spontaneously hydrolyzed and liber-
ated. AChE becomes active again. Carbamates are prob-
ably able to bind through non-covalent interactions81. The
mechanism of AChE inhibition by an organophosphonate
is depicted below (Fig. 2).
Fig. 1. Tetraisopropyl pyrophosphoramide (iso-OMPA).
Compounds inhibiting AChE can be divided into three
basic groups79:
1. Compounds binding at the active site interact with
either esteratic (e.g. nerve agents) or anionic site (e.g.
tacrine).
2. Compounds interacting with the aromatic gorge (e.g.
decamethonium).
Fig. 2. Inhibition of AChE by nerve agent sarin (reaction
1) and the consequent aging (reaction 2). Serine
hydroxyl is indicated in AChE molecule by an
abbreviation (Ser-OH).
Nerve agents are organophosphonate compounds
used in chemical warfare. The older group of nerve agents
called G series was discovered before World War II. Tabun
was the first known nerve agent first synthesized in 1936
by professor Gerhard Schrader. After World War II, the
most toxic nerve agents called the V series were exten-
sively investigated82. Tabun (abbreviated GA according to
NATO), sarin (GB), soman (GD) and cyclosarin (GF)
are representatives of the G series nerve agents. Among
VX nerve agents are the Russian VX (VR) and Chinese
(VC) variants83. The chemical structures of selected nerve
agents are depicted in (Fig. 3).
223
Cholinesterases, a target of pharmacology and toxicology
Fig. 3. Selected nerve agents.
Nerve agents are extremely hazardous due to superior
penetration ability into the human body by all routes and
their high toxicity. Nerve agents differ from pesticides in
their toxicity and rapid bodily dissemination The median
lethal dose (LD50) is different for individual nerve agents:
e.g. for subcutaneous administrations to rat, the LD50s
are 193 μg/kg for tabun, 103 μg/kg for sarin, 75 μg/kg for
soman, and 12 μg/kg for VX (ref.84-86). Median lethal con-
centration and time (LCt50) in rats for sarin is 150 mg/m3
for ten minutes lasting inhalation87. The toxicity of nerve
agents is much more apparent than standard pesticides:
e.g. the commercially available organophosphate pesticide
primiphos-methyl (e.g. preparation Actellic 50EC) has
a declared LD50 for rat males and per oral administration
≥ 1,500 mg/kg.
The G series of nerve agents penetrate the body by all
routes and spread quickly through the organism. In com-
parison with G series, the V series of nerve agents are able
to penetrate via the lungs and skin with ease; however,
V agents create sub-epithelial reservoirs and the agent is
slowly released from the reservoirs88. It should be empha-
sized that nerve agents as well as organophosphate and
carbamate pesticides are quite reactive. Apart from bind-
ing to AChE and BuChE, they can bind to multiple organs
and tissues in the body. Inhibition of AChE is the most
crucial from the toxicological point of view whereas from
the therapeutic point of view the most significant fact is
the inhibition of carboxylesterase 1. Carboxylesterase 1 is
able to recover its activity even after sarin inhibition. This
fact encourages scientists in the search for an effective
scavenger that would serve as a prophylactic against nerve
agent intoxication89. The other effective enzyme is serum
paraoxonase (PON). The PON is able to split organo-
phosphate and thereby detoxify the poisoned person. On
the other hand, PON activity is quite low and fluctuates
greatly in the general population90.
The less toxic variant of organophosphonate nerve
agents are organophosphate pesticides. Highly toxic
organophosphate pesticides are e.g. paraoxon ethyl,
paraoxon methyl, and malaoxon. These compounds are
approximately equally toxic to warm-blooded as well as
cold-blooded organisms. Due to the effort to enhance pes-
ticide specificity, numerous derivatives of highly toxic pes-
ticides have been prepared to reduce the toxicity towards
warm-blooded organisms and retain toxicity to insects.
Thioforms of organophosphates such as parathion ethyl,
parathion methyl and malathion are some relevant exam-
ples. The thioforms of organophosphate pesticides are
converted into the above mentioned oxoforms by mixed
function oxidases (MFO). The activation proceeds in
cold-blooded organisms but this is not common in warm-
blooded organisms where no metabolizing or dealkylation
into non toxic compound take place91.
Carbamates are the second group of pesticides inhib-
iting cholinesterases. From the chemical point of view,
they are N-alkyl and N,N-dialkyl carbamates. The natural
derivate of carbamate is physostigmine. It is produced as
a secondary metabolite in the African plant Physostigma
venenosum (Fabaceae). Physostigmine is a strong revers-
ible inhibitor of AChE. It has broad use in Myasthenia
gravis treatment as it increases acetylcholine levels in the
damaged neurosynaptic clefts and also as a prophylac-
tic to nerve agent exposure as it blocks the irreversible
binding of nerve agents92,93. Carbamates are pseudo-
irreversible inhibitors of cholinesterases; the carbamoyl
moiety can be split from cholinesterase by spontaneous
hydrolysis94. Carbamates cannot penetrate the blood
brain barrier in the healthy body; however, stress con-
ditions can enhance diffusion into the central nervous
system95. Organophosphate and carbamate compounds
are not only used in agriculture but for medical purposes
too. Rivastigmine is a drug available for the symptomatic
treatment of Parkinson’s as well as Alzheimer’s disease96.
Trichlorfon (metrifonate) has similar application in medi-
cine to rivastigmine though it was used as a pesticide in
the past97.
The majority of countries have strong regulations on
the application of pesticides; e.g. in the European Union it
is regulated by the directive 91/41/EHS. Individual prepa-
rations are approved for commercialization and the list is
regularly updated. Commonly used and relatively safe for
warm-blooded organisms, are mainly pesticides: organo-
phosphates – chlorpyrifos, fenitrothion, pirimiphos-me-
thyl, dimethoate, phosalone and carbamates – pirimicarb,
224 M. Pohanka
Fig. 4. Selected organophosphate and carbamate inhibitors of cholinesterases.
carbofuran, carbosulfan, methiocarb, fenoxycarb. The
structures of selected organophosphate and carbamate
compounds are shown below (Fig. 4).
Inhibitors of the α-anionic site
Cholinesterase inhibitors binding to the α-anionic
site are a group of chemical compounds containing cer-
tain common motives. Firstly, these compounds typi-
cally contain condensed aromatic cores. Secondly, there
should be quarternary ammonium or nitrogen included
as a heteroatom. Acrdines and tetrahydroacridines can be
mentioned as examples. Quinolines and isoquinolines are
other common structures interacting with the α-anionic
site of cholinesterases. In comparison with the esteratic
site inhibitors, compounds interacting with the α-anionic
site are reversible inhibitors. 9-amino-1,2,3,4,-tetrahy-
droacridine known as tacrine, which is also considered
one of the most important inhibitors of the α-anionic site
able to suppress Alzheimer’s disease manifestation. It is
marketed worldwide under the trade name Cognex. The
main disadvantage of tacrine is its relatively high hepato-
toxicity98. There is an effort underway to find less toxic
derivatives of tacrine99. In the past, 7-methoxytacrine
was extensively investigated as a promising substitute
to tacrine. This compound is less toxic than tacrine and
some in vitro as well as in vivo tests proved superior to
tacrine100,101. Protoberbrine alkaloids are strong natural
inhibitors of AChE. Berberine, palmatine, jatrorrhizine
and epiberberine are examples. These substances are con-
sidered promising drugs for Alzheimer’s disease sympto-
matic treatment102.
Galantamine (Nivalin) is another well known drug
interacting with the α-anionic site. It is an alkaloid
from the Caucasian snowdrop (Galanthus woronowii,
Amaryllidaceae). The properties of galantamine were
firstly recognized by Mashkovsky and Kruglikova-Lvova in
the 1950s (ref.103). Beside the α-anionic site, galantamine
also binds at another important part of the AChE active
site including aromatic gorge104,105. The formulas of these
compounds are shown in (Fig. 5).
Inhibitors binding into aromatic gorge
The aromatic gorge is not a typical target for
cholinesterase inhibitors. On the other hand, inhibitors
interacting with the α-anionic site will probably also in-
teract with the aromatic gorge. Galantamine can be men-
tioned as an example (see above). In silico methods and
structural analyses have shown that some bisquarternary
compounds such as the depolarizing muscle relaxant
decamethonium (Fig. 6) provide a stable complex with
225
Cholinesterases, a target of pharmacology and toxicology
Fig. 5. Selected inhibitors that bind onto the α-anionic site.
Fig. 6. Decamethonium (anions are not considered).
Fig. 7. Selected structures that bind to peripheral anionic site (anions are not
considered).
the aromatic gorge due to electrostatic interaction106. But
the main decamethonium effect is not on AChE (ref.107).
Inhibitors of peripheral (β) anionic site
The peripheral anionic site is the main target of many
pharmacologically important compounds rather than
toxins. Much attention is given to the peripheral anionic
site due to the link to Alzheimer’s disease. Lack of ace-
tylcholine was considered as a major factor in the cause
of Alzheimer’s disease (AD). The deposition of amyloid
plaque in AD may be accelerated or even triggered by
interaction of β-amyloid with the peripheral anionic
site. Inhibitors binding at the peripheral anionic site are
considered not only symptomatic drugs for Alzheimer’s
disease, but also probably causative ones108. It should be
emphasized though that the etiology of Alzheimer’s dis-
ease is not thoroughly understood and the actual function
of AChE is still being investigated.
Aflatoxins are natural hepatocarcinogens activated
by liver cytochrome P450. They probably interact with
the peripheral anionic site. Despite strong inhibition of
AChE, it seems that BuChE has no sensitivity to aflatoxin
as shown for aflatoxin B1 (ref.109-111). Inhibition of AChE
was confirmed after the onset of cholinergic symptoms
following aflatoxin exposure112. The mechanism of aflatox-
in interaction with AChE is not well-explained and more
supporting experiments are needed. Double-stranded
DNA fluorescence dyes are also inhibitors of AChE.
Propidium113 as well as ethydium114 are proven inhibitors,
binding at the peripheral anionic site. The structure of
alfatoxin B1, ethidium and propidium are depicted below
(Fig. 7).
The effects of certain ions on AChE remain unclear.
Oxidative state III+ aluminum ions have been investigated.
X
XX
226 M. Pohanka
REFERENCES
1. Loewi O. Uberhumerole ubertragbarkeit der herznervenwirkung.
I Mitteilung Pflugers Arch 1921;189:239-42.
2. Wessler I, Kirkpatrick CJ. Acetylcholine beyond neurons: the
non-neuronal cholinergic system in humans. Br J Pharmacol
2008;154:1558-71.
3. Bajgar J. Organophosphates/nerve agent poisoning: mechanism
of action, diagnosis, prophylaxis, and treatment. Adv Clin Chem
2004;38:151-216.
4. Manoharan I, Boopathy R, DArvesh S, Lockridge O. A medial
health report on individuals with silent butyrylcholinesterase in
Vysya community of India. Clin Chim Acta 2007;378:128-35.
5. Li B, Duysen EG, Carlson M, Lockridge O. The butyrylcholines-
terase knockout mouse as a model for human butyrylcholinesterase
deficiency. J Pharmacol Exp Ther 2008;324:1146-54.
6. Duysen EG, Li B, Lockridge O. The butyrylcholinesterase knock-
out mouse a research tool in the study of drug sensitivity, bio-
distribution, obesity and Alzheimer’s disease. Expert Opin Drug
Metab Toxicol 2009;5:523-8.
7. Yen T, Nightingale BN, Burns JC, Sullivan DR, Stewart PM.
Butyrylcholinesterase (BCHE) genotyping for post-succinylcholine
apnea in an Australian population. Clin Chem 2003;49:1297-1308.
8. Hashim Y, Shepherd D, Wiltshire S, Holman R, Levy JC, Clark
A, Cull CA. Butyrylcholinesterase K variant on chromosome 3 q
is associated with Type II diabetes in white Caucasian subjects.
Diabetologia 2001;44:2227-30.
9. Holmes C, Ballard C, Lehmann D, Smith AD, Beaumont H, Day
IN, Khan MN, Lovestone S, McCulley M, Morris CM, Munoz DG,
O’Brien K, Russ C, Del Ser T, Warden D. Rate of progression of
cognitive decline in Alzheimer’s disease effect of butyrylcholineste-
rase K gene variation. J Neurol Neurosurg Psychiatry 2005;76:640-
3.
10. Kallow W, Genest K. A method for the detection of human se-
rum cholinesterase: determnation of dibucaine numbers. Can J
Biochem 1957;35:339-46.
11. Iwasaki T, Yoneda M, Nakajima A, Terauchi Y. Serum butyrylcho-
linesterase is strongly associated with adiposity, the serum lipid
profile and insulin resistance. Intern Med 2007;46:1633-9.
12. Ostergaard D, Viby-Moogensen J, Hanel HK, Skovgaard LT.
Half-life of plasma cholinesterase. Acta Anaesthesiol Scand
1988;32:266-9.
13. Yuan J, Yin J, Wang E. Characterization of procaine metabolism
as probe for the butyrylcholinesterase enzyme investigation by si-
multaneous determination of procaine and its metabolite using
capillary electrophoresis with electrochemiluminescence detection.
J Chromatogr A 2007;1154:368-72.
14. Zelinski T, Coghlan G, Mauthe J, Triggs-Raine B. Molecular basis
of succinylcholine sensitivity in a prairie Hutterite kindred and
genetic characterization of the region containing the BCHE gene.
Mol Genet Metab 2007;90:210-16.
15. Duysen EG, Li B, Carlson M, Li YF, Wieseler S, Hinrichs SH,
Lockridge O. Increased hepatotoxicity and cardiac fibrosis in
cocaine-treated butyrylcholinesterase knockout mice. Basic Clin
Pharmacol Toxicol 2008;103:514-21.
16. Kolarich D, Weber A, Pabst M, Stadlmann J, Teschner W, Ehrlich
H, Schwarz HP, Altmann F. Glycoproteomic characterization of
butyrylcholinesterase from human plasma. Proteomics 2008;8:254-
63.
17. Saxena A, Sun W, Luo C, Myers TM, Koplovitz I, Lenz DE, Doctor
BP. Bioscavenger for protection from toxicity of organophosphorus
compounds. J Mol Neurosci 2006;30:145-8.
18. Rozengart EV, Basova NE, Suvorov AA, Khovanskikh AE.
Indophenol chromogenic substrates of cholinesterases of differ-
ent origin. J Evol Biochem Physiol 2002;38:16-23.
19. Grigoryan H, Halebyan G, Lefebvre B, Brasme B, Masson
P. Mechanism of hydrolysis of dicholine esters with long polymeth-
ylene chain by human butyrylcholinesterase. Biochim Biophys Acta
2008;1784:1818-24.
20. Saeed A, de Boeck S, Debruyne I, Wouters J, Stockx J. Hen’s egg
yolk cholinesterase. Purification, characterization and comparison
Fig. 8. Alzheimer disease drugs binding into peripheral
anionic site.
Aluminum can act as a neurotoxin and it has been inves-
tigated as the causative agent of Alzheimer’s disease for
many years; however, no decisive connection has been
found115,116 . Contradictory changes in brain AChE activ-
ity after aluminum exposure have been found in different
brain regionss117,118 . On the other hand, AChE is not only
affected by aluminum. Double-valent ions also have the
ability to inhibit AChE activity. Magnesium ions are also
an AChE inhibitor119.
The peripheral anionic site is a target of newly synthe-
sized drugs for Alzheimer’s disease treatment. Inhibition
of the peripheral anionic site in Alzheimer’s disease has
probably not only symptomatic effects due to enhance-
ment of acetylcholine availability. It can also slow down
the deposition of amyloid plaque120,121. Inhibition of the
peripheral anionic site can be considered the most prom-
ising for Alzheimer’s disease treatment. Drugs that bind
at the peripheral (e.g. donepezil, huperzine) as well as at
the -anionic site (e.g. tacrine, galantamine) cause elevated
expression of AChE. In contrast, inhibitors binding at
the esteratic site do not cause significant expression of
AChE (ref.122 ). The up-regulation of AChE expression is
an unwanted effect that lowers treatment efficacy. The
commercially available drugs for Alzheimer’s disease that
inhibit AChE through the peripheral anionic site are e.g.
alkaloids huperzine A as well as B and man-made done-
pezil. Huperzine comes from the firmoss Huperzia serrata
(Huperziaceae) (ref.123,124). Huperzine A is a more potent
AChE inhibitor than huperzine B. Huperzine can be iso-
lated from the H. serrata that contains above 0.025 (w/w)
of huperzine A and less huperzine B; synthetic production
is available as well125. Donepezil is another drug suitable
for Alzheimer’s disease treatment with good penetration
through the blood brain barrier and slow excretion. It is
marketed under the trade name Aricept126. The structure
of huperzine and donepezil is depicted in (Fig. 8).
CONCLUSION
Cholinesterases play an important role in the human
body. They are a regular target of a large number of tox-
ins including chemical warfare agents. However, the cur-
rent focus of investigation is development of Myasthenia
gravis and Alzheimer’s disease drugs. Understanding of
cholinesterase structure and the biological mechanism
of their inhibition is necessary for novel effective drug
development.
227
Cholinesterases, a target of pharmacology and toxicology
with hen’s liver and blood plasma cholinesterase. Biochim Biophys
Acta 1980;614:389-99.
21. Giacobini E. Cholinesterases: new roles in brain function and in
Alzheimer’s disease. Neurochem Res 2003;28:515-22.
22. Grigoryan HA, Hambardzumyan AA, Mkrtchyan MV, Topuzyan
VO, Halebyan GP, Asatryan RS. Alpha, beta-dehydrophenylalanine
choline esters, a new class of reversible inhibitors of human ace-
tylcholiensterse and butyrylcholinesterase. Chem Biol Interact
2008;171:108-16.
23. Debord J, Laubarie C, Dantoine T. Microcalorimetric study of the
inhibition of butyrylcholinesterase by carbamates. Anal Biochem
2008;373:247-52.
24. Kamal MA, Klein P, Luo W, Li Y, Holloway HW, Tweedie D, Greig
NH. Kinetics of human serum butyrylcholinesterase inhibition by
a novel experimental Alzheimer therapeutic, dihydrobenzodiox-
epine cymserine. Neurochem Res 2008;33:745-53.
25. Nagasawa T, Sugisaki H, Tani Y, Ogata K. Purification and
characterization of butyrylcholine-hydrolyzing enzyme from
Pseudomonas polycolor. Biochim Biophys Acta 1976;429:817-27.
26. Monteiro M, Quintaneiro C, Morgado F, Soares AM, Guilhermino
L. Characterization of the cholinesterases present in head tissues
of the estuarine fish Pomatoschistus microps: application to bio-
monitoring. Exotoxicol. Environ. Saf. 2005;62:341-7.
27. Plageman LR, Puletti GM, Skau KA. Characterization of ace-
tylcholinesterase in Caco-2 cells. Exp Biol Med (Maywood)
2002;227:480-6.
28. Zhukovskii YG. On establishment of individuality of the cholien-
sterase enzyme in the studied preparation. J Evol Biochem Physiol
2003;39:281-90.
29. Blong RM, Bedows E, Lockridge O. Tetramerization domain of
human butyrylcholinesterase is at the C-terminus. Biochem J
1997;327:747-57.
30. Lockridge O, Bartels CF, Vaughan TA, Wong CK, Norton SE,
Johnson LL. Complete amino acid sequene of human serum cho-
linesterase. J Biol Chem 1987;262:549-57.
31. Masson P, Nachon F, Bartels CF, Froment MT, Ribes F, Matthews
C, Lockridge O. High activity of human butyrylcholinesterase at
low pH in the presence of excess butyrylthiocholine. Eur J Biochem
2003;270:315-24.
32. Nicolet Y, Lockridge O, Masson P, Fontecilla-Camps JC, Nachon
F. Crystal structure of human butyrylcholinesterase and of its com-
plexes with substrate and products. J Biol Chem 2003;278:41141-7.
33. Fujii T, Mori Y, Tominaga T, Hayasaka I, Kawashima K.
Maintenance of constant blood acetylcholine content before and
after feeding in young chimpanzees. Neurosci Lett 1997;227:21-4.
34. Fukuto TR. Mechanism of action of organophosphorus and car-
bamate insecticides. Environ Health Perspect 1990;87:245-54.
35. Vignaud A, Fougerousse F, Mousel E, Guerchet N, Hourde C,
Bacou F, Butler-Browne GS, Chatonnet A, Ferry A. Genetic
inactivation of acetylcholinesterase causes functional and struc-
tural impairment of mouse soleus muscles. Cell Tissue Res
2008;333:289-96.
36. Hartmann J, Kiewert C, Duysen EG, Lockridge O, Greig NH,
Klein J. Excessive hippocampal acetylcholine levels in acetylcho-
linesterase-deficient mice are moderated by butyrylcholinesterase
activity. J Neurochem 2007;100:1421-9.
37. 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;253:872-9.
38. Ollis DL, Cheah E, Cygler M, Dijkstra B, Frolow F, Franken
SM, Harel M, Remington SJ, Silman I, Schrag J, Sussman JL,
Verschueren KHG, Goldman A. The α/β hydrolase fold. Protein
Eng 1992;5:197-211.
39. Cygler M, Schrag JD, Sussman JL, Harel M, Silman I, Gentry
MK, Doctor BP. Relationship between sequence conservation and
three-dimensional structure in a large family of esterases, lipases,
and related proteins. Protein Sci 1993;2:366-82.
40. Brenner S. The molecular evolution of genes and proteins: a tale
of two serines. Nature 1988;334:528-30.
41. Silman I, Sussman JL. Acetylcholinesterase: how is structure re-
lated to function? Chem Biol Interact 2008;175:3-10.
42. Kovarik Z, Radic Z, Berman HA, Simeon-Rudolf V, Reiner E,
Taylor P. Acetylcholinesterase active centre and gorge conforma-
tions analysed by combinatorial mutations and enantiomeric phos-
phonates. Biochem J 2003;373:33-40.
43. Bartolucci C, Haller LA, Jardis U, Fels G, Lamba D. Probing
Torpedo californica acetylcholinesterase catalytic gorge with
two novel bis-functional galanthamine derivatives. J Med Chem
2010;53:745-51.
44. Kreienkamp HJ, Weise C, Raba R., Aaviksaar A, Hucho F. Anionic
subsites of the catalytic center of acetylcholinesterase from Torpedo
and from cobra venom. Proc Natl Acad Sci USA 1991;88:6117-21.
45. Ripoll DR, Faerman CH, Axelsen PH, Silman I, Sussman JL.
An electrostatic mechanism for substrate guidance down the
aromatic gorge of acetylcholinesterase. Proc Natl Acad Sci USA
1993;90:5128-32.
46. Koellner G, Steiner T, Millard CB, Silman I, Sussman JL. A neu-
tral molecule in a cation-binding site: specific binding of a PEG-
SH to acetylcholinesterase from Torpedo californica. J Mol Biol
2002;320:721-5.
47. Changeux JP. Responses of acetylcholinesterase from Torpedo mar-
marata to salts and curarizing drugs. Mol Pharmacol 1996;2:369-
92.
48. Johnson G, Moore SW. The peripheral anionic site of acetylcho-
linesterase: structure, functions and potential role in rational drug
design. Curr Pharm Des 2006;12:217-25.
49. Haviv H, Wong DM, Silman I, Sussman JL. Bivalent ligands de-
rived from Huperzine A as acetylcholinesterase inhibitors. Curr
Top Med Chem 2007;7:375-87.
50. Eichler J, Anselment A, Sussman JL, Massoulie J, Silman I.
Differential effects of „peripheral“site ligands on Torpedo and
chicken acetylcholinesterase. Mol Pharmacol 1994;45:335-40.
51. Inestrosa NC, Dinamarca MC, Alvarez A. Amyloid-cholinesterase
interactions. Implications for Alzheimer’s disease. FEBS J
2008;275:625-32.
52. Chatonnet A, Lockridge O. Comparison of butyrylcholinesterase
and acetylcholinesterase. Biochem J 1989;260:625-34.
53. Andres C, elMourabit M, Stutz C, Mark J, Waksman A. Are solu-
ble and membrane-bound rat brain acetlcholinesterase different?
Neurochem Res 1990;15:1065-72.
54. Fernandez HL, Moreno RD, Inestrosa NC. Tetrameric (G4) acetyl-
cholinesterase: structure, localization, and physiological regulation.
J Neurochem 1996;66:1335-46.
55. Inestrosa NC, Roberts WL, Marshall TL, Rosenberry TL.
Acetylcholinesterase from bovine caudate nucleus is attached to
membranes by a novel subunit distinct from those of acetylcholin-
esterases in other tissues. J Biol Chem 1987;262:4441-4.
56. Mehlert A, Varon L, Silman I, Homans SW, Ferguson MAJ.
Structure of the glycosyl-phosphatidylinositol membrane anchor
of acetylcholinesterase from the electric organ of the electric-fish,
Torpedo californica. Biochem J 1993;296:473-9.
57. Rosenberry TL, Roberts WL, Haas R. Glycolipid membrane-bind-
ing domain of human erythrocyte acetylcholinesterase. Fed Proc
1986; 45:2970-5.
58. Liao J, Norgaard-Pedersen B, Brodbeck U. Subunit association
and glycosylation of acetylcholinesterase from monkey brain. J
Neurochem 1993;61:1127-34.
59. Massoulie J, Bon S. The molecular forms of cholinesterse and ace-
tylcholinesterase in verterbrates. Ann Rev Neurosci 1982;5:57-106.
60. Feng G, Krejci E, Molgo J, Cunningham JM, Massoulie J, Sanes
JR. Genetic analysis of collagen Q: roles in acetylcholinesterse
and butyrylchoilnesterase assembly and in synaptic structure and
function. J Cell Biol 1999;144:1349-60.
61. Harel M, Sussman JL, Krejci E, Bon S, Chanal P, Massoulie J,
Silman I. Conversion of acetylcholinesterase to butyrylcholin-
esterase: modeling and mutagenesis. Proc Natl Acad Sci USA
1992;89:10827-31.
62. Haigh JR, Johnston SR, Peppernay A, Mattern PJ, Garcia GE,
Doctor BP, Gordon RK, Aisen PS. Protection of red blood cell
acetylcholinesterase by oral huperzine A against ex vivo soman
exposure: next generation prophylaxis and sequestering of ace-
tylcholinesterase over butyrylcholinesterase. Chem Biol Interact
2008;175:380-6.
228 M. Pohanka
63. Ahmed M, Rocha JB, Correa M, Mazzanti CM, Zanin RF, Morschi
AL, Morsch VM, Schetinger MR. Inhibition of two different cho-
linesterases by tacrine. Chem Biol Interact 2006;162:165-71.
64. Pohanka M, Jun D, Kuca K. Amperometric biosensor for evalu-
ation of competitive cholinesterase inhibition by the reactivator
HI-6. Anal Lett 2007;40:2351-9.
65. Pohanka M, Musilek K, Kuca K. Progress of biosensors based on
cholinesterase inhibition. Curr Med Chem 2009;16:1790-8.
66. Bartling A, Worek F, Szinicz L, Thiermann H. Enzyme-kinetic
investigation of different sarin analogues reacting with hu-
man acetylcholinesterase and butyrylcholinesterase. Toxicology
2007;233:166-72.
67. Tecles F, Ceron JJ. Determination of whole blood cholinesterase
in different animal species using specific substrates. Res Vet Sci
2001;70:233-8.
68. Rosenberry TL. Catalysis by acetylcholinesterase: evidence that
the rate-limitng step for acylation with certain substrates general
acid-base catalysis. Proc Natl Acad Sci USA 1975;72:3834-8.
69. Krupka RM. The mechanism of action of acetylcholinesterase:
substrate inhibition and the binding of inhibitors. Biochemistry
1963;2:76-82.
70. Reed MC, Lieb A, Nijhout HF. The biological significance of sub-
strate inhibition: a mechanism with diverse functions. Bioassays
2010;32:422-9.
71. Guedes RN, Zhu KY, Kambhampati S, Dover BA. Characterization
of acetylcholinesterase purified from the lesser grain borer,
Rhyzopertha dominica (Coleoptera: Bostrichidae). Comp Biochem
Physiol C Pharmacol Toxicol Endocrinol 1998;119:205-10.
72. Gao JR, Zhu KY. An acetylcholinesterase purified from the
greenbug (Schizaphis graminum) with some unique enzymologi-
cal and pharmacological characteristics. Insect Biochem Mol Biol
2001;31:1095-104.
73. Paraoanu LE, Layer PG. Acetylcholinesterase in cell adhesion,
neurite growth and network formation. FEBS J 2008;275:618-24.
74. Johnson G, Swart C, Moore SW. Non-enzymatic developmental
functions of acetylcholiensterase – the question of redundancy.
FEBS J 2008;275:519-38.
75. Adler M, Filbert MG. Role of butyrylcholinesterase in canine tra-
cheal smooth muscle function. FEBS Lett 1990;267:107-10.
76. Villarroya M, Garcia AG, Marco JL. New classes of AChE inhibi-
tors with additional pharmacological effects of interests for the
treatment of Alzheimer’s disease. Curr Pharm Des 2004;10:3177-84.
77. Giacobini E. Selective inhibitors of butyrylcholinesterase: a val-
id alternative for therapy of Alzheimer’s disease? Drugs Aging
2001;18:891-8.
78. Giacobini E. Cholinesterase inhibitors: new roles and therapeutic
alternatives. Pharmacol Res 2004;50:433-40.
79. Weiner L, Shnyrov VL, Konstantinovskji L, Roth E, Ashani Y,
Silman I. Stabilization of Torpedo californica acetylcholinesterase
by reversible inhibitors. Biochemisty 2009;48:563-74.
80. Millard CB, Kryger G, Ordentlich A, Greenblatt HM, Harel M,
Raves ML, Segall Y, Barak D, Shafferman A, Silman I, Sussman
JL. Crystal structures of aged phosphonylated acetylcholinesterase:
nerve agent reaction products at the atomic level. Biochemistry
1999;38:7032-9.
81. Barak D, Ordentlich A, Stein D, Yu QS, Greig NH, Shafferman A.
Accomodation of physostigmine and its analogues by acetylchoil-
nesterase is dominated by hydrophobic interactions. Biochem J
2009;417:213-22.
82. Schaltz F. Neurosciences and research on chemical weapons of
mass destruction in Nazi Germany. J Hist Neurosci 2006;15:186-
209.
83. Kuca K, Pohanka M. Chemcial warfare agents. EXS 2010; 100:543-58.
84. Jovanovic D. The effect of bispyridinium oximes on neuromuscular
blockade induced by highly toxic organohposhates in rat. Archives
Internationales de Pharmacodynamie et de Therapie 1983;262:231-41.
85. Brimblecomb RW, Green DM, Stratton JA, Thompson PB. The
protective actions of some anticholinergic drugs in sarin poisoning.
Brit J Pharmacol 1970;39:822-30.
86. Boskovic B, Kovacevic V, Jovanovic D. 2-PAM chloride HI6
and HGG12 in soman and tabun poisoning. Fund Appl Toxicol
1984;4:106-15.
87. Rengstorff RH. Accidental exposure to sarin: vision effects. Arch
Toxicol 1985;56:201-3.
88. Chilcott RP, Dalton CH, Hill I, Davison CM, Blohm KL, Clarkson
ED, Hamilton MG. In vivo skin absorption and distribution of
the nerve agent VX (O-ethyl-S-[2(diisopropylamino)ethyl] methyl-
phosphonothioate) in the domestic white pig. Hum Exp Toxicol
2005;24:347-52.
89. Hemmer AC, Otto TC, Wierdl M, Edwards CC, Fleming CD,
MacDonald M, Cashman JR, Potter PM, Cerasoli DM, Redinbo
MR.. Human carboxylesterase 1 stereoselectively binds the nerve
agents cyclosarin and spontaneously hydrolyzes the nerve agent
sarin. Mol Pharmacol 2010;77:508-16.
90. Kanamori-Kataoka M, Seto Y. Paraoxonase activity against nerve
gases measured by capillary electrophoresis and characterization
of human serum paraoxonase (PON1) polymorphism in the cod-
ing region (Q192R). Anal Biochem 2009;385:94-100.
91. Kasagami T, Miyamoto T, Yamamoto I. Activated transformations
of organophoshorus insecticides in the case of non-AChE inhibi-
tory oxons. Pest Manag Sci 2002;58:1107-17.
92. Haigh JR, Adler M, Apland JP, Desphpande SS, Barham CB,
Desmond P, Koplovitz I, Lenz DE, Gordon RK. Protection by
pyridostigmine bromide of marmoset hemi-diaphragm acetylcho-
linesterase activity after soman exposure. Chem Biol Interact, In
press.
93. Andersen JB, Engeland A, Owe JF, Gilhus NE. Myasthenia gra-
vis requiring pyridostigmine treatment in a national population
cohort. Eur J Neurol, In press.
94. Darvesh S, Darvesh KV, McDonald RS, Mataija D, Walsh R,
Mothana S, Lockridge O, Martin E. Carbamates with differential
mechanism of inhibition toward acetylchoilnesterase and butyr-
ylcholinesterase. J Med Chem 2008;51:4200-12.
95. Friedman A, Kaufer D, Shemer J, Hendler I, Soreq H, Tur-Kaspa I.
Pyridostigmine brain penetration under stress enhances neuronal
excitability and induces early immediate transcriptional response.
Nat Med 1996;2:1382-5.
96. Chitnis S, Rao J. Rivastigmine in Parkinson’s disease dementia.
Expert Opin Drug Metab Toxicol 2009;5:941-55.
97. Cummings JL, Nadel A, Masterman D, Cyrus PA. Efficacy of met-
rifonate in imporving the psychiatric and behavioral disturbances
of patients with Alzheimer’s disease. J Geriatr Psychiatry Neurol
2001;14:101-8.
98. Alfirevic A, Mills T, Carr D, Barratt BJ, Jawaid A, Sherwood
J, Smith JC, Tugwood J, Hartkoorn R, Owen A, Park KB,
Pirmohamed M. Tacrine-induced liver damage: an analysis of 19
candidate genes. Pharmacogenet Genomics 2007;17:1091-100.
99. Tumiatti V, Minarini A, Bolognesi ML, Milelli A, Rosini M,
Melchiorre C. Tacrine derivatives and Alzheimer’s disease. Curr
Med Chem 2010;17:1825-38.
100. Bajgar J, Bisso GM, Michalek H. Differential inhibition of rat
brain acetylcholinesterase molecular forms by 7-methoxytacrine
in vitro. Toxicol Lett 1995;80:109-14.
101. Pohanka M, Kuca K, Kassa J. New performance of biosensor
technology for Alzheimer’s disease drugs: in vitro comparison of
tacrine and 7-methoxytacrine. Neuroendocrinol Lett 2008;29:755-
8.
102. Jung HA, Min BS, Yokozawa T, Lee JH, Kim YS, Choi JS. Anti-
Alzheimer and antioxidant activities of Coptidis Rhizoma alka-
loids. Biol Pharm Bull 2009;32:1433-8.
103. Mashkovsky MD, Kruglikova-Lvova RP. On the pharmacol-
ogy of the new alkaloid galantamine. Farmakologia Toxicologia
(Moscow) 1951;14:27-30.
104. Bartolucci C, Perola E, Pilger C, Fels G, Lamba D. Three-
dimensional structure of a complex of galanthamine (Nivalin)
with acetylcholinesterase from Torpedo californica: implications
for the design of new anti-Alzheimer drugs. Proteins 2001;42:182-
91.
105. Pilger C, Bartolucci C, Lamba D, Tropsha A, Fels G. Accurate pre-
diction fo the bound conformation of galanthamine in the active
site of Torpedo californica acetylcholinesterase using molecular
docking. J Mol Graph Model 2001;19:288-96.
106. Axelsen PH, Harel M, Silman I, Sussman JL. Structure and dy-
namics of the active site gorge of acetylcholinesterase: synergistic
229
Cholinesterases, a target of pharmacology and toxicology
use of molecular dynamics simulation and X-ray crystallography.
Protein Sci 1994;3:188-97.
107. Riker WF, Okamoto M, Artusio JF. In vivo reversal of depolar-
izing neuromuscular blockade. Arch Int Pharmacodyn Ther
1995;330:90-101.
108. Castro A, Martinez A. Targeting beta-amyloid pathogen-
esis through acetylchoinesterase inhibitors. Curr Pharm Des
2006;12:4377-87.
109. Hansmann T, Sanson B, Stojan J, Weik M, Marty JL, Fournier
D. Kinetic insight into the mechanism of cholinesterase inhibi-
tion by aflatoxin B1 to develop biosensors. Biosens Bioelectron
2009;24:2119-24.
110. Pohanka M, Musilek K, Kuca K. Evaluation of aflatoxin B1 –
acetylcholinesterase dissociation kinetic using the amperometric
biosensor technology: prospect for toxicity mechanism. Protein
Pept Lett 2010;17:340-2.
111. Pohanka, M, Kuca K, Jun D. Aflatoxin assay using an ameromet-
ric sensor strip and acetylcholinesterase as recognition element.
Sens Lett 2008;6:450-3.
112. Egbunike GN, Ikegwuonu FI. Effect of aflatoxicosis on acetylcho-
linesterse activity in the brain and adenohypophysis of the male
rat. Neurosci Lett 1984;52:171-4.
113. Cavalli A, Bottegoni G, Raco C, De Vivo M, Recanatini M.
A computational study of the binding of propidium to the pe-
ripheral anionic site of human acetylcholinesterase. J Med Chem
2004;47:3991-9.
114. Mazzanti CM, Spanevello RM, Obregon A, Pereira LB, Streher
CA, Ahmed M, Mazzanti A, Graca DL, Morsch VM, Schetinger
MR. Ethidium bromide inhibits rat brain acetylcholinesterase ac-
tivity in vitro. Chem Biol Interact 2006;162:121-7.
115. Drago D, Bolognin S, Zatta P. Role of metal ions in the abeta
oligomerization in Alzheimer’s disease and in other neurological
disorders. Curr Alzheimer Res 2008;5:500-7.
116. Jansson ET. Aluminum exposure and Alzheimer’s disease. J
Alzheimers Dis 2001;3:541-9.
117. Kaizer RR, Correa MC, Spanevello RM, Morsch VM, Mazzanti
CM, Goncalves JF, Schetinger MR. Acetylcholinesterase activa-
tion and enhanced lipid peroxidation after long-term exposure to
low levels of aluminum on different mouse brain regions. J Inorg
Biochem 2005;99:1865-70.
118. Kaizer RR, Correa MC, Gris LR, da Rosa CS, Bohrer D, Morsch
VM, Schetinger MR. Effect of long-term exposure to aluminum
on the acetylcholinesterase activity in the central nervous system
and erythrocytes. Neurochem Res 2008;33:2294-2301.
119. Inestrosa NC, Perez CA, Simpfendorfer RW. Sensitivity of ace-
tylcholinesterase molecular forms to inhibition by high MgCl2
concentration. Biochim Biophys Acta 1994;1208:286-93.
120. Arce MP, Rodriguez-Franco MI, Gonzalez-Munoz GC, Perez C,
Lopez B, Villaroya M, Lopez MG, Garcia AG, Conde S.
Neuroprotective and cholinergic properties of multifunctional
glutamic acid derivatives for the treatment of Alzeheimer’s dis-
ease. J Med Chem 2009;52:7249-57.
121. Berson A, Knobloch M, Hanan M, Diamant S, Sharoni M,
Schuppli D, Geyer BC, Ravid R, Mor TS, Nitsch RM, Soreg H.
Changes in readthrough acetylcholinesterase expression modulate
amyloid-beta pathology. Brain 2008;131:109-19.
122. Darreh-Shori T, Soininen H. Effects of cholinesterse inhibitors on
the activities and protein levels of cholinesterases in the cerebros-
pinal fluid of patients with Alzheimer’s disease: a review of recent
clinical studies. Curr Alzheimer Res 2010;7:67-73.
123. Gao X, Zheng CY, Yang L, Tang XC, Zhang HY. Huperzine A pro-
tects isolated rat brain mitochondria against beta-amyloid peptide.
Free Radic Biol Med 2009; 46:1454-62.
124. Zhang HY, Tang XC. Huperzine B, a novel acetylcholinesterase
inhibitor, attenuates hydrogen peroxide induced injury in PC12
cells. Neurosci Lett 2000;292:41-44.
125. Bai D. Development of huperzine A and B for treatment of
Alzheimer’s disease. Pure Appl Chem 2007;79:469-79.
126. Kr yger G, Silman I, Sussman JL. Three-dimensional structure
of a complex of E2020 with acetylcholinesterase from Torpedo
californica. J Physiol Paris 1998;92:191-4.
... Some compounds may inhibit AcChE and BuChE at various level or even react only with one of them [13]. Numerous cholinesterase biosensors were proposed for the detection of pesticides [14][15][16][17], heavy metal ions [18,19], glycoalkaloids [20,21], fluorides [22], drugs [23,24], chemical warfare agents [25], dyes [26], etc. ...
Article
Fast and simple detection of toxic compounds in aqueous solutions is important for agriculture and pollution monitoring. Enzyme‐based electrochemical biosensors are a promising platform for this task, but they usually lack the ability to distinguish between toxicants if more than one toxicant is present in the sample. Herein, we propose a new method of detection of various toxic compounds in complex multi‐component water samples using an electrochemical biosensor and additional stages of enzyme reactivation. The biosensor is based on butyrylcholinesterase immobilized on the surface of conductometric transducers using glutaraldehyde cross‐linking. It was shown that the biosensor is sensitive to organophosphorus pesticides, heavy metals ions, glycoalkaloids, but has a limited sensitivity to mycotoxins and surfactants. We propose a procedure for the analysis of complex samples with several reactivation stages to be able to determine which category of toxicants is present. Glycoalkaloids are reversible inhibitors and biosensor's activity is restored by washing in working buffer; heavy metal ions and pesticides are irreversible inhibitors and biosensor's activity is restored by incubation in EDTA and PAM‐2 solutions, correspondingly. This biosensor can be used for the detection of separate toxicants or for analysis of their mixtures in aqueous samples. It can be also used for the evaluation of total toxicity of the samples and applied for water quality monitoring.
... AChE, which belongs to the carboxylesterase family of enzymes, is found mainly in neuromuscular and cholinergic brain synapses, where it terminates synaptic transmissions. It is the primary target that inhibits this enzyme with organophosphate derivative compounds such as pesticides and nerve gas (Pohanka 2011, Zılbeyaz et al., 2018, Cavdar et al. 2019). It has a very high activity at a rate that can break up 25,000 ACh molecules per second (Sussman et al. 1991). ...
Article
Full-text available
This study aimed to determine the relationship between temperature and neurotoxicity of a new generation insecticide, the active ingredient of which is flubendiamide, on non-target aquatic organisms (Gammarus pulex (L., 1758)). Firstly, LC 50 values of flubendiamide on G. pulex were determined at 16, 18 and 20 °C as 256.83 ± 19.59 µgL-1 , 169.38 ± 34.17 µgL-1 and 155.84 ± 17.22 µgL-1 , respectively. We found that the acute toxicity sensitivity of the test organism to the pesticide containing flubendiamide increased with increasing temperature. Secondly, G. pulex was exposed to non-lethal concentrations of flubendiamide pesticide for 96 hours at the above temperatures. Finally, the changes in acetylcholinesterase (AChE) enzyme activity were determined for each temperature using ELISA kits. The exposure time between the 24th and 96th h at the same temperature was found to be statistically insignificant (p˃0.05). It was determined that the inhibition value of AChE enzyme activity on the test organism of the pesticide with active ingredient flubendiamide increased depending on the temperature increase. Considering the changes in AChE enzyme activities in G. pulex at different temperatures against flubendiamide insecticide, it was concluded that it can be used as a useful biomarker.
... In addition, galantamine is a cholinesterase inhibitor that interacts with the anionic subsite, as well as with the aromatic gorge [135,136]. Galantamine is also an allosteric ligand at nicotinic cholinergic receptors inducing their modulation and acts specifically to enhance the activity (sensitize) of nicotinic receptors in the presence of acetylcholine [137,138]. In addition, tacrine is the potent inhibitor of anionic active site, and physostigmine decreases acetylcholine hydrolysis rate, and thereby increases its level in damaged neurosynaptic clefts, improving nerve impulse transmission [139]. ...
Article
Objective: Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common type of dementia. The early diagnosis of AD is an important factor for the control of AD progression. Electroencephalography (EEG) can be used for early diagnosis of AD. Acetylcholinesterase inhibitors (AChEIs) are also used for the amelioration of AD symptoms. In this systematic review, we reviewed the effect of different AChEIs including donepezil, rivastigmine, tacrine, physostigmine, and galantamine on EEG patterns in patients with AD. Methods: PubMed electronic database was searched and 122 articles were found. After removal of unrelated articles, 24 articles were selected for the present study. Results: AChEIs can decrease beta, theta, and delta frequency bands in patients with AD. However, conflicting results were found for alpha band. Some studies have shown increased alpha frequency, while others have shown decreased alpha frequency following treatment with AChEIs. The only difference was the type of drug. Conclusions: We found that studies reporting the decreased alpha frequency used donepezil and galantamine, while studies reporting the increased alpha frequency used rivastigmine and tacrine. It was suggested that future studies should focus on the effect of different AChEIs on EEG bands, especially alpha frequency in patients with AD, to compare their effects and find the reason for their different influence on EEG patterns. Also, differences between the effects of AChEIs on oligodendrocyte differentiation and myelination may be another important factor. This is the first article investigating the effect of different AChEIs on EEG patterns in patients with AD.
Article
Full-text available
Several of 3,4,5,6‐tetrahydropyrimidinium salts with 1‐methyl functionalization are produced. By using techniques for ¹H‐NMR, ¹³C‐NMR, and IR spectroscopy, all compounds were investigated. Additionally, these compounds’ abilities to block enzymes were looked into. They had a highly effective inhibitory effect on the isoenzymes of carbonic anhydrases I and II, butyrylcholinesterase (BChE), and acetylcholinesterase (AChE). Ki values were found in the range of 57.43±7.09–170.09±50.91 nM for AChE, 7.19±0.42–69.08±2.44 nM for BChE, and 46.48±5.74–203.38±46.15 nM for hCA I, and 30.19±4.03–171.96±30.27 nM for hCA II. As a result, 1,3‐disubtitituted tetrahydroprimidinium salts exhibited potent inhibition profiles toward indicated metabolic enzymes. One of the most important methods for designing and creating novel, potent medications to treat Alzheimer's disease (AD) worldwide is the synthesis and discovery of new AChE and BChE inhibitors. The activities of synthesized 3,4,5,6‐tetrahydropyrimidinium salts were compared against various proteins that are crystal structure of AChE (PDB ID: 4 M0E), crystal structure of BChE (PDB ID: 5NN0), crystal structure of hCA I (PDB ID: 2CAB), and crystal structure of hCA II (PDB ID: 3DC3), and then the drug properties of these molecules were examined.
Article
Full-text available
Pyrazolines are preferred to be stable heterocycles in medication development. It has been established from previous research that the 2‐pyrazolone scaffold is a common pattern that can be found in many pharmacologically significant therapeutic compounds. They are said to have a wide range of biological actions and have been thoroughly investigated by the scientific community. 2‐pyrazoline has been used as an intriguing pharmacophore to create novel compounds with enhanced potency, less toxicity, and the necessary pharmacokinetic profile to treat unprecedented diseases and global drug resistance. Experiments done by docking confirmed the hypothesized neuropharmacological mode of action and demonstrated a strong affinity towards the monoamine oxidase, β‐amyloid plaques, and AChE. Furthermore, additional computational pharmacokinetic assessments signify that it may not have any significant risk of carcinogenic effects, neurotoxic effects, reproductive intoxication, acute toxicity, mutagenicity, or irritations suggesting their possible utility in depressive, convulsions, and anxiety disorders. This discussion outlines a reliable approach for medicinal chemists to develop pyrazolines for the establishment of neurodegenerative research as well as future research programs on neurodegenerative disorders.
Article
Full-text available
The investigation into human butyrylcholinesterase (hBChE) inhibitors as therapeutic agents for Alzheimer's disease (AD) holds significant promise, addressing both symptomatic relief and disease progression. In the pursuit of novel drug candidates with a selective BChE inhibition pattern, we focused on naturally occurring template structures, specifically Amaryllidaceae alkaloids of the carltonine-type. Herein, we explored a series of compounds implementing an innovative chemical scaffold built on the 3- and 4-benzyloxy-benzylamino chemotype. Notably, compounds 28 (hBChE IC50 = 0.171 ± 0.063 μM) and 33 (hBChE IC50 = 0.167 ± 0.018 μM) emerged as top-ranked hBChE inhibitors. In silico simulations elucidated the binding modes of these compounds within hBChE. CNS availability was predicted using the BBB score algorithm, corroborated by in vitro permeability assessments with the most potent derivatives. Compound 33 was also inspected for aqueous solubility, microsomal and plasma stability. Chemoinformatics analysis validated these hBChE inhibitors for oral administration, indicating favorable gastrointestinal absorption in compliance with Lipinski's and Veber's rules. Safety assessments, crucial for the chronic administration typical in AD treatment, were conducted through cytotoxicity testing on human neuroblastoma (SH-SY5Y) and hepatocellular carcinoma (HepG2) cell lines.
Article
Background Cholinesterase is a biomarker for poisonings by anticholinesterase agents, but its reference values are scarce, and possible interaction with collars containing parasiticides has not been studied. Objectives We aimed to evaluate the serum cholinesterase activity of healthy dogs without a history of contact with anticholinesterase agents and healthy animals exposed to commercial collars containing organophosphate. Methods Ninety‐nine dogs were used and included healthy animals without recent exposure to anticholinesterase agents and healthy animals previously exposed to diazinon collars. Serum quantification of the enzyme butyrylcholinesterase (BuchE) through spectrophotometry was conducted on all samples. In experiment 1, BuchE activity was quantified at time 0 and 7 days after, a time when the samples were kept at −18°C. In experiment 2, sampling times were 0, 7, 14, 21, 28, and 56 days. Results Time 0 values were 4622.38 ± 1311.53 U/L. After 7 days, a significant decay was observed, with a mean of 3934.45 ± 1430.45 U/L. Spearman's test was performed, finding a weak correlation between ALT, creatinine, total plasma proteins, age, weight, red blood cells, platelets, leukocytes, and BuchE activities. In experiment 2, the mean at time 0 was 4753 ± 454.8 U/L. With exposure to the collar, there was a decay of up to 93% after 14 days. Conclusions Normality values of serum BuchE in healthy dogs without a history of exposure to anticholinesterase agents were 4360.8–4883.96 U/L. Freezing serum caused a decrease in BuchE activity. Exposure to commercial collars containing diazinon also reduced BuchE activity without clinical signs, indicating that previously exposed animals should be evaluated carefully.
Article
Full-text available
Phytochemicals are plant secondary metabolites that show health benefits for humans due to their bioactivity. There is a huge variety of phytochemicals that have already been identified, and these compounds can act as antimicrobial and neuroprotection agents. Due to their anti-microbial activity and neuroprotection, several phytochemicals might have the potency to be used as natural therapeutic agents, especially for Helicobacter pylori infection and neurodegenerative disease, which have become a global health concern nowadays. According to previous research, there are some connections between H. pylori infection and neurodegenerative diseases, especially Alzheimer’s disease. Hence, this comprehensive review examines different kinds of phytochemicals from natural sources as potential therapeutic agents to reduce H. pylori infection and improve neurodegenerative disease. An additional large-scale study is needed to establish the connection between H. pylori infection and neurodegenerative disease and how phytochemicals could improve this condition.
Article
Full-text available
Our goal was to design, synthesize, and evaluate new cholinesterase inhibitors. Fourteen dehydroamino acids esterified to choline and to its ternary analog were synthesized by a new method that gave a yield of 84-93%. The potency of the amino acid ester derivatives was tested by measuring K(i) values for inhibition of human red cell acetylcholinesterase and human plasma butyrylcholinesterase. The most potent compound was a choline ester of dehydrophenylalanine where the amine group of the amino acid was derivatized with a benzoyl group containing a methoxy in the 2-position, CH(3)O(C(6)H(4))CONHC(CHC(6)H(5))COOCH(2)CH(2)N(+)(CH(3))(3). This compound was a strong inhibitor of both human acetylcholinesterase and human butyrylcholinesterase, with K(i) values of 10 microM and 0.08 microM, respectively. These K(i) values are comparable to that of Rivastigmine. Docking of the most potent compound into the active site of human butyrylcholinesterase showed that the lowest energy model had two benzene rings oriented towards Trp 82 and Tyr 332 whereas the positively charged nitrogen group was stabilized by Trp 231. This orientation placed the ester group 3.89 A from the active site Ser 198, a distance too far for covalent bonding, explaining why the esters are inhibitors rather than substrates. This class of anticholinesterase agents has the potential for therapeutic utility in the treatment of disorders of the cholinergic system.
Article
To date, the pharmacotherapy of Alzheimer's disease (AD) has relied on acetylcholinesterase (AChE) inhibitors (AChEIs) and, more recently, an N-methyl-D-aspartate receptor (NMDAR) antagonist. AD is a multifactorial syndrome with several target proteins contributing to its etiology. "Multi-target-directed ligands" (MTDLs) have great potential for treating complex diseases such as AD because they can interact with multiple targets. The design of compounds that can hit more than one specific AD target thus represents an innovative strategy for AD treatment. Tacrine was the first AChEI introduced in therapy. Recent studies have demonstrated its ability to interact with different AD targets. Furthermore, numerous tacrine homo- and heterodimers have been developed with the aim of improving and enlarging its biological profile beyond its ability to act as an AChEI. Several tacrine hybrid derivatives have been designed and synthesized with the same goal. This review will focus on and summarize the last two years of research into the development of tacrine derivatives able to hit AD targets beyond simple AChE inhibition.
Article
Background: Measurement of plasma butyrylcholinesterase (BChE) activity and inhibitor-based phenotyping are standard methods for identifying patients who experience post-succinylcholine (SC) apnea attributable to inherited variants of the BChE enzyme. Our aim was to develop PCR-based assays for BCHE mutation detection and implement them for routine diagnostic use at a university teaching hospital. Methods: Between 1999 and 2002, we genotyped 65 patients referred after prolonged post-SC apnea. Five BCHE gene mutations were analyzed. Competitive oligo-priming (COP)-PCR was used for flu-1, flu-2, and K-variant and direct DNA sequencing analysis for dibucaine and sil-1 mutations. Additional DNA sequencing of BCHE coding regions was provided when the five-mutation screen was negative or mutation findings were inconsistent with enzyme activity. Results: Genotyping identified 52 patients with primary hypocholinesterasemia attributable to BCHE mutations, and in 44 individuals the abnormalities were detected by the five-mutation screen (detection rate, 85%). Additional sequencing studies revealed mutations in eight other patients, including five with novel mutations. The most common genotype abnormality was compound homozygous dibucaine and homozygous K-variant mutations. No simple homozygotes were found. Of the remaining 13 patients, 3 had normal BChE activity and gene, and 10 were diagnosed with hypocholinesterasemia unrelated to BCHE gene abnormalities. Conclusion: A five-mutation screen for investigation of post-SC apnea identified BCHE gene abnormalities for 80% of a referral population. Six new BCHE mutations were identified by sequencing studies of 16 additional patients.
Article
The regulatory agencies of the United States and Canada have placed aluminum on priority lists for research designed to fill data gaps relating to neurotoxicity. This is to create a factual basis for the establishment of health standards for drinking water. In this review, we consider evidence for a significant role for aluminum in AD. Aluminum has been implicated as a potential risk factor in Alzheimer's Disease (AD) and for elderly cognitive impairment by epidemiology studies of drinking water and a food study. Most people experience aluminum brain overload in the aging process. Aluminum levels over 20 times higher than those of a middle-aged group were found in a brain autopsy study of elderly persons, roughly correlating over the age period with densities of senile plaques and neurofibrillary tangles. Persons with AD have been found to experience increased absorption of aluminum and higher blood levels. More controversially, the majority of brain studies also show elevated aluminum levels, though there is disagreement over location of metal buildup. Clinical intervention to lower brain aluminum by chelation has slowed the progression of AD.
Article
Theoretical analysis is carried out and a reliable kinetic method for establishment of individuality of cholinesterases in studied preparation is proposed. For reliable conclusion about the presence of single cholinesterase or a mixture of cholinesterases in the biosample, it is necessary to determine values of the experimental kinetic constant of irreversible inhibition by several (three or more) inhibitors, using for evaluation of the catalytic activity of the biomaterial not less than three different substrates consecutively, for example, acetyl--methylcholine (substrate selective to typical acetylcholinesterase), butyrylcholine (substrate selective to typical butyrylcholinesterase), and acetylcholine (substrate hydrolyzed easily by cholinesterases of different types).
Article
Acute sc toxicity of soman increased in the order, mice → rats → guinea pigs → dogs, being 12.6 times more toxic to dogs (LD50 = 0.05 μmol/kg) than to mice. It was 2.8 times more toxic than tabun to mice and 35 times more toxic to dogs. HI-6 was the least toxic and had similar toxicity values to the four animal species studied and HGG-12 the most toxic of the three oximes used. HGG-12 has shown the greatest interspecies variation (rats:dogs = 1:19.5). HI-6, HGG-12, and PAM-2 Cl (in conjunction with atropine and diazepam) revealed the best protective effect in soman-poisoned dogs, with the respective protective indices of 9, 6.3, and 3.5, followed by guinea pigs. In tabun poisoning the best, but relatively low, protective effect was found only in guinea pigs. The introduction of diazepam increased the protective effects of atropine-oxime combination in soman and tabun poisoning by 10 to 80%. We suggest that the high toxicity of soman and low toxicity of HI-6 may be anticipated in man. The inefficiency of HI-6, HGG-12, and PAM-2 Cl in tabun poisoning points either to the search of new compounds or to the use of the mixture of the oximes found to be effective against the known chemical warfare nerve agents.
Article
Amperometric biosensors containing enzymes butyrylcholinesterase or acetylcholinesterase were prepared. The biosensors were employed for studying of cholinesterase reactivator: HI‐6. Competitions between HI‐6 and acetylthiocholine as enzyme substrate were used for determination of IC50 value. Biosensors with butyrylcholinesterase from human serum determined IC50 as (1.00±0.02)×10 M; the biosensor with acetylcholinesterase from human erythrocytes performance provided IC50 (3.31±0.13)×10 M, the one with human recombinant acetylcholinesterase (2.00±0.06)×10 M and finally biosensor with acetylcholinesterase from electric eel (6.17±0.17)×10 M when 5 mM acetylthiocholine as substrate was used. We are encouraged to consider presented biosensors as a very useful for evaluation of newly prepared cholinesterase reactivators.
Article
Recent studies have proved that huperzine A (HupA) possesses different pharma- cological actions other than the inhibition of hydrolysis of ACh. These noncholinergic roles, for instance, the antagonist effect on NMDA receptor, the protection of neuronal cells against β-amyloid, free radicals, and hypoxia-ischemia-induced injury, could be important too in Alzheimer's disease (AD) treatment. The therapeutic effects of HupA are probably based on a multitarget mechanism. By targeting dual active sites of AChE, a series of bis- and bifunc- tional HupB compounds with various lengths of tether were designed, synthesized, and tested for the inhibition and selectivity of AChE. The most potent bis-HupB compound exhibited increase by three orders of magnitude in AChE inhibition and two orders of magnitude in se- lectivity for AChE than its parent HupB.