You are on page 1of 173

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/333614149

Identification of novel scaffolds for Monoamine oxidase B inhibitors.

Thesis · May 2014


DOI: 10.13140/RG.2.2.30232.44800

CITATION READS
1 316

1 author:

Hasanain Odhar
Al-Zahrawi University College
23 PUBLICATIONS   93 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Computer aided design and evaluation of novel acetylcholinesterase enzyme inhibitor View project

Association between Helicobacter pylori prevalence and extra-gastric diseases View project

All content following this page was uploaded by Hasanain Odhar on 04 June 2019.

The user has requested enhancement of the downloaded file.


Identification of novel scaffolds for Monoamine
oxidase B inhibitors

A thesis submitted

to Kent State University in partial

Fulfillment of the requirements for the

Degree of Master of Science

By

Hasanain Odhar

May, 2014
Thesis written by

Hasanain Odhar

B.S., University of Baghdad, 2007

M.S., Kent State University, 2014

Approved by

Werner Geldenhuys
__________________________, Chair, Master’s Thesis Committee

Altaf Darvesh
__________________________, Member, Master’s Thesis Committee

Richard Carroll
__________________________, Member, Master’s Thesis Committee

Eric Mintz
__________________________, Director, School of Biomedical Sciences

Janis Crowther
__________________________, Dean, College of Arts and Sciences

ii
Table of Contents

List of figures ..................................................................................................................... vi

List of tables ....................................................................................................................... ix

Acknowledgments............................................................................................................... x

Chapter one: Introduction ............................................................................................... 1

1.1- Parkinson's disease (PD) .......................................................................................... 1

1.2- Monoamine oxidase enzyme (MAO)....................................................................... 3

1.3- Structure of MAO-B ................................................................................................ 4

1.3.1- membrane binding domain ................................................................................ 6

1.3.2- Substrate binding domain .................................................................................. 6

1.3.3- Flavin binding domain ...................................................................................... 7

1.3.4- Structural comparison between human MAO-B and human MAO-A .............. 9

1.4- Reaction mechanism of MAO enzyme .................................................................. 11

1.5- Functional role of aromatic cage............................................................................ 18

1.6- MAO inhibitors with a possible or approved clinical application ......................... 20

1.7- MAO-B inhibitors with a possible disease modifying effect ................................. 22

iii
Chapter two: Materials and Methods ........................................................................... 28

2.1- Prospective candidates ........................................................................................... 28

2.2- Docking studies ...................................................................................................... 36

2.3- Monoamine oxidase inhibition assay ..................................................................... 36

2.4- Bovine serum albumin (BSA) binding assay ......................................................... 40

2.5- Parallel Artificial Membrane Permeability Assay (PAMPA) ................................ 41

2.6- Oxygen Radical Absorbance Capacity (ORAC) assay .......................................... 43

2.7- Data analysis .......................................................................................................... 44

Chapter three: Results .................................................................................................... 45

3.1- Selection of the most efficient candidates ............................................................. 45

3.2- Docking studies results .......................................................................................... 47

3.3- Monoamine oxidase inhibition assay results ......................................................... 71

3.4- Bovine serum albumin (BSA) binding assay results ............................................. 76

3.5- Parallel artificial membrane permeability assay (PAMPA) results ....................... 82

3.6- Oxygen radical absorbance capacity (ORAC) assay results .................................. 84

3.7- Structure activity relationship (SAR) study results................................................ 87

Chapter four: Discussion.............................................................................................. 105

iv
References ...................................................................................................................... 138

v
List of figures

Figure 1.1 - Three dimensional structure of human MAO-B enzyme (monomer A) ......... 5

Figure 1.2 - A schematic representation for catalytic reaction pathway followed by MAO

enzyme. .......................................................................................................... 13

Figure 1.3 - A schematic representation for the proposed polar nucleophilic mechanism

of MAO enzyme catalysis ............................................................................. 16

Figure 1.4 - A schematic representation for the proposed single electron transfer (SET)

mechanism of MAO catalysis ........................................................................ 17

Figure 1.5 - A schematic representation for the effect of dipole moments of Tyr398 and

Tyr435 in MAO-B on the electron lone pair of the substrate amine ............. 20

Figure 2.1 - Conversion of kynuramine into 4-Hydroxyquinoline as catalyzed by both

MAO-A and MAO-B ..................................................................................... 39

Figure 3.1 - A scatter plot of MAO-B inhibition potential (percentage) against antioxidant

potential (percentage) for our chemicals. ...................................................... 46

Figure 3.2 - Docking of compound 5140311 into human MAO-B crystal (2D and 3D

images) ........................................................................................................... 49

Figure 3.3 - Docking of compound 5140321 into human MAO-B crystal (2D and 3D

images) ........................................................................................................... 51

Figure 3.4 - Docking of compound 5143886 into human MAO-B crystal (2D and 3D

images) ........................................................................................................... 53
vi
Figure 3.5 - Docking of compound 7919469 into human MAO-B crystal (2D and 3D

images) ........................................................................................................... 55

Figure 3.6 - Docking of compound 7954103 into human MAO-B crystal (2D and 3D

images) ........................................................................................................... 57

Figure 3.7 - Docking of compound 5476423 into human MAO-B crystal (2D and 3D

images) ........................................................................................................... 59

Figure 3.8 - Docking of compound 7730595 into human MAO-B crystal (2D and 3D

images) ........................................................................................................... 63

Figure 3.9 - Docking of compound 7747412 into human MAO-B crystal (2D and 3D

images) ........................................................................................................... 66

Figure 3.10 - MAO-B inhibition potential as produced by the selected compounds ....... 73

Figure 3.11 - The potency of the inhibitory effect against MAO-A as produced by the

tested compounds........................................................................................... 74

Figure 3.12 - BSA binding potential of the tested chemicals ........................................... 77

Figure 3.13 - Relative BSA binding curves for the selected compounds ......................... 78

Figure 3.14 - Percentage of antioxidant power for the selected chemicals ...................... 85

Figure 3.15 - MAO-B inhibition potential, antioxidant power, chemical structural formula

and MAO-B docking images for compounds 5140311 and 5140321 (SAR

study). ............................................................................................................ 89

Figure 3.16 - MAO-B inhibition potential, antioxidant power and chemical structural

formula for compounds 5143886, 5144779, 5108394 and 5108400 (SAR

vii
study). MAO-B docking images for compounds 5108394 and 5108400 are

also shown ..................................................................................................... 96

Figure 3.17 - MAO-B inhibition potential, antioxidant power and chemical structural

formula for compounds 7935085 and 5476423 (SAR study) ...................... 102

Figure 4.1 - Chemical structural formula, IUPAC name and MAO-B docking images for

our first prospective scaffold (scaffold A) ................................................... 114

Figure 4.2 - Chemical structural formula, IUPAC name and MAO-B docking images for

our second prospective scaffold (scaffold B) .............................................. 117

Figure 4.3 - Chemical structural formula, IUPAC name and MAO-B docking image for

the modified scaffold (scaffold C) ............................................................... 124

Figure 4.4 - Chemical structural formula, IUPAC name and MAO-B docking image for

the modified scaffold (scaffold D) ............................................................... 126

Figure 4.5 - Chemical structural formula, IUPAC name and MAO-B docking images for

the modified scaffolds E and F (L-alanine derivatives)............................... 130

Figure 4.6 - Chemical structural formula, IUPAC name and MAO-B docking image for

our novel design (scaffold G) ...................................................................... 134

viii
List of tables

Table 1.1 - A brief summary about MAO-inhibitors with a possible or approved clinical

application...................................................................................................... 21

Table 2.1 - Prospective candidates' specifications. ........................................................... 29

Table 3.1 - Docking results for BSA crystal. .................................................................... 70

Table 3.2 - Inhibition selectivity toward MAO isozymes................................................. 75

Table 3.3 - EC50 values for (antioxidant capacity – concentration) curve of each tested

compound. ..................................................................................................... 86

Table 4.1 - Classification of PAMPA-BBB results. ....................................................... 111

Table 4.2 - The obedience degree to Lipinski’s rule of five (scaffolds A, B, C and D). 122

Table 4.3 - The obedience degree to Norinder’s rules for CNS permeability. ............... 123

Table 4.4 - The application of Lipinski’s rule of five to scaffolds E, F and G. .............. 136

ix
Acknowledgments

I would like to express my deep gratefulness to my parents, brother and sisters for

their endless support, help and encouragement throughout my life journey.

I would like to thank my advisor (mentor) Dr. Werner Geldenhuys for his

priceless support and substantial guidance throughout research course and thesis writing.

I would also like to express my gratefulness to Dr. Altaf Darvesh and Dr. Richard Carroll

for serving on my thesis committee and for their vital contribution to my project and

thesis.

Finally, I would like to extend my gratitude to Dr. Eric Mintz and Mrs. Judith

Wearden for their precious support.

x
Chapter one: Introduction

1.1- Parkinson's disease (PD)

It is an age related neurodegenerative disease that affects about one million

persons in the United States [1]. The prevalence of Parkinson's disease usually increases

from 1-2% at age 50 to about 5% at age 85 [2, 3]. Pathologically the disease is

characterized by a progressive degeneration of dopaminergic neurons in the substantia

nigra pars compacta, this depletion of dopaminergic neurons will result in four major

motor related hallmarks including tremor, rigidity, bradykinesia and postural imbalance

[4]. These manifestations usually appear when about 60-70% of dopaminergic neurons

are lost [5]. Non-motor symptoms (like depression and dementia) may also appear as the

disease progress over time [6]. Another characteristic pathological feature of Parkinson's

disease is the presence of proteinaceous inclusions called Lewy bodies within the

surviving dopaminergic neurons, these inclusions consist mainly of α-synuclein which is

highly ubiquitinated [7]. Although the exact cause of Parkinson's disease remains unclear,

several factors may play a role in increasing susceptibility to develop the disease. These

factors include: age, environmental factors (ex: Paraquat PQ) [8, 9] and genetic factors

(autosomal dominant and recessive genes like α-synuclein, parkin, DJ-1, LRRK2) [10].

Current therapeutic approach focuses on symptoms attenuation only [11] through

dopamine replacement therapy by using either L-dopa (the immediate

1
2

metabolic precursor of dopamine) or dopamine receptor agonist [12, 13]. Dopamine

replacement therapy does not halt or slow down the pathological degeneration process

and as the disease is advancing, the efficiency of such therapeutic approach is falling

down [14]. In this situation, higher medication dose is required to maintain adequate

therapeutic benefit. Such decision is usually restricted by the fact that the tolerated

levodopa dose is limited by several side effects associated with long term therapy such as

dyskinesia and behavioral disturbances [15, 16]. Drugs that can lower L-dopa dose and/

or delay the need for L-dopa are considered essential for Parkinson's disease therapy

approach [17, 18]. These adjunct drugs involve the following categories: Monoamine

oxidase B (MAO-B) inhibitors, Catechol-O-methyltransferase inhibitors and amantadine

[11]. No drug with neuroprotective activity has been approved by FDA for Parkinson's

disease treatment till this point [19, 20].

Parkinson's disease is a complex neurologic condition with multiple underlying

etiological pathways and treating such disorder with a molecule that target a single

pathway is inefficient, a drug molecule that target different pathways seems to be more

convenient [21-28]. A possible candidate for such multi-targeted drug design is MAO-B

inhibitors, by inhibiting MAO-B enzyme; these compounds can elevate the level of both

endogenous and exogenous dopamine [29, 30]. By blocking the activity of MAO-B

enzyme, these compounds can also lower the production of hydrogen peroxide (a

byproduct of oxidative deamination reaction) and by this way oxidative stress can be

minimized [31]. The rationale for using MAO-B inhibitors as a basic model for the

development of a new antiparkinsonian therapy is encouraged by several facts. The first


3

one is that the activity level of MAO-B enzyme inside the brain increases with age [32-

34] while MAO-A level is not affected by age [87]. Also in patients with Parkinson's

disease, the level of MAO-B is elevated as a result of gliosis. Finally, the activity level of

MAO-B in human basal ganglia is higher than that of MAO-A and since both isozymes

metabolize dopamine with the same level of efficiency, then it is more reasonable to

target MAO-B and not MAO-A [36].

1.2- Monoamine oxidase enzyme (MAO)

MAO is an outer mitochondrial membrane bound flavin containing enzyme with a

molecular weight of about 60 kDa, it plays an important role in the metabolism of

endogenous and exogenous amines [35, 45]. Intraneuronal MAO terminates the action of

endogenous amine neurotransmitters, regulates intraneuronal amine stores and protects

cells from dietary amines (false transmitters) [36].

This enzyme exists in two isoforms (MAO-A) and (MAO-B), these isoforms

shares about 70% sequence identity and represent separate X chromosome related gene

products [37]. These isoforms have different substrate specificities in that MAO-A

catalyzes the oxidation of serotonin (5-HT) while MAO-B catalyzes the oxidation of both

benzylamine and 2-phenylethylamine. Other substrates like dopamine, epinephrine,

norepinephrine, tyramine and tryptamine are oxidized by both isoforms [38]. During

embryonic development, MAO-A is expressed first but the expression level of MAO-B

inside the brain is increased dramatically after birth [32, 39, 40]. MAO enzyme level of

activity is different among various regions of human brain, where the hypothalamus and
4

basal ganglia show the highest activity while cerebellum and neocortex show the lowest

activity [41]. Radio labeled inhibitors were used to identify the distribution of MAO

isozymes throughout the brain, it was found that basal ganglia contains mainly MAO-B

[42].

Immunohistochemical analysis reveals that serotonergic neurons contain

predominantly MAO-B isoform while catecholaminergic neurons contain MAO-A

mainly [43]. These immunohistochemical analysis data would suggest two conclusions:

first, the role of MAO-B in serotonergic neurons is to eliminate any foreign amines and

prevent them from reaching the synaptic cleft while the second conclusion is that MAO-

A in glial cells should be responsible for degradation of serotonin in brain regions rich in

serotonergic neurons. Noradrenergic neurons contain MAO-A and MAO-B, both

isoforms can catalyze the oxidative deamination of norepinephrine efficiently but because

the uptake of norepinephrine into synaptic vesicle is strongly favorable over binding to

MAO therefore the substrate can escape metabolic degradation to some extent [44].

1.3- Structure of MAO-B

MAO-B enzyme consists mainly from protein (520 amino acids) and FAD (redox

cofactor), it crystallizes as a dimer and each monomer involves the following subunits:

membrane binding domain, substrate binding domain and flavin binding domain [46, 47].

Figure 1.1 shows a three dimensional representation for human MAO-B monomer.

MAO-B undergoes post translational modification (acetylation) where the amino terminal

methionine is cleaved and the resultant N-terminal serine is acetylated [48, 49].
5

Figure 1.1- Three dimensional structure of human MAO-B enzyme (monomer A). The

membrane binding domain is in green color (residues 489–500), the substrate binding

domain is in red color (residues 80–210, 286–390 and 454–488) and the flavin binding

domain is in blue color (residues 4–79, 211–285 and 391–453). This figure was generated

by using PyMOL v.1.3 (www.schrodinger.com) [154] and human MAO-B crystal with

the code 2BK3; the sequences of amino acids for the designated domains were obtained

from reference [55].


6

1.3.1- membrane binding domain

C-terminal truncation experiments together with sequence analysis had suggested

that the C-terminal region can function as the membrane binding domain (C-terminal α

helix), this 27 residue transmembrane helix has apolar surface which would encourage its

insertion into mitochondrial membrane [50, 51]. Truncation experiments showed that C-

terminal truncated MAO-B can retain some affinity towards membranes but not as strong

or as specific as intact enzyme and this would suggest the presence of other membrane

binding regions. The electrostatic surface maps show positively charged domains that can

form electrostatic interactions with the anionic membrane surface [54].

1.3.2- Substrate binding domain

In order to access covalent FAD and undergo metabolism, substrates must cross a

specific path with a distance of about 20 Å [52]. The journey starts by negotiation with a

flexible loop (residue 99-112) which function as a gating switch at the surface of outer

mitochondrial membrane, this loop must be moved so that the substrate can reach active

site cavities [53]. The substrate then must pass through two hydrophobic cavities; the first

one is called the entrance cavity with a volume of ~290 ų, the second larger cavity is

called the substrate cavity with a volume of ~400 ų [47, 54]. These two cavities are

separated by an Isoleucine 199 side chain which can function as a gate; depending on the

nature of the substrate, the Isoleucine (Ile) 199 side chain can exhibit either an open

conformation (fuse both cavities) or closed conformation (separate both cavities) [53].
7

Tyr326 residue also plays a role in separating the active site cavities of MAO-B; the side

chain of Tyr326 undergoes conformational changes upon substrate binding [72]

The outer mitochondrial membrane is covered with negative charges and these

charges will attract amine substrates (positively charged) into MAO-B enzyme [52]. FAD

cofactor is located at the end of substrate cavity where it is covalently bound to Cysteine

397 through thioether linkage [46].

1.3.3- Flavin binding domain

Flavin adenine dinucleotide (FAD) is considered as an essential entity for MAO

enzyme catalytic activity (redox cofactor); it is bound covalently to MAO-B apoenzyme

through a thioether linkage which is formed between cysteine 397 residue and the 8α-

methylene of isoalloxazine ring [46, 37]. This binding is located near the C-terminus of

MAO enzyme [55].

FAD coenzyme is incorporated in an extended conformation into a hydrophobic

environment within MAO-B, specific interactions are found between FAD and MAO-B

apoenzyme [55]. These interactions are mainly hydrogen bonds between FAD and either

amino acids side chains or amide linkages in the protein. Mutagenesis experiments had

shown that if Arg42 was replaced by Ala or Lys then this would adversely influence FAD

incorporation into MAO-B, These results showed the requirement of electrostatic

interaction between anionic pyrophosphate group of FAD and the positively charged

guanidino group of Arg42 [56]. The amino acid residues that interact with FAD in MAO-
8

B enzyme are conserved in MAO-A enzyme and this suggests that FAD binding site is

identical in both enzymes [55]. The only difference between these two enzymes is that

Ser59 is replaced with Ala in MAO-A and since the peptide bond (not amino acid side

chain) at this position is involved in hydrogen bonding with the flavin C-4 carbonyl

oxygen, then such amino acid substitution is not thought to cause a significant difference

in FAD binding domain between the these two isozymes. Flavin coenzyme analogs

analysis had shown that both MAO-A and MAO-B have a similar FAD incorporation

mechanism and a similar FAD binding sit [59, 60]. Analogs analysis results also provide

additional support for covalent flavinylation mechanism (quinone methide); it is believed

that both MAO-A and MAO-B follow this mechanism for FAD incorporation [61].

Structural data, obtained through diffraction analysis, had shown that the amide

bond between Cys397 and Tyr398 is in cis rather than the favorable trans configuration

[53]. This energetically unfavorable configuration (cis configuration) of the peptide bond

is necessary to make the phenolic side chain of Tyr398 in a perpendicular orientation to

the flavin ring, this orientation is essential for catalytic activity (aromatic cage) [58]. Two

main steric constraints that affect flavin reactivity had been recognized, first one is the

bending about (N5)-(N10) axis of isoalloxazine ring so that it will deviate 30˚ from

normal planar conformation while the second one is being puckered about the pyrimidine

portion of the ring. These steric deformations will make the reactive sites of flavin ring

(N-5 and C-4) assuming a more sp3 rather than a planar sp2 configuration and this will

enhance MAO enzyme reactivity [53].


9

To determine the effect of covalent binding of FAD coenzyme on the activity and

general stability of MAO enzyme, two mutagenesis experiments were performed. First

experiment had shown that replacement of Cys397 with Ser would result in an inactive

enzyme, it is clear that the hydroxyl group of serine does not have enough nucleophilicity

to form covalent bond with FAD [62]. The second experiment involved the expression of

Cys406Ala mutant MAO-A in riboflavin dependent strain of S. cerevisiae. Mutant MAO-

A was expressed in the absence of riboflavin and it did not show any activity until FAD

was added to the medium. The level of MAO-A activity was dependent on the

concentration of FAD added. This experiment suggested that enzyme activity is not

absolutely dependent on covalent FAD linkage and it also showed the importance of this

covalent linkage in structural stabilization of the enzyme against detergents solubilization

[63].

1.3.4- Structural comparison between human MAO-B and human MAO-A

The most important structural difference between these two isozymes is that

human MAO-B crystallizes as a dimer while human MAO-A crystallizes as a monomer

[64, 47, 58]. It was suggested that in human MAO-A, a Glu-151 is replaced by a Lys

residue. This selective mutation at the dimeric interface may be responsible for

destabilization of the dimeric form in human MAO-A [65]. This shift in the oligomeric

state of human MAO-A mainly affects the temperature stability of the enzyme; it was

found that rat MAO-A (dimer) is more thermally stable than human MAO-A (monomer)

[66]. Dipole moment calculations show a significant change in the direction of


10

monomeric MAO-B dipole moment upon dimerization. In dimeric MAO-B, the

orientation of positively charged end is directed towards the anionic membrane surface

which will provide addition interaction. Such alteration in dipole direction is less

pronounced in MAO-A [54]. New results of distance measurements by pulsed EPR-

DEER (electron paramagnetic resonance-double electron-electron resonance)

experiments showed that human MAO-A is present in dimeric form in the mitochondrial

membrane, it is believed that solubilization of human MAO-A by using detergent will

result in a mixture of both monomeric and dimeric forms of human MAO-A and the

monomeric form will be crystallized more readily [71]

Other significant differences are also present between human MAO-B and human

MAO-A regarding active site shape and volume. The active site in human MAO-B is

bipartite with a total volume of ~700ų (entrance cavity: 290ų and substrate cavity:

~400ų) while in human MAO-A, the active site is monopartite with a volume of ~550ų

[67, 64]. The shape of active site in human MAO-B is longer and narrower than that of

human MAO-A. This difference in active site shape and volume may be in part as a result

of variation in the conformation of cavity shaping loop, this six residue loop is present in

a more compact conformation in human MAO-B as compared with human MAO-A [64,

68]. Active site in both isozymes also show important amino acid replacements for

example human MAO-A contains Phe-208 (Ile-199 in human MAO-B) and Ile-335 (Tyr-

326 in human MAO-B) [64]. A combination of both conformational changes and residue

replacements affect the shape and size of the active site and this in turn will modify the

substrate specificity of the isozyme.


11

Rat MAO-A is about 92% identical with human MAO-A in terms of sequence

identity [64]. Rat MAO-A crystallizes as a dimer with active site cavity volume of about

450ų, its cavity shaping loop conformation is very similar to that of human MAO-B [64,

70]. These structural data about rat and human MAO-A would suggest a possible

relationship between dimer formation and the conformation of cavity shaping loop

through a long range effect [64]. Such long range effect hypothesis had been observed in

a number of other enzymes like dihydrofolate reductase [69]. Kinetic studies showed that

rat MAO-A metabolizes human MAO-B substrates (like benzylamine) more efficiently

than does human MAO-A [66]. Based on the facts mentioned above, the approach of

using rat MAO-A as a laboratory model for the development of human MAO-A specific

inhibitor should be re-evaluated [64].

1.4- Reaction mechanism of MAO enzyme

The enzyme catalyzes the oxidative deamination of primary, secondary and some

tertiary amines, this reaction will result in the formation of hydrogen peroxide, an

aldehyde and either ammonia (if the substrate is primary amine) or substituted amine (if

the substrate is secondary amine) [36, 52]. For MAO-B, the reaction pathway depends on

the type of substrate. In this regard the oxidation of benzylamine follows ternary complex

mechanism where the imine product dissociates from the oxidized enzyme. On the other

hand the binary complex mechanism is usually followed by phenethylamine and in this

case the imine product will dissociate from reduced enzyme before oxidation of the

reduced enzyme-imine complex takes place [73, 74]. Figure 1.2 shows the catalytic
12

reaction pathway for MAO enzyme. The existence of these two pathways (binary and

ternary) depends on the relationship between the rate of imine dissociation from reduced

enzyme and the rate of reduced enzyme-imine complex oxidation [75]. For ternary

complex mechanism (benzylamine), the oxidation rate of reduced enzyme-imine complex

is faster than the rate of imine dissociation from reduced enzyme. In binary complex

mechanism (phenethylamine), the rate of imine dissociation is faster than the rate of

reduced enzyme-imine complex oxidation [73, 75]. Catalysis pathway for MAO-A had

been found to follow ternary complex mechanism only [76].


13

Figure1.2- A schematic representation for catalytic reaction pathway followed by MAO

enzyme. This figure was adapted from reference [52].


14

MAO enzyme catalytic mechanism involves the cleavage of Cα-H bond in amine

substrate with the transfer of two reducing equivalents to the enzyme (flavin ring) and

this will result in formation of imine product and flavin hydroquinone [68]. It is believed

that the amine substrate should be in the deprotonated form so that it can enter the active

site and undergo metabolism, however the exact mechanism of substrate deprotonation at

physiological condition is not known yet [68, 77, 86]. Kinetic isotope studies had

demonstrated that Cα-H bond cleavage is the rate limiting step in oxidative deamination

reaction [77, 78]. Stereochemical analysis showed that only pro-R hydrogen can be

transferred in this catalytic reaction (undergoes abstraction) [73, 79, 80]. The abstraction

of pro-R H can happen in one of three possibilities: as a proton, a hydride ion or a

hydrogen atom. To identify hydrogen form that undergo abstraction, the effect of electron

withdrawing group in para substituted substrate on reaction rate was tested. It was found

that increasing electron withdrawing power will increase reaction rate and this fact

supports the possibility of proton abstraction [77, 78].

Two hypotheses had been suggested for the transfer of reducing equivalents in

MAO catalyzed reaction. The first and more acceptable hypothesis is called polar

nucleophilic mechanism. In this mechanism, substrate amine will attack flavin C4a and

this will form flavin-substrate adduct. Once this adduct is formed, N5 position of flavin

ring becomes a strong base that can abstract proton from Cα [77]. Figure 1.3 shows a

schematic representation for polar nucleophilic mechanism of MAO catalysis. NMR

studies had observed that the pKa value of N5 proton for flavin hydroquinone to be ~24

and this support the idea of N5 sufficient basicity to perform abstraction [81]. As
15

mentioned previously in FAD binding domain structure, the isoalloxazine ring is in a bent

conformation about (N5-N10) axis which deviates about 30˚ from normal planarity [53].

This bending will make N5 atom closer to sp3 than sp2 conformation and this will lead to

a higher electron density at N5 position (better nucleophile) and a lower electron density

at C4a position (better electrophile). This will facilitate the nucleophilic attack by a

suitable substrate at C4a position [68].

The other and less acceptable hypothesis is called single electron transfer (SET)

mechanism; this mechanism involves one electron oxidation of substrate amine nitrogen

leading to the formation of aminium cationic radical and flavin anionic radical. The

production of aminium radical will lower pKa value of Cα-H and this will facilitate

proton abstraction by a base in the active site of MAO enzyme [82-84]. Figure 1.4 shows

a schematic picture for single electron transfer hypothesis. Different structural data

showed that no amino acid within MAO active site can function as an active base or acid

[52]. Experimental monitoring failed to identify intermediate radical and thus it is

difficult to provide any support for SET hypothesis [85]. The formation of aminium

cationic radical is expected to interact with tyrosine residues in aromatic cage (π-cationic

interaction) and this can halt any further reaction [52, 75].
16

Figure 1.3- A schematic representation for the proposed polar nucleophilic mechanism of

MAO enzyme catalysis. This figure was adapted from reference [52].
17

Figure 1.4- A schematic representation for the proposed single electron transfer (SET)

mechanism of MAO catalysis. This schematic figure was adapted from reference [52].
18

1.5- Functional role of aromatic cage

Previous structural analyses had observed that the active site of MAO enzyme

contains two tyrosine residues (Tyr398 and Tyr435 in MAO-B), these residues are

situated in front of the re face of flavin ring and they exhibit an approximately

perpendicular conformation relative to the flavin ring. The phenolic side chains of these

two residues are separated from each other by about 7.8Å [47]. A number of mutagenesis

analyses targeting Tyr435 in MAO-B were performed to identify possible functional

roles of the aromatic cage. Targeting Tyr398 in these analyses was excluded because this

residue is involved in a cis bonding with Cys397 which binds covalently through

thioether linkage with flavin. It is believed that any change in Tyr398 may adversely

affect covalent flavin binding [68, 88].

The aromatic cage does not seem to perform any important structural role within

the active site of MAO enzyme but mutant enzymes show lower stability upon

solubilization by detergents as compared to wild enzyme [88]. Other flavin dependent

amine oxidase enzymes contain aromatic amino acids in a conformation similar to that

observed in the aromatic cage of MAO enzyme and this observation suggests a possible

functional role for aromatic cage in catalytic activity [89, 90]. One of the expected roles

of aromatic cage is to polarize the electron single pair of substrate amine and this will

enhance substrate nucleophilicity. As mentioned above, the distance between phenolic

side chains of aromatic cage was calculated to be 7.8Å which is believed to be optimal

for substrate amine polarization [88]. Aromatic cage may also play a steric role through
19

dipole moment effect of Tyrosine side chain. These dipolar effects can simulate the

presence of electron withdrawing group at para position of substrate leading to a

reduction in the distance between amine nitrogen and flavin C4a, this will facilitate the

transfer of charge from amine to flavin and encourage the formation of flavin C4a adduct

[91]. Figure 1.5 shows us a schematic representation for the role of aromatic cage in

substrate oxidation.

The dipole effect of aromatic cage may also contribute to substrate specificity of

MAO-B, it is suggested that using a substrate having a dipole moment in the same

direction as do the aromatic cage dipoles will result in repulsive interaction and a change

in substrate orientation within active site cavity [88]. All these facts about aromatic cage

structure and function strengthen polar nucleophilic mechanism of MAO-B catalysis

[88].
20

Figure 1.5- A schematic representation for the effect of dipole moments of Tyr398 and

Tyr435 residues in MAO-B on the electron lone pair of the substrate amine before the

nucleophilic attack takes place. This figure was adapted from reference [88].

1.6- MAO inhibitors with a possible or approved clinical application

A summary about these chemicals is shown in the below table (table 1.1) which

was adapted from reference [134].


21

Table 1.1- A brief summary about MAO-inhibitors with a possible or approved clinical

application.

MAO inhibitor Selectivity Binding Applications

Irreversible, Antidepressant and


Phenelzine MAO-A/ MAO-B
covalent. anxiolytic.

Irreversible, Antidepressant and


Tranylcypromine MAO-A/ MAO-B
covalent. anxiolytic.

Selegiline Irreversible,
MAO-B Antiparkinsonian
(L-deprenyl) covalent.

Irreversible,
Rasagiline MAO-B Antiparkinsonian
covalent.
Development as
Reversible, antiparkinsonian was
Lazabemide MAO-B
covalent. discontinued due to
toxicity issues.
Currently undergo phase
Reversible,
Safinamide MAO-B III clinical trials as
non-covalent.
antiparkinsonian.
Antiepileptic and it is
Reversible,
Zonisamide MAO-B approved in Japan as
non-covalent.
antiparkinsonian.
Antidepressant and
Reversible,
Moclobemide MAO-A anxiolytic. It is not
non-covalent.
approved in USA.
22

1.7- MAO-B inhibitors with a possible disease modifying effect

Traditional drug design approach focuses on a single target within disease

pathologic pathway; this discovery tactic is based on the idea of minimizing adverse

effects by designing a highly selective ligand that will avoid interaction with other

unwanted biological targets (cross reactivity) [22, 23]. Such method for drug design and

discovery proved to be incompetent when dealing with diseases that involve complex

pathological picture; a good example for such conditions is the neurodegenerative

diseases. New attempts, to address an effective therapeutic approach for

neurodegenerative diseases, concentrate on designing a ligand that targets multiple

abnormalities within disease pathway. This novel pharmaceutical instrument shows some

successful results regarding disease process modification with no significant elevation in

the incidence of side effects [21, 92]. Below we will mention some compounds that were

designed using designed multiple ligand approach.

A- Rasagiline:

It is an irreversible and selective inhibitor of MAO-B [93]. It was approved by

FDA for Parkinson's disease treatment in 2006 either as an early monotherapy or as an

adjunct treatment to levodopa [11]. Rasagiline exhibits a disease modifying capability

when given in a dose of 1 mg/ day [94]. It seems that the propargylamine moiety is

responsible for the neuroprotective feature of rasagiline; it was observed that the S-

enantiomer is more than 1000 times weaker than rasagiline (R-enantiomer) as a MAO

inhibitor however the S-enantiomer is still able to show neuroprotective activity [95-98].
23

The main metabolite of rasagiline (aminoindan) also shows neuroprotective effect in vitro

studies [98, 99].

The neuroprotective mechanism of propargylamine moiety depends on defending

the mitochondria through several means like activation of protein kinase C (PKC), Bcl-2,

phosphoinositide kinase 3 and mitogen activated protein kinase (MAPK). It also down

regulates pro-apoptotic proteins like Bax, Bad and Fas [93]. Rasagiline also stimulates

the liberation of the soluble form of amyloid precursor protein α (sAPPα) through PKC-

MAP mediated activation of α-secretase [27, 28]. Rasagiline seems to possess

neurorescuing ability through induction of brain derived neurotrophic factors (BDNF)

and glial derived neurotrophic factors (GDNF) [100, 155].

B- Ladostigil:

This drug was designed by combining the propargylamine moiety of rasagiline

with the carbamate cholinesterase inhibitory moiety of rivastigmine. The resultant drug

can inhibit acetylcholine esterase, butyrylcholine esterase, MAO-A and MAO-B [101].

The drug shows a neuroprotective effect similar to that of rasagiline [101, 102].

Ladostigil also shows antidepressant activity through inhibition of MAO-A inside the

brain leading to an elevation in the level of dopamine, norepinephrine and serotonin

[108]. This compound is believed to be effective in the treatment of Alzheimer's disease

and depression.
24

C- HLA20:

This lead compound was prepared by combining the antioxidant chelator moiety

of 8-hydroxyquinoline derivative of the iron chelator VK-28 with the propargylamine

moiety of rasagiline or selegiline [103-105]. This compound exhibits a moderate

selectivity for MAO-B enzyme and also acts as a free radical scavenger. The rationale

behind combining iron chelation effect with MAO-B inhibition emerges from the fact

that brain level of iron and MAO-B increases with age and it also increases in

neurodegenerative diseases as a result of gliosis. The rise in MAO-B level will elevate

the level of oxidative deamination leading to an increase in hydrogen peroxide production

as a byproduct. The resultant hydrogen peroxide will undergo Fenton reaction in the

presence of elevated iron content leading to exaggerated hydroxyl radical level and

inevitable oxidative stress [95, 106].

Based on this scenario M30 compound was introduced, this compound is a potent

inhibitor of MAO-A and MAO-B (inhibits both of them equally) and it has iron chelation

characteristics similar to desferoxamine. The neuroprotection features of M30 are like

those of rasagiline and ladostigil [103, 107].

D- Istradefylline (KW-6002):

It is MAO-B enzyme inhibitor and Adenosine A2a receptor blocker [92].

Adenosine A2a receptor is a G-protein coupled receptor which will stimulate adenylate

cyclase upon activation leading to an increase in cAMP level [109]. This receptor subtype
25

is found mainly in the striatum (GABAergic striatopallidal neurons) where it is co-

localized with dopamine D2 receptor [110-112]. It is believed that activation of

adenosine A2a receptor can produce a functional antagonistic effect on dopamine D2

receptor through reduction in G-protein coupling activity of dopamine D2 receptor and

lowering the binding affinity of dopamine D2 agonist [113, 114]. Because both dopamine

D2 receptor and adenosine A2a receptor are coupled to adenylate cyclase and have some

similarities in signaling system, it is possible that the functional antagonistic effect can

also be mediated at the downstream of signaling pathway (second messenger level) [115].

Based on the localization of adenosine A2a receptor and their functional

relationship with dopamine D2 receptor, it is obvious that the blockade of adenosine A2a

receptor can be used as a non-dopaminergic approach to treat Parkinson's disease.

Blockade of adenosine A2a receptor can improve motor activity in animal models of

Parkinson's disease through strengthening dopamine D2 neurotransmission [116-118].

Some studies showed that adenosine A2a receptor antagonism can also produce a

beneficial motor effect in dopamine D2 receptor knockout mice suggesting that

adenosine A2a receptor antagonism can produce such effect through dopamine D2

receptor independent pathway [119].

Many epidemiological studies had shown that blockade of adenosine A2a

receptors can provide a protective effect against dopaminergic neurons degeneration

observed in Parkinson's disease and MPTP induced neurotoxicity [115, 120, 121].

Caffeine ingestion (non-selective adenosine A1/ A2a receptor blocker) is shown to be


26

associated with a lower tendency to develop Parkinson's disease in men [122-125]. The

neuroprotective effect produced by adenosine A2a receptor antagonists is not fully

understood, a possible explanation for such effect involves the reduction of glutamate

release through adenosine A2a receptor blockade and this can lower neurodegeneration

induced excitotoxicity [115, 120].

E- Pioglitazone and rosiglitazone:

These drugs are used for the treatment of type II diabetes (rosiglitazone is

subjected to usage restrictions due to possible cardiotoxic effects) [126, 127]. These

drugs (Thiazolidinedione TZD) reduce insulin resistance through targeting peroxisome

proliferator activated receptor gamma (PPARγ) [128]. Recently these compounds receive

a lot of attention for a possible use as neuroprotective agents in stroke and Parkinson's

disease [129]. The neuroprotective effect is attributed to three main effects: modulation

of inflammation, MAO-B inhibition and activation of mitoNEET.

These compounds can manipulate inflammatory pathways leading to a reduction

in reactive oxygen species (ROS) production through inhibition of matrix

metallopeptidase 9 and Inhibition of iNOS [92]. They can also increase the level of

CuZn-superoxide dismutase and this can enhance the neuroprotective effect [130]. It had

been shown that pioglitazone can lower the liberation of cytokines in cultured cells upon

lipopolysaccharide treatment [131]. Both rosiglitazone and pioglitazone can inhibit the

deposition of beta amyloid in the brain and lower the release of inflammatory mediator

induced by beta amyloid [132, 156, 157].


27

The neuroprotective effect of pioglitazone and rosiglitazone can be explained

partially through the inhibition of MAO-B enzyme and this will prevent the conversion of

MPTP to the neurotoxic form MPP+ in MPTP Parkinsonian mouse model [133].

Finally pioglitazone can function as a ligand for a mitochondrial protein named

mitoNEET and through this pathway pioglitazone can stabilize mitoNEET and change

the functional state of mitochondria leading to a neuroprotective output. Binding of

ligands to mitoNEET protein can result in depolarization of mitochondrial membrane,

alteration in mitochondrial oxidative condition and attenuation of rotenone toxic effect

[92].
Chapter two: Materials and Methods

2.1- Prospective candidates

Twenty four chemical compounds were purchased from Chembridge online

chemical store (www.hit2lead.com) as possible candidates for MAO-B inhibition. The

chemical structures for these compounds were predicted using similarity analysis

approach which depends on molecular shape comparison using Gaussian function for

volume matching [135]. Some specifications about these candidates are shown in table

2.1. Each compound was dissolved in an appropriate volume of dimethyl sulfoxide

(DMSO) so that a stock solution with 10 mM concentration was produced.

Initially, virtual approaches (docking studies) were used to build an early

impression about the binding tendency of these compounds to both MAO-B and bovine

serum albumin. Based on docking results, these compounds were screened in the

laboratory for both inhibition potential and selectivity against MAO-A and MAO-B. In

order to obtain a better understanding of these compounds' pharmacokinetic profile,

bovine serum albumin (BSA) binding assay and parallel artificial membrane permeability

assay (PAMPA) were performed. Finally in our hope to discover a possible

neuroprotective activity, these compounds were tested for antioxidant effect using

oxygen radical absorbance capacity (ORAC) assay. All these experimental procedures

can facilitate drawing the complete picture of possible scaffolds for potent, selective and

28
29

centrally acting MAO-B inhibitors with disease modifying feature for future use in the

treatment of Parkinson's disease.

Table 2.1- Prospective candidates' specifications.

Mol.

ID code Chemical structure Chemical name Weight

g/ mole

4-[3-(6-chloro-2-hydroxy-

4-phenyl-3-quinolinyl)-5-

7909619 (2-pyridinyl)-4, 5-dihydro- 500.9

1H-pyrazol-1-yl]-4-

oxobutanoic acid.

7, 7'-[1,4-piperazinediylbis

5476423 (methylene)] di (8- 400.5

quinolinol).

4-[(2, 4-dimethyl-1, 3-

thiazol-5-yl) carbonyl]-3-

hydroxy-5-[4-(methylthio)
7747412 451.6
phenyl]-1-(3-pyridinyl

methyl)-1, 5-dihydro-2H-

pyrrol-2-one.
30

Table 2.1- (Continued) Prospective candidates' specifications.

Mol.

ID code Chemical structure Chemical name Weight

g/ mole

2-methyl-5-(3-methyl-4-

oxo-3, 4-dihydro-1-

7655826 phthalazinyl)-N-[2-(4- 442.5

morpholinyl) ethyl]

benzene sulfonamide.

1-[3-(dimethyl amino)

propyl]-4-(2-furoyl)-3-

6656215 hydroxy-5-(3-nitrophenyl)- 399.4

1, 5-dihydro-2H-pyrrol-2-

one.

6-methoxy-N-[3-(4-

6405277 morpholinyl) propyl]-2- 364.5

naphthalene sulfonamide.

1-(4, 5-dimethyl-1, 3-

thiazol-2-yl)-5-(2-furyl)-3-

hydroxy-4-[(7-methoxy-1-
7917584 450.5
benzofuran-2-yl)

carbonyl]-1, 5-dihydro-

2H-pyrrol-2-one.
31

Table 2.1- (Continued) Prospective candidates' specifications.

Mol.

ID code Chemical structure Chemical name Weight

g/ mole

5-(2-fluorophenyl)-3-

hydroxy-4-[(4-methyl-2-

phenyl-1, 3-thiazol-5-yl)
7730595 507.6
carbonyl]-1-[2-(4-

morpholinyl) ethyl]-1, 5-

dihydro-2H-pyrrol-2-one.

1-(3, 4-dimethoxybenzyl)-

3-hydroxy-3-[2-(2-
7839510 467.5
naphthyl)-2-oxoethyl]-1,

3-dihydro-2H-indol-2-one.

3-hydroxy-4-[(7-methoxy-

1-benzofuran-2-yl)

7917586 carbonyl]-1-(5-methyl-3- 430.4

isoxazolyl)-5-phenyl-1, 5-

dihydro-2H-pyrrol-2-one.
32

Table 2.1- (Continued) Prospective candidates' specifications.

Mol.

ID code Chemical structure Chemical name Weight

g/ mole

3-{5-[4-(dimethylamino)

benzylidene]-4-oxo-2-
5140311 336.4
thioxo-1, 3-thiazolidin-3-

yl}propanoic acid.

5-(2, 4-dimethoxy

benzylidene)-2-
5108394 340.4
(phenylimino)-1, 3-

thiazolidin-4-one.

5-(4-chlorobenzylidene)-2-

5108400 (phenylimino)-1, 3- 314.8

thiazolidin-4-one.

1-(3, 4-dihydroxy phenyl)-

2-{[1-(1-naphthyl)-1H-
7919469 378.4
tetrazol-5- yl]

thio}ethanone.
33

Table 2.1- (Continued) Prospective candidates' specifications.

Mol.

ID code Chemical structure Chemical name Weight

g/ mole

5-[(5-phenyl-2-furyl)

5143886 methylene]-2-thioxo-1, 3- 287.4

thiazolidin-4-one.

[5-(4-methoxybenzylidene

)-4-oxo-2-thioxo-1,3-
5140321 309.4
thiazolidin-3-yl]acetic

acid.

5-(1, 3-benzodioxol-5-

ylmethylene)-3-(4-
5144779 355.4
methylphenyl)-2-thioxo-1,

3-thiazolidin-4-one.
34

Table 2.1- (Continued) Prospective candidates' specifications.

Mol.

ID code Chemical structure Chemical name Weight

g/ mole

Ethyl N-(1, 1-dioxido-1, 2-

7542216 benzisothiazol-3-yl)-N- 282

methylglycinate.

5, 6-dimethyl-4-{[(2-

methyl-1, 3-thiazol-4-yl)
7738249 307
methyl]thio}thieno[2,3-d]

pyrimidine.

[(1-methyl-1H-pyrrol-2-

yl)methyl](3-
7929249 238
pyridinylmethyl) amine

hydrochloride.

3-methyl-2-{[2-(4-methyl-

1, 3-thiazol-5-yl)
7954103 317
ethyl]thio}-4(3H)-

quinazolinone.
35

Table 2.1- (Continued) Prospective candidates' specifications.

Mol.

ID code Chemical structure Chemical name Weight

g/ mole

N-[(2-hydroxy-8-methyl-

3-quinolinyl) methyl]-N-
7935085 372.4
(4-methoxyphenyl)

methane sulfonamide.

[5-(anilinomethylene)-4-

oxo-2-thioxo-1, 3-
5124176 294.4
thiazolidin-3-yl] acetic

acid.

Methyl [(4-methyl-5, 6, 7,

7697413 8-tetrahydro-2- 252

quinazolinyl)thio] acetate.
36

2.2- Docking studies

The 2D and 3D structures of the tested compounds were prepared using

MarvinSketch 6.0.3 (www.chemaxon.com). Human MAO-B crystal (2BK3) and bovine

serum albumin (BSA) crystal (4F5S) were obtained from RCSB Protein Data Bank

(www.rcsb.org). Both MAO-B and BSA crystals were prepared for docking using UCSF

Chimera 1.8 (www.cgl.ucsf.edu/chimera) [150] and AutoDockTools (ADT) 1.5.6

(http://mgltools.scripps.edu) [151, 152]. ADT 1.5.6 was also used to prepare the tested

compounds (ligands) for docking operation. Docking of the ligands into these targets

(crystals) was done using AutoDock 4.2 (http://autodock.scripps.edu) [151]. The final

results were analyzed using AutoDockTools (ADT) 1.5.6, LigPlot+ v.1.4.5 [153], UCSF

Chimera 1.8 and PyMOL v.1.3 (www.schrodinger.com) [154].

2.3- Monoamine oxidase inhibition assay

This assay measure the compound's inhibition strength and degree of selectivity

toward MAO-A and MAO-B. The basic concept of this assay involves the ability of both

MAO-A and MAO-B to convert the non-fluorescent and non-selective substrate

(kynuramine) into the intermediate 3-(2-aminophenyl)-3-oxo-propionaldehyde which will

undergo spontaneous intramolecular condensation leading to the formation of the

fluorescent product 4-Hydroxyquinoline (4-HQ) [136]. Based on this concept, the level

of MAO activity can be determined through measuring the fluorescence level of 4-

Hydroxyquinoline (4-HQ). In the presence of the tested compound, any reduction in


37

fluorescence level will reflect the inhibition capacity of that compound. Figure 2.1 shows

the basic principle of this assay.

In this test, KPO4 solution (0.1 M, pH 7.4) was used as a buffer which was

warmed to 37˚ C prior to performing the test. Falcon black 96 well plates (Cat#353241)

were also used in this test. Kynuramine was purchased from Sigma-Aldrich (Cat#: k-

3250) and a stock solution with a concentration of 25 mM was prepared by dissolving

kynuramine in the appropriate volume of distilled water. This stock solution was further

diluted with KPO4 buffer to yield a final concentration that is close to the Km value for

the reaction catalyzed by either MAO-A or MAO-B (the final concentration of

kynuramine solution after dilution was 40 µM for MAO-A and 20 µM for MAO-B).

Human MAO-A and human MAO-B were purchased from Fisher Scientific

(www.fishersci.com). These enzymes were used in this assay as 5 mg/ ml stock solutions

and in order to minimize thermal induced inactivation of MAO, both enzymes were

stored at -80˚ C and they even kept on ice till their employment in the assay. The stock

solutions of MAO-A and MAO-B were further diluted with KPO4 buffer to obtain final

solutions with concentrations of 0.006 mg/ ml for MAO-A and 0.015 mg/ ml for MAO-

B. 2 N NaOH was used as a stopping reagent by causing protein denaturation.

In this assay, the tested compounds were serially diluted with KPO4 buffer (100

µl/ well in duplicate) across the plate to produce eight points dose inhibition curve. As

mentioned previously, the tested compounds were dissolved in DMSO and the final

concentration of DMSO in the assay was less than 0.5%. It is reported that DMSO shows
38

the least MAO inhibitory effect. The enzyme activity (fluorescence level) in the absence

of any inhibitor (only DMSO) was considered as a control (100% MAO activity). After

the compounds were serially diluted, the plate was covered and incubated at 37˚ C for 10

minutes so that both the compounds and the plate can reach a temperature of 37˚ C. Then

the substrate (kynuramine) was added as 50 µl/ well after that the enzyme (either MAO-

A or MAO-B) was added as 50 µl/ well in order to start the reaction. The plate was then

covered and incubated at 37˚ C for 20 minutes in order to obtain the ultimate level of

activity for the enzyme. The final assay volume was 200 µl per well. After incubation

period, 75 µl of 2 N NaOH solution was added per well to stop the reaction. The

fluorescence level of 4-Hydroxyquinoline was measured at 310/ 380 nm wavelength pair

by using a Flourometer (top reader, BMG-FlouStar).


39

Figure 2.1- Conversion of kynuramine into 4-Hydroxyquinoline as catalyzed by both

MAO-A and MAO-B, this figure was adapted from reference [137].
40

2.4- Bovine serum albumin (BSA) binding assay

This assay measures the binding affinity of the tested compounds toward serum

bovine albumin. Fluorescence quenching represents the main principle of this assay and it

involves the reduction in fluorescence level (quantum yield) of a fluorophore upon

interaction with a quencher molecule [138, 139]. Bovine serum albumin (BSA) shows

intrinsic fluorescence activity (fluorophore) due to the presence of two types of aromatic

amino acids (tryptophan and tyrosine). When BSA is excited at 280 nm, it will show a

strong fluorescence emission at 342 nm and this emission is mainly attributed to

tryptophan [140-142]. This test is based on the idea that higher binding of the tested

compound to BSA will result in greater quenching of the fluorescence activity of BSA

and this can be measured experimentally.

In this assay, KPO4 solution (0.1 M, pH 7.4) was used as a buffer. Bovine serum

albumin was obtained from Boston BioProducts (Ashland, MA, USA). A stock solution

of BSA (1 mg/ ml) was prepared by dissolving BSA in KPO4 buffer just before starting

the test. Falcon black 96 well plates (Cat# 353241) were used for this test. The final assay

volume was 200 µl/ well. The tested compounds were serially diluted (100 µl/ well in

duplicate) with KPO4 buffer across the plate so that the desired range of concentrations

(an eight points data curve) was obtained. As mentioned before, dimethyl sulfoxide

(DMSO) was used to dissolve the tested compounds and the fluorescence level of BSA in

the absence of any tested compound (only DMSO) was considered as a control (no BSA

binding). The stock solution of BSA was added as 100 µl/ well (the final concentration of
41

BSA in each well was 7.5 µM) after that the plate was covered and incubated at room

temperature for 30 minutes. After incubation, the plate was read at 280/ 340 nm

wavelength pair using a Flourometer (BMG-FlouStar).

2.5- Parallel Artificial Membrane Permeability Assay (PAMPA)

This assay represents a high throughput approach to predict passive, transcellular

permeability of the tested compounds in early stages of drug development process. This

assay is cheap, highly successful and reproducible [143]. It uses a synthetic lipid layer

immobilized on filter support to simulate the passive permeability of the compound

across different biological barriers (blood brain barrier, skin and gastrointestinal tract)

[143-146]. This assay can't determine active transport of the compound across biological

membranes [144].

The Hexadecane Method of PAMPA (HDM-PAMPA) measures the passive

diffusion of the tested compound from the donor compartment (which contains the

desired concentration of the compound dissolved in buffer medium) to an acceptor

compartment (compound free compartment, contains only buffer medium). These two

compartments are separated by a hexadecane liquid layer on a polycarbonate membrane

support. The concentration of the tested compound can be determined in these two

compartments by measuring the fluorescence level for the compound.

For this assay, 5% DMSO in phosphate buffered saline pH 7.4 was used as a

buffer system. Hexadecane and hexane were obtained from commercial sources and a 5%
42

(v/v) hexadecane in hexane mixture was prepared. Compound under investigation

(5140311) was diluted with buffer system (5% DMSO in BPS) to yield a final solution

with a concentration of 500 µM. A 96 well MultiScreen filter plate for permeability

(donor plate) was ordered from Millipore Corporation (Billerica, MA, USA, Cat#

MAPBMN310). The 96 well acceptor plate was also obtained from Millipore (Cat#

MSSACCEPTOR).

The assay was initiated by the addition of 15 µl/ well of 5% (v/v) hexadecane in

hexane solution into the donor plate; the plate was then placed uncovered in a fume hood

for one hour in order to ensure that hexane was completely evaporated and a fine layer of

hexadecane was generated on polycarbonate film. This was followed by the addition of

buffer solution (5% DMSO in PBS) into the acceptor plate as 300 µl per each well. The

donor plate was then placed on the acceptor plate so that the underside of the membrane

for donor plate is in contact with the buffer solution in each well of the acceptor plate.

The compound (5140311) solution was added to the donor plate as 150 µl/ well in

triplicate. After that, the donor plate was covered with its lid and incubated at room

temperature for a period of four hours. During incubation period, equilibrium solution for

compound (5140311) was prepared by simply mixing donor compartment volume (150

µl of compound solution) with acceptor compartment volume (300 µl of buffer solution).

After incubation period, 250 µl was transferred from each well of the acceptor plate into

Falcon black 96 well plate (Cat# 353241). A similar volume of equilibrium solution for

compound (5140311) was transferred into the same 96 well black plate. Also compound

(5140311) solution was serially diluted with assay buffer across the black plate to yield a
43

suitable eight points fluorescence-concentration curve. The generation of such curve will

help us to predict the concentration of the compound in the acceptor compartment based

on the observed fluorescence (interpolation). The fluorescence level of the compound

(5140311) was measured at 540/ 620 nm wavelength pair by using FLX 800 microplate

fluorescence reader (Biotek instruments).

2.6- Oxygen Radical Absorbance Capacity (ORAC) assay

This assay measures the ability of the tested compounds to scavenge peroxyl

radicals that are generated by 2,2'-azobis(2-amidinopropane) dihydrochloride (AAPH),

thereby it can screen these compounds for any possible antioxidant activity [147, 148].

The assay principle is based on the idea that the presence of peroxyl radicals can lower

the fluorescence level of a fluorophore (fluorescein) as a result of oxidative degradation.

A compound with antioxidant power can preserve the fluorescence activity by reducing

the oxidative degradation of fluorescein [149].

For this assay, KPO4 solution (0.1 M, pH 7.4) was used as a buffer, Falcon black

96 well plates (Cat# 353241) were also used and the final volume for this assay was 200

µl per each well. AAPH was obtained from Sigma-Aldrich (St. Louis, MO, USA) and it

was stored in a dry environment. Before starting the assay, AAPH was dissolved in an

appropriate volume of KPO4 buffer to yield a stock solution with a concentration of 72

mM. This stock solution was kept on ice until assay initiation. Fluorescein was also

purchased from Sigma-Aldrich (St. Louis, MO, USA) and prior to assay initiation, it was

also dissolved in KPO4 buffer to prepare a stock solution with a concentration of 112
44

nM. Vitamin C (L-ascorbic acid) was also ordered from Sigma-Aldrich (St. Louis, MO,

USA); before starting the test, it was dissolved in KPO4 buffer to obtain a solution with a

concentration of 10 mM. Vitamin C was used in this assay as a reference substance

(positive control). As mentioned previously, DMSO was used as a vehicle for the tested

compounds, the fluorescence level of fluorescein in the absence of any tested compound

(only DMSO) was considered as a negative control.

The compounds under investigation were serially diluted with KPO4 buffer across

the assay plate (20 µl/ well in duplicate) to yield the required range of concentrations.

Fluorescein solution was then added as 80 µl per each well. This was followed by the

addition of peroxyl radical generator (AAPH) as 100 µl per each well. The plate was then

covered and incubated at room temperature for one hour. After that, the fluorescence

level was read at 485 nm excitation and 528 nm emission wavelengths through using

FLX 800 microplate fluorescence reader (Biotek instruments).

2.7- Data analysis

Most of the results for the previously mentioned experiments were displayed as

(Mean ± SEM). Prism 5 (www.graphpad.com) was used for columns generation, curves

fitting (nonlinear regression) and lines fitting (linear regression).


Chapter three: Results

3.1- Selection of the most efficient candidates

Based on the results of our experiments, a scatter plot was generated by plotting

MAO-B enzyme inhibition potential (capacity) against antioxidant potential (capacity) of

the prospective candidates. The scatter plot is shown in figure 3.1 and it was generated by

using 100µM concentration of each compound in MAO-B inhibition assay and 20µM

concentration of each compound in antioxidant (ORAC) assay.

By careful examination of this plot, we can easily choose the most efficient

candidates that display a perfect combination of both MAO-B inhibition capacity and

antioxidant effect. According to the scatter graph, six compounds show such efficient

combination and the identification codes for these chemicals are: 5143886, 5140311,

7730595, 5140321, 7919469 and 7747412. In this section we will focus only on the

results of these compounds since they represent encouraging candidates for possible

antiparkinsonian agents with disease modifying capability. We will also mention the

results for additional two chemicals and these are: 5476423 and 7954103. According to

the plot, compound 5476423 shows excellent antioxidant behavior with minor MAO-B

inhibition ability. On the other hand, compound 7954103 shows a potent MAO-B

inhibition effect with minor antioxidant activity.

45
46

Figure 3.1- A scatter plot of MAO-B inhibition potential (percentage) against antioxidant

potential (percentage) for our chemicals. For antioxidant potential, vitamin C was used as

a reference (100% potential).


47

3.2- Docking studies results

In order to obtain preliminary expectations about the pharmacodynamics and

pharmacokinetics characteristics of our candidates, docking analyses were performed by

targeting human MAO-B crystal and bovine serum albumin crystal respectively. Docking

studies can also give us a virtual insight into compound's orientation within the active site

of our target.

Based on our results of docking studies, it is obvious that compounds: 5140311,

5140321, 5143886, 7919469 and 7954103 exhibit an extended conformation that will

occupy both the entrance cavity and the substrate cavity within the active site of MAO-B

crystal. Such orientation will force Ile 199 residue to gain an open configuration leading

to the fusion of both entrance cavity and substrate cavity. The results of docking studies

for these five compounds are shown in figure 3.2, 3.3, 3.4, 3.5 and 3.6 respectively.

According to these figures, we can easily notice that these five candidates are situated in

the active site with a position that directly facing the FAD moiety. In figure 3.2, we can

recognize that there is a hydrogen bond between the oxygen atom attached to position

four of the 2-thioxo-1, 3-thiazolidine moiety of compound 5140311 and the hydroxyl

group of phenyl moiety in Tyr 435 (residue of aromatic cage). Another important finding

to mention is the presence of two hydrogen bonds in the docking results of compound

7919469 as can be seen in figure 3.5. The first hydrogen bond is formed between the

nitrogen atom at position three of tetrazole moiety of compound 7919469 and the

hydroxyl group in carboxylic acid moiety of Phe 168. The second and most important
48

hydrogen bond is formed between the p-hydroxyl group of dihydroxy phenyl moiety in

compound 7919469 and N (5) in the isoalloxazine ring system of FAD moiety. For these

five compounds, hydrophobic interactions with residues of active site contribute

significantly to the binding energy.

The results for the three remaining compounds: 5476423, 7730595 and 7747412

are shown in figure 3.7, 3.8 and 3.9 respectively. From these results, we can see clearly

that these compounds do not enter to the active site of MAO-B crystal. These three

compounds occupy a position close to the gating switch (loop 99-112) and through

hydrophobic interactions with the amino acid residues in this loop (mainly Phe 103 and

Pro 102); these compounds can hinder the entrance of substrates into the entrance cavity

of MAO-B enzyme. The values for energy of binding and dissociation constant (KI) for

the last three compounds are higher than those for the first five compounds and this may

be attributed to the difference in the location of binding.


49

Figure 3.2(A)- Two dimensional representation for the docking of compound 5140311

into MAO-B crystal. This image was generated by LigPlot+ v.1.4.5 [153]. Carbon,

oxygen, nitrogen and sulfur atoms are represented by black, red, blue and yellow colors

respectively. Hydrogen bonds are represented by green dashed line while hydrophobic

interactions are drawn as small multiple red lines.


50

Figure 3.2(B)- Three dimensional representation for the docking of compound 5140311

into MAO-B crystal. This image was generated by PyMOL v.1.3

(www.schrodinger.com) [154]. Our ligand is colored by red and the hydrogen bond is

shown as a green line.


51

Figure 3.3(A)- Two dimensional representation for the docking of compound 5140321

into MAO-B crystal. This image was generated by LigPlot+ v.1.4.5 [153]. Carbon,

oxygen, nitrogen and sulfur atoms are represented by black, red, blue and yellow colors

respectively. Hydrophobic interactions are drawn as small multiple red lines.


52

Figure 3.3(B)- Three dimensional representation for the docking of compound 5140321

into MAO-B crystal. This image was generated by PyMOL v.1.3

(www.schrodinger.com) [154]. Our ligand is colored by red.


53

Figure 3.4(A)- Two dimensional representation for the docking of compound 5143886

into MAO-B crystal. This image was generated by LigPlot+ v.1.4.5 [153]. Carbon,

oxygen, nitrogen and sulfur atoms are represented by black, red, blue and yellow colors

respectively. Hydrophobic interactions are drawn as small multiple red lines.


54

Figure 3.4(B)- Three dimensional representation for the docking of compound 5143886

into MAO-B crystal. This image was generated by PyMOL v.1.3

(www.schrodinger.com) [154]. Our ligand is colored by red.


55

Figure 3.5(A)- Two dimensional representation for the docking of compound 7919469

into MAO-B crystal. This image was generated by LigPlot+ v.1.4.5 [153]. Carbon,

oxygen, nitrogen and sulfur atoms are represented by black, red, blue and yellow colors

respectively. Hydrogen bonds are represented by green dashed line while hydrophobic

interactions are drawn as small multiple red lines.


56

Figure 3.5(B)- Three dimensional representation for the docking of compound 7919469

into MAO-B crystal. This image was generated by PyMOL v.1.3

(www.schrodinger.com) [154]. Our ligand is colored by red and the hydrogen bond is

shown as a green line.


57

Figure 3.6(A)- Two dimensional representation for the docking of compound 7954103

into MAO-B crystal. This image was generated by LigPlot+ v.1.4.5 [153]. Carbon,

oxygen, nitrogen and sulfur atoms are represented by black, red, blue and yellow colors

respectively. Hydrophobic interactions are drawn as small multiple red lines.


58

Figure 3.6(B)- Three dimensional representation for the docking of compound 7954103

into MAO-B crystal. This image was generated by PyMOL v.1.3

(www.schrodinger.com) [154]. Our ligand is colored by red.


59

Figure 3.7(A)- Two dimensional representation for the docking of compound 5476423

into MAO-B crystal. This image was generated by LigPlot+ v.1.4.5 [153]. Carbon,

oxygen and nitrogen atoms are represented by black, red and blue colors respectively.

Hydrophobic interactions are drawn as small multiple red lines.


60

Figure 3.7(B)- Three dimensional representation for the docking of compound 5476423

into MAO-B crystal. This image was generated by PyMOL v.1.3

(www.schrodinger.com) [154]. Our ligand is colored by red.


61

Figure 3.7(C)- This figure shows the location of binding for compound 5476423 (red

color) according to docking studies in MAO-B crystal. This compound occupies a

location close to the gating loop and may interfere with substrates accessibility to the

active site. This figure was generated by using UCSF Chimera 1.8

(www.cgl.ucsf.edu/chimera) [150].
62

Figure 3.7(D)- A close view for the previous figure, Here we can see how compound

5476423 (red color) is located near the gating loop and may interact with residues located

in that loop like Pro 102 and Phe 103. This figure was generated by using UCSF Chimera

1.8 (www.cgl.ucsf.edu/chimera) [150].


63

Figure 3.8(A)- Two dimensional representation for the docking of compound 7730595

into MAO-B crystal. This image was generated by LigPlot+ v.1.4.5 [153]. Carbon,

oxygen, nitrogen, fluoride and sulfur atoms are represented by black, red, blue, green and

yellow colors respectively. Hydrophobic interactions are drawn as small multiple red

lines.
64

Figure 3.8(B)- Three dimensional representation for the docking of compound 7730595

into MAO-B crystal. This image was generated by PyMOL v.1.3

(www.schrodinger.com) [154]. Our ligand is colored by red.


65

Figure 3.8(C)- A close view for the binding position of compound 7730595 (red color) as

indicated by docking studies using MAO-B crystal as a target. As shown here the

compound is located close to the gating loop and may be involved in interactions with

residues located within the gating loop like Pro 102 and Phe 103. This figure was

generated by using UCSF Chimera 1.8 (www.cgl.ucsf.edu/chimera) [150].


66

Figure 3.9(A)- Two dimensional representation for the docking of compound 7747412

into MAO-B crystal. This image was generated by LigPlot+ v.1.4.5 [153]. Carbon,

oxygen, nitrogen, fluoride and sulfur atoms are represented by black, red, blue, green and

yellow colors respectively. Hydrophobic interactions are drawn as small multiple red

lines. The green dashed line represents hydrogen bond between the amine group at

position two of Threonine 478 residue and the hydroxyl group attached to position three

at the 1, 5- dihydro-pyrrol-2-one moiety of compound 7747412.


67

Figure 3.9(B)- Three dimensional representation for the docking of compound 7747412

into MAO-B crystal. This image was generated by PyMOL v.1.3

(www.schrodinger.com) [154]. Our ligand is colored by red and the hydrogen bond is

shown as a green line.


68

Figure 3.9(C)- A close view for the binding position of compound 7747412 (red color) as

indicated by docking studies using MAO-B crystal as a target. As shown here the

compound is located close to the gating loop and may be involved in hydrophobic

interactions with residues located within the gating loop like Pro 102 and Phe 103. The

green line represents hydrogen bond between Thr 478 residue and our compound. This

figure was generated by using UCSF Chimera 1.8 (www.cgl.ucsf.edu/chimera) [150].


69

Because the main binding sites in serum albumin for aromatic and heterocyclic

compounds are subdomain IIA (site I) and subdomain IIIA (site II) [158,159], docking

studies in BSA crystal were targeted toward these two binding sites only. By taking the

virtual binding parameters (energy of binding and dissociation constant) into our

consideration, it is clear that our candidates bind efficiently with nearly equal affinity to

site I and site II. The only compound that deviates from this established principle is

7919469, which shows some preference for binding to site I over site II. Table 3.1 shows

virtual binding parameters for our selected compounds in BSA docking studies.
70

Table 3.1- Docking results for BSA crystal.

Subdomain IIA Subdomain IIIA

Compound ID Energy of binding KI Energy of binding KI

(kcal/mol) (µM) (kcal/mol) (µM)

5140311 -5.53 89.11 -5.12 177.88

5140321 -5.78 58.01 -5.07 192.7

5143886 -6.35 22.03 -7.01 7.31

7919469 -7.52 3.1 -4.78 311.84

7954103 -5.97 42.09 -5.66 70.47

5476423 -5.98 41.7 -5.71 65.74

7730595 -5.29 131.79 -5.76 60.34

7747412 -4.96 232.32 -4.7 361.79


71

3.3- Monoamine oxidase inhibition assay results

The results of this assay provide a clear insight into the potential and selectivity of

inhibition produced by our candidates against MAO enzyme isoforms. Our results for this

test are summarized in figure 3.10, figure 3.11 and table 3.2. Figure 3.10 describes the

strength of MAO-B inhibition (potency of inhibition) produced by using 100µM

concentration of each selected compound. According to this figure, the highest inhibition

is produced by compound 5143886 where MAO-B activity falls down to a percentage

value of (2.148 ± 0.2697) and statistically this value represents (Mean ± SEM). The

lowest inhibition is produced by compound 5476423 where the activity of MAO-B

enzyme only falls down to a percentage value of (78.99 ± 0.1677). The remaining

compounds produce a strong inhibitory effect which generally lowers the activity of

MAO-B to below 50% level.

On the other hand, figure 3.11 shows the potency of the inhibitory effect against

MAO-A for only four compounds (5140311, 5140321, 5143886 and 5476423). This

potency of effect was generated by using a concentration of 100µM for each tested

compound. In this figure, the strongest inhibitory effect is produced by compound

5143886 with a reduction in MAO-A level of activity into a percentage value of (42.37 ±

1.737) while the weakest effect is produced again by compound 5476423 with a

reduction in MAO-A activity into a percentage value of (78.81 ± 0.4983). The other two

compounds appear to be able to lower enzyme activity to around 60%. By comparing

data from figure 3.10 with those in figure 3.11, we can obtain some information about the
72

selectivity in action of the tested compounds against MAO enzyme isoforms. It is very

clear that compound 5476423 shows no preference in its inhibitory effect against MAO

enzyme isoforms. Compounds 5140311 and 5143886 appear to be highly selective for

MAO-B inhibition while compound 5140321 shows a slight preference for the inhibition

of MAO-B over MAO-A.

Table 3.2 gives us information about the degree of inhibition selectivity toward

MAO-B and MAO-A isozymes. This table compares the IC50 value for MAO-B

inhibitory effect against that value for MAO-A inhibitory effect. Unfortunately, we were

only able to determine MAO-A IC50 for compounds 5140311 and 5476423 due to

technical restrictions. Compound 5140311 seems to be highly selective for inhibiting

MAO-B enzyme since the MAO-B IC50 value is about 16 times lower than IC50 value

for MAO-A.
73

Figure 3.10- MAO-B inhibition potential as produced by our compounds. Values are

presented as (Mean ± SEM). The concentration used for each compound was 100µM.
74

Figure 3.11- The potency of the inhibitory effect against MAO-A as produced by the

tested compounds. Results are presented as (Mean ± SEM); the concentration used to

generate such effect is 100µM.


75

Table 3.2- Inhibition selectivity toward MAO isozymes.

Compound ID MAO-B IC50 µM MAO-A IC50 µM

5140311 9.71 155

5140321 105 N/A

5143886 2.64 N/A

7919469 1.19 N/A

7730595 62.8 N/A

7747412 94.3 N/A

5476423 N/A 242

7954103 6.27 N/A


76

3.4- Bovine serum albumin (BSA) binding assay results

This assay provides a general view about compounds' degree of binding to serum

albumin and the influence of such interaction on the pharmacokinetics features of these

selected compounds. The binding potentials for these chemicals are shown in figure 3.12.

The results in this figure were generated by using 100µM concentration for each tested

compound; the outcomes in this figure were reported as (Mean ± SEM).

Examination of this figure reveals that these compounds, in general, show high

tendency for BSA binding. The lowest binding percentage is produced by compound

5476423 with a binding value of (44.16 ± 5.205) while the highest binding percentage is

produced by compound 5143886 with a value of (97.57 ± 0.1944). With the exception of

compound 5140311, all the remaining compounds show high degree of BSA binding that

is greater than 70%.

By using different concentrations for each compound (serial dilution), we were

able to generate a relative BSA binding curve (nonlinear regression). This curve was

utilized to obtain an estimation of EC50 value. Figure 3.13 shows the BSA binding curve

for each compound together with its calculated EC50. With some exceptions these EC50

values can be correlated with those results presented in figure 3.12. For instance, the

smallest EC50 values (highest binding) were calculated for compounds 5143886 and

5140321 while the largest value (least binding) was reported for compound 5476423.
77

Figure 3.12- BSA binding potential of the tested chemicals. These results were generated

by using a concentration of 100µM for each compound and the outcomes were presented

as (Mean ± SEM).
78

Figure 3.13- Relative BSA binding curves for the selected compounds. The calculated

EC50 values are also presented with these curves.


79

Figure 3.13- (Continued) Relative BSA binding curves for the selected compounds. The

calculated EC50 values are also presented with these curves.


80

Figure 3.13- (Continued) Relative BSA binding curves for the selected compounds. The

calculated EC50 values are also presented with these curves.


81

Figure 3.13- (Continued) Relative BSA binding curves for the selected compounds. The

calculated EC50 values are also presented with these curves.


82

3.5- Parallel artificial membrane permeability assay (PAMPA) results

This assay gives us an early prediction about the ability of the compound (under

investigation) to pass the blood brain barrier and reach CNS. This assay was only applied

to compound 5140311 because it was the only one that fluoresced at the excitation wave

length range provided by the fluorescence reader. By the end of this assay, the

fluorescence level was determined for both the acceptor compartment and equilibrium

solution as this level of fluorescence reflects the concentration of compound 5140311 in

these solutions. The results of fluorescence reader were then applied to the following

equation (provided by the manufacturer of assay apparatus) in order to calculate the rate

of permeation or effective permeability (Pe):

[𝑑𝑟𝑢𝑔]𝑎𝑐𝑐𝑒𝑝𝑡𝑜𝑟
log 𝑃𝑒 = log {𝐶 ∗ −ln(1 − )}
[𝑑𝑟𝑢𝑔]𝑒𝑞𝑢𝑖𝑙𝑖𝑏𝑟𝑖𝑢𝑚

𝑉𝐷 ∗𝑉𝐴
Where 𝐶 = ( )
(𝑉𝐷 +𝑉𝐴 ) 𝐴𝑟𝑒𝑎∗𝑡𝑖𝑚𝑒

𝑉𝐷 = Volume of donor compartment= 0.15 cm³.

𝑉𝐴 = Volume of acceptor compartment= 0.3 cm³.

Area= Active surface area of membrane= 0.048 cm².

Time= Incubation time for the assay and in our case it was four hours= 14400 seconds.
83

By applying all of the previously mentioned variables into our equation we get the

following:

0.15∗0.3
𝐶 = ((0.15+0.3) )
0.048∗14400

C = 1.446 × 10−4 cm s −1

611
log 𝑃𝑒 = log {(1.446 × 10−4 ) ∗ −ln(1 − )}
17013

Log Pe = -5.276 cm s −1

Pe = 5.2966× 10−6 cm s −1

According to the classification of PAMPA-BBB results (based on Pe ranges)

[160], it is very clear that our compound 5140311 is highly permeable through blood

brain barrier since our calculated Pe value is greater than 4.0 × 10−6 cm s −1. As we

mentioned previously in the materials and methods chapter (chapter two), compound

5140311 was serially diluted to generate a standard curve (eight points fluorescence-

concentration curve). We can estimate the concentration of our compound in the acceptor

compartment based on the measured level of fluorescence (interpolation using the

standard curve). In our case the concentration of compound 5140311 in the acceptor

compartment was 2.098125µM.


84

3.6- Oxygen radical absorbance capacity (ORAC) assay results

This assay predicts the antioxidant capacity of compound under investigation

through screening the ability of that compound to scavenge peroxyl radicals that are

generated by AAPH (radical initiator) [147, 148]. By scavenging these radicals, our

compound will be able to prevent the degradation of fluorescein thereby preserving the

fluorescence level of this fluorophore [149]. According to this principle, the fluorescence

level will reflect the antioxidant capacity of the tested compound.

The results for this assay are summarized in figure 3.14; these results were

obtained by using 20µM concentration of each tested compound. The outcomes are

presented as (Mean ± SEM); vitamin C (Ascorbic acid) was used as a positive control

(100% activity). In this figure, all the tested compounds show excellent antioxidant

potentials that are either equivalent or higher than that reported for Ascorbic acid (with

the exception of compound 7954103). The highest radical scavenging capacity was

reported for compound 7730595 with a percentage value of (205.2 ± 1.952) while the

least scavenging capacity was observed in compound 7954103 with a percentage value of

(31.24 ± 0.2169).

In this assay, serial dilution was used for each compound in order to generate

eight points fluorescence – concentration curve (nonlinear regression). The generation of

such curve helped us to estimate the value of EC50 for each compound. Unfortunately,

we were unable to correlate the calculated EC50 values (shown in table 3.3) with results
85

in figure 3.14. This may be due to technical restrictions within curve fitting (nonlinear

regression).

Figure 3.14- Percentage of antioxidant power for the selected chemicals. This figure was

generated by using 20µM concentration for each compound. The outcomes are presented

as (Mean ± SEM); Ascorbic acid was used as a positive control (100% activity).
86

Table 3.3- EC50 values for (antioxidant capacity – concentration) curve of each tested

compound.

Compound ID EC50 µM

Ascorbic acid 5.570

5140311 0.5748

5140321 3.698

5143886 N/A

7919469 12.56

7730595 0.9124

7747412 0.6416

5476423 3.256

7954103 3.491
87

3.7- Structure activity relationship (SAR) study results

We established a basic structure activity relationship (SAR) study as an attempt to

build a hypothetical scaffold for the development of potent, selective and centrally active

MAO-B inhibitors with a possible neuroprotective effect that can be applied for the

treatment of Parkinson's disease. Through comparing the structure and activity for

compounds with related building blocks, we tried to identify the structural units that are

essential for MAO-B inhibition and those that are critical for peroxyl radical quenching.

For simplicity in presentation, the data of the scatter plot in figure 3.1 are re-

introduced here in the form of columns (only for compounds under investigation in SAR

study). Figures 3.15, 3.16 and 3.17 show the relative activity for the concerned

compounds (MAO-B inhibition activity and antioxidant potential) together with the

structural formula of these compounds.

Based on figure 3.15, it is very easy to notice that both compounds 5140311 and

5140321 have almost the same antioxidant potential, compound 5140311 appears to have

a stronger MAO-B inhibition effect. This variation in MAO-B inhibition potential can be

attributed in part to the difference in the substitution type at position four of the

benzylidene moiety in these compounds. The dimethyl amino group in compound

5140311 has more electron donating effect than does the methoxy moiety in compound

5140321; this may provide a more favorable position and electron density for compound

5140311 within MAO-B active site so that a hydrogen bond can be formed between

oxygen atom (attached to position four of the 2-thioxo-1, 3-thiazolidine ring) and the
88

hydroxyl group of phenyl moiety in Tyr 435 residue. The presence of such hydrogen

bonding was confirmed for compound 5140311 by using docking analysis. Compounds

5140311 and 5140321 also show difference in substitution length at position three of the

2-thioxo-1, 3-thiazolidin-4-one ring; longer substitution (propanoic acid in 5140311

versus acetic acid in 5140321) may have a positive impact on the potential and selectivity

of MAO-B inhibition effect by providing a more extended conformation.


89

Figure 3.15(A)- The potency of the inhibitory effect against MAO-B as produced by

compounds 5140311 and 5140321; the data are displayed as (Mean ± SEM).
90

Figure 3.15(B)- Percentage of peroxyl radical quenching potential (antioxidant) for

compounds 5140311 and 5140321, values are presented as (Mean ± SEM) and Ascorbic

acid was used as positive control (100% activity).


91

Figure 3.15(C)- Chemical structural formula for compounds 5140311 and 5140321.
92

Figure 3.15(D)- Two dimensional representation for the docking of compound 5140311

into human MAO-B crystal, this image was generated by using LigPlot+ v.1.4.5 [153].
93

Figure 3.15(E)- Two dimensional representation for the docking of compound 5140321

into human MAO-B crystal, this image was generated by using LigPlot+ v.1.4.5 [153].
94

In figure 3.16(A), only compounds 5143886 and 5108394 show high MAO-B

inhibition potential. The other two compounds 5144779 and 5108400 have a minor

MAO-B inhibitory effect. Compounds 5108394 and 5108400 are very similar in terms of

structural formula but they show significant difference regarding MAO-B inhibition

effect (in vitro). Structural comparison for these two compounds show that the only

difference between them is the presence of two electron donating groups (methoxy

group) attached to positions two and four of the benzylidene moiety in compound

5108394 while compound 5108400 has an electron withdrawing group (chlorine atom)

attached to position four of the benzylidene moiety. This variation in substituent type will

affect the orientation and the electron density of these compounds within MAO-B active

site. Docking studies showed that compound 5108394 occupies a position opposite in

direction to that occupied by compound 5108400 within MAO-B crystal; such orientation

will favor the formation of hydrogen bond between oxygen atom attached to position four

of 1, 3-thiazolidine ring in compound 5108394 and the sulfhydryl group attached to

position three of Cysteine 172 residue within MAO-B active site. The reversed

orientation in compound 5108400 will facilitate the formation of hydrogen bond between

the sulfur atom at position one of 1, 3-thiazolidin-4-one ring and the sulfhydryl group in

Cysteine 172 residue, this hydrogen bond is weaker than that formed in case of

compound 5108394 and this may account for the difference in MAO-B inhibition

potential.

Previous studies showed that nitrogen atom in position three and oxygen atom

attached to position four of the thiazolidinedione ring within pioglitazone molecule are
95

involved in hydrogen bonding with conserved active site water molecules within the

aromatic cage of Tyr398 and Tyr435 [128]. We believe that such hydrogen bonding can

also exist between the conserved active site water molecules and (nitrogen atom in

position three and oxygen atom attached to position four of the 1, 3-thiazolidine ring).

We expect that attachment of 4-methylphenyl group to the nitrogen atom at position three

of the 2-thioxo-1, 3-thiazolidin-4-one ring in compound 5144779 can hinder the

formation of hydrogen bond with conserved active site water molecule and this may

adversely impact MAO-B inhibitory effect.

In figure 3.16(B), we can clearly notice that only compound 5143886 has peroxyl

radical quenching activity, the other three compounds (5144779, 5108400 and 5108394)

have very low or almost no antioxidant activity. Chain breaking antioxidants produce

their radicals scavenging activity through donating hydrogen atoms (radicals reduction)

and formation of stable radicals that are unable to continue chain reaction [149, 182]. We

believe that nitrogen atom at position three of 2-thioxo-1, 3-thiazolidin-4-one ring

represents a good hydrogen donor, and we also expect that the presence of thioxo group

at position two of 1, 3-thiazolidin-4-one ring is considered essential for antioxidant

activity. Such criteria are readily available in compound 5143886. Similar structural

criteria for antioxidant activity can be also observed in compounds 5140311 and 5140321

by taking into consideration that the nitrogen atom at position three of 2-thioxo-1, 3-

thiazolidin-4-one is attached to carboxylic acid moiety that can still donate hydrogen

atom.
96

Figure 3.16(A)- The potency of the inhibitory effect against MAO-B as produced by

compounds 5143886, 5144779, 5108394 and 5108400; the data are displayed as (Mean ±

SEM).
97

Figure 3.16(B)- Percentage of peroxyl radical quenching potential (antioxidant) for

compounds 5143886, 5144779, 5108394 and 5108400, values are presented as (Mean ±

SEM) and Ascorbic acid was used as positive control (100% activity).
98

Figure 3.16(C)- Chemical structural formula for compounds 5108394, 5108400, 5144779

and 5143886.
99

Figure 3.16(D)- Two dimensional representation for the docking of compound 5108394

into human MAO-B crystal, this image was generated by using LigPlot+ v.1.4.5 [153].
100

Figure 3.16(E)- Two dimensional representation for the docking of compound 5108400

into human MAO-B crystal, this image was generated by using LigPlot+ v.1.4.5 [153].
101

Figure 3.17 clearly shows that compounds 7935085 and 5476423 have a very

weak MAO-B inhibition activity. Compound 5476423 appears to be a potent radical

quencher; on the other hand compound 7935085 shows a weaker potency for radical

reduction. This variation in antioxidant power can be attributed in part to the difference in

the substitution type and position within quinoline moiety. Previous researches showed

that attachment of hydroxyl group to position eight in quinoline moiety can greatly

enhance the radical scavenging activity [182].Such structural requirement for antioxidant

potential is clearly evident in compound 5476423 with the presence of two 8-

hydroxyquinoline moieties; the weaker scavenging activity of compound 7935085 can be

partially explained by the presence of methyl group attached to position eight of

quinoline moiety and the attachment of hydroxyl group to position two of quinoline

system.
102

Figure 3.17(A)- The potency of the inhibitory effect against MAO-B as produced by

compounds 7935085 and 5476423; the data are displayed as (Mean ± SEM).
103

Figure 3.17(B)- Percentage of peroxyl radical quenching potential (antioxidant) for

compounds 5476423 and 7935085, values are presented as (Mean ± SEM) and Ascorbic

acid was used as positive control (100% activity).


104

Figure 3.17(C)- Chemical structural formula for both compounds 5476423 and 7935085.
Chapter four: Discussion

Parkinson's disease is a progressive neurodegenerative disorder that mainly

affects elderly people; it holds the second rank after Alzheimer's disease in terms of

neurodegenerative disorders prevalence among this segment of population [161].

Parkinson's disease is characterized by a progressive loss (degeneration) of dopaminergic

neurons in the substantia nigra pars compacta region of the midbrain, this gradual damage

will negatively impact nigrostriatal pathway leading to the emergence of classical motor

related symptoms. As the disease progresses in course, non-motor symptoms (like

depression and dementia) may also appear due to sporadic degeneration within

cholinergic, noradrenergic and serotonergic pathways in the brain [162].

Parkinson's disease is a complex neurologic condition with multiple etiological

factors being involved in guiding the pathophysiological course of this disorder. Current

therapeutic approaches for this disorder focus only on symptomatic mitigation through

dopamine replacement therapy [11-13]. No treatment is currently available that can

hinder or slow down the progression of the neurodegenerative process [19, 20]. The

application of designed multiple ligands (DMLs) approach seems to be the most

convenient tool to produce a molecule capable of targeting more than one pathway within

the pathological process with a possible disease course modifying effect [21-28].

105
106

Monoamine oxidase B (MAO-B) enzyme appears to be a promising target for

such therapeutic approach. MAO-B enzyme catalyzes the oxidative deamination of

dopamine and several other amines [38] and by blocking the effect of this enzyme we can

elevate the level of both endogenous and exogenous dopamine [29, 30]. The inhibition of

MAO-B enzyme can also lower the production of hydrogen peroxide as a byproduct of

oxidative deamination reaction [31]. It is believed that the produced hydrogen peroxide

can undergo Fenton reaction in the presence of ferrous iron to yield hydroxyl radicals

[163]. By lowering the level of hydrogen peroxide, we can minimize the activity of

Fenton reaction and protect neurons from detrimental oxidative stress status.

𝐅𝐞𝟐+ → 𝐇𝟐 𝐎𝟐 → 𝐅𝐞𝟑+ + 𝐇𝐎. + 𝐎𝐇 − (Fenton reaction)

It is important to mention that the expression level of antioxidant enzymes

decreases with age leading to a reduction in the antioxidant capacity of the brain [164,

165]. The cellular iron level inside the brain is elevated with age and with

neurodegenerative conditions [92]. MAO-B but not MAO-A expression level inside the

brain is increased with age [32, 33, 34, 87]. MAO-B is expressed in glial cells; the level

of MAO-B inside the brain is elevated in patients with Parkinson's disease as a direct

result of gliosis. Human basal ganglia shows high level of activity for MAO-B as

compared with MAO-A and since both isozymes catalyze the oxidative deamination

(termination) of dopamine neurotransmitter with equal efficiency then it is very logical

from pharmacological perspective to target MAO-B selectively [36].


107

Based on monoamine oxidase inhibition assay results, it is very obvious that

compounds 5143886 and 5140311 are highly potent and selective in terms of inhibition

effect against MAO-B enzyme. This potency and selectivity in action can be explained

through docking studies results; it appears that these two compounds can exhibit an

extended conformation that traverses both the entrance cavity and substrate cavity of the

active site in MAO-B enzyme. Such conformation will force Ile 199 residue to acquire an

open configuration that lead to the fusion of these two cavities. It is believed that the

configuration of Ile 199 residue plays a critical role in determining substrate and inhibitor

binding specificity for MAO-B [167]. When small molecule (substrate or inhibitor) enters

the active site of MAO-B, Ile 199 residue will obtain a closed conformation and this will

produce the bipartite configuration of the active site. On the other hand, Isoleucine 199

residue will obtain an open conformation when long, planar molecule (extended

conformation) enters the active site and this will result in the fusion of both entrance

cavity and substrate cavity.

Another interesting finding in MAO-B inhibition assay results is that both

compounds 7730595 and 7747412 seem to be potent inhibitors, however docking studies

predict that these compounds don't enter the active site of MAO-B. The images generated

by docking programs reveal that these two compounds occupy a position that is very

close to the gating switch (loop 99-112), it is very clear to notice that these compounds

are involved in hydrophobic interactions with amino acid residues in the gating loop

(mainly Phe 103 and Pro 102). Based on these images, we can build an assumption that

these compounds produce their inhibitory effect through retarding the accessibility of
108

substrates to the active site of MAO-B enzyme. This difference in docking position

between compounds (5140311, 5143886) and compounds (7747412, 7730595) can be

attributed to the difference in structural geometry, the structures of compounds 7747412

and 7730595 are bulkier (four to five rings system) than those for compounds 5140311

and 5143886 (two to three rings system). Although the chemical structure of compound

7919469 is composed of four rings, this compound appears to be capable of entering the

active site of MAO-B enzyme as shown in docking images. According to MAO-B

inhibition assay results, compound 7919469 seems to be a potent inhibitor. We expect

that the four rings in this compound are arranged in a way that can provide a planar and

an extended conformation and this will eventually allow compound 7919469 to access

MAO-B active site and produce a potent inhibitory effect.

A recent literature proposed the presence of a conformational relationship

between Ile 199 residue and Phe 103 residue [166]. When Ile 199 residue exhibits an

open conformation, the side chain of Phe 103 residue will be forced to acquire a

conformation that makes the gating switch (loop) closes the entrance into the active site

of MAO-B. On the other hand, when Ile 199 residue is in closed conformation then the

side chain of Phe 103 will obtain a conformation that favors opening of the gating loop.

In the ligand free condition, these two conformations are rapidly interchangeable and this

will provide accessibility to the active site of MAO-B enzyme.


109

Albumin is the most abundant form of plasma protein in human beings; it acts as

a vehicle for the transport of many endogenous and exogenous compounds through

reversible binding [142, 168]. Exploring the profile of drug binding to serum albumin is

considered an important step in drug development process; such binding can influence

pharmacokinetics and pharmacodynamics parameters of drug under investigation [169-

173]. Binding of drug molecules to plasma albumin can change the volume of

distribution; lower the rate of clearance and increase plasma half-life for that drug [174,

175]. There are two main binding regions in albumin molecule for aromatic and

heterocyclic compounds, these binding regions are hydrophobic in nature and are located

in subdomain IIA (site I) and subdomain IIIA (site II) [158,159]. Drug molecules usually

bind to plasma albumin in a dynamic and reversible style with a very fast dissociation

rate [176].

We have used bovine serum albumin (BSA) as an alternative for human serum

albumin (HSA) for the binding assay because BSA is readily available and because of its

structural homology with HSA [177]. Our results for BSA binding assay clearly show

that most of the tested compounds have a high tendency for BSA binding; this may

negatively affects the distribution of these compounds throughout body tissues and limits

the accessibility to site of action. The only exceptions are compounds 5476423 and

5140311 which show moderate tendency to bind BSA.


110

One of the main challenges in producing a successful neurotherapeutic agent is to

design a molecule capable of passing through the blood brain barrier (BBB) and reaching

the required site of action [178]. Blood brain barrier (BBB) is the main site for molecular

exchange between the brain and blood stream; it functions to maintain a stable

microenvironment for the brain. This barrier is composed mainly from endothelial cells

that line cerebral microvesseles; it represents a combination of both physical and

metabolic barriers. The physical barrier involves the presence of intercellular tight

junctions between the endothelial cells with a very limited vesicular transport. The blood

brain barrier also contains active transporters; some of these transporters transfer

nutrients and other materials from blood to brain (influx) while the other transporters

mediate the clearance of waste products and some drugs from brain to blood (efflux). The

metabolic barrier involves phase I and phase II metabolic enzymes that can limit the

ability of lipophilic compounds to penetrate BBB by converting them into a more

hydrophilic products [179-181]. Most CNS active drugs penetrate BBB through

transcellular passive diffusion process [143].

Parallel artificial membrane permeability assay (PAMPA) is used to predict BBB

permeability potential for compound under investigation. We performed this assay only

for compound 5140311; this compound was the only one that fluoresced at the excitation

wave length range produced by the fluorescence reader. We calculated the value of

effective permeability (Pe) as mentioned previously and then we used the following table

(classification of PAMPA-BBB results based on Pe ranges provided by reference [160])

to interpret our results.


111

Table 4.1-Classification of PAMPA-BBB results.

BBB permeability Pe (𝟏𝟎−𝟔 cm 𝐬 −𝟏 )

High (CNS+) >4.0

Low (CNS-) <2.0

Uncertain (CNS+/-) 4.0-2.0

The calculated Pe value for compound 5140311 was equal to 5.2966× 10−6 cm

s−1 and according to this table, our compound is highly permeable.

Oxidation reaction can produce free radicals; these radicals can start a chain

reaction that involves several steps (initiation, propagation, branching and termination of

these radicals). Such chain of oxidative events can eventually damage cellular contents

like lipid membranes, DNA and proteins. In general, antioxidants can block such reaction

through two major mechanisms: They can either inhibit the formation of free radicals

from their unstable precursors (preventive antioxidants) or they can break the reaction

chain through inhibiting the propagation and branching steps (chain breaking

antioxidants). Most antioxidants are classified as chain breaking agents; they exert their

effect through scavenging free radicals. The chain breaking antioxidants should be able to

act as good hydrogen donors in order to reduce free radicals. Once they donate hydrogen

atom, they should form stable radicals that are unable to continue the chain reaction or

they continue it but with very low efficiency [149, 182].


112

Based on our results for oxygen radical absorbance capacity (ORAC) assay, it is

very clear that all of the tested compounds show high antioxidant capacity that is either

equivalent or higher than that reported for Ascorbic acid (positive control). The only

exception is compound 7954103 which shows minor antioxidant effect.

It is evident from all these virtual and in vitro assays that candidate 5140311

represents a potential lead compound for the development of new antiparkinsonian

agents. This compound is a potent and selective inhibitor for MAO-B, it is highly

permeable through BBB and shows moderate tendency for BSA binding. Finally this

compound shows excellent potential for peroxyl radicals quenching.

As a conclusion for our basic structure activity relationship (SAR) study, it is very

clear that 2-thioxo-1, 3-thiazolidin-4-one ring plays an essential role as a structural

requirement for both MAO-B inhibition activity and radicals quenching potential. For

MAO-B inhibitory effect, we believe that the oxygen atom attached to position four of 2-

thioxo-1, 3-thiazolidine ring can be involved in hydrogen bonding with a certain amino

acid residue or a conserved water molecule within the active site. The nitrogen atom in

position three of the 2-thioxo-1, 3-thiazolidin-4-one ring can be also involved in the

formation of hydrogen bond with a conserved active site water molecule. For antioxidant

potential, we assume that amine group at position three of the 2-thioxo-1, 3-thiazolidin-4-

one ring can act as a good hydrogen donor. The presence of thioxo group at position two

of the 1, 3-thiazolidin-4-one ring appears to be essential for antioxidant activity.


113

8-hydroxyquinoline has been shown to exhibit a strong iron chelating activity and

an antioxidant potential [183, 184]. This molecule has been also shown to be capable of

passing through blood brain barrier [57]. 8-hydroxyquinoline has been successfully used

as a building block for the design and synthesis of several novel bi-functional neuroactive

compounds like HLA20 and M30 [103, 104, 105, 107]. Based on these facts and our

results, it is very clear that 8-hydroxyquinoline represents a promising building block for

iron chelating activity and radical scavenging potential.

By using 2-thioxo-1, 3-thiazolidin-4-one ring and 8-hydroxyquinoline as building

blocks, we were able to design two possible hypothetical scaffolds for MAO-B inhibitors.

We hope that the combination of these two building blocks can help us in producing

potent, selective and centrally acting MAO-B inhibitors with a promising iron chelating

effect and a possible antioxidant potential. Figure 4.1 and figure 4.2 show the chemical

formula for our scaffolds (scaffolds A and B) together with initial results of docking

analysis. Docking results for scaffold A in figure 4.1 confirm the presence of two

hydrogen bonds; the first bond is between the oxygen atom attached to position four of 2-

thioxo-1, 3-thiazolidine ring and the hydroxyl group of phenyl moiety in Tyr 435 (residue

of aromatic cage) while the second hydrogen bond is between the hydroxyl group

attached to position eight of the quinoline moiety and the sulfhydryl group of Cysteine

172 residue. In figure 4.2, docking images for scaffold B confirm the presence of one

hydrogen bond between the oxygen atom attached to position four of 2-thioxo-1, 3-

thiazolidine ring and the sulfhydryl group of Cysteine 172 residue.


114

Scaffold A

Molecular weight =288.34 g/mole

5-[(8-hydroxyquinolin-7-yl) methylidene]-2-sulfanylidene-1,3-thiazolidin-4-one

Figure 4.1(A)- Chemical structure together with IUPAC name for our first prospective

scaffold (scaffold A).


115

Scaffold A

Figure 4.1(B)- Two dimensional representation for the docking of scaffold A into MAO-

B crystal, this image was generated by using LigPlot+ v.1.4.5 [153]. Hydrogen bonds are

represented by green dashed lines.


116

Figure 4.1(C)- Three dimensional representation for the docking of scaffold A into

MAO-B crystal. This image was generated by PyMOL v.1.3 (www.schrodinger.com)

[154]. Our ligand is colored by red and the hydrogen bonds are shown as cyan lines.
117

Scaffold B

Molecular weight =288.34 g/mole

5-[(8-hydroxyquinolin-5-yl) methylidene]-2-sulfanylidene-1,3-thiazolidin-4-one

Figure 4.2(A)- Chemical structure together with IUPAC name for our second prospective

scaffold (scaffold B).


118

Scaffold B

Figure 4.2(B)- Two dimensional representation for the docking of scaffold B into MAO-

B crystal, this image was generated by using LigPlot+ v.1.4.5 [153]. Hydrogen bonds are

represented by green dashed lines.


119

Figure 4.2(C)- Three dimensional representation for the docking of scaffold B into MAO-

B crystal. This image was generated by PyMOL v.1.3 (www.schrodinger.com) [154].

Our ligand is colored by red and the hydrogen bonds are shown as cyan lines.
120

As part of our attempts to refine the overall structure of these scaffolds and to

optimize their expected pharmacokinetic profile, virtual evaluation of oral bioavailability

and CNS permeability was performed for both scaffolds. For this purpose, a number of

physicochemical parameters were initially calculated by using ChemAxon’s calculators

and calculator plugins (www.chemaxon.com). These parameters were then assessed for

their degree of obedience to Lipinski’s rule of five for oral bioavailability [185] and

Norinder’s two rules (AstraZeneca’s rules) for CNS permeability [186]. For simplicity in

presentation, we have listed the standards for the above rules together with our calculated

parameters in tables 4.2 and 4.3.

Based on data in table 4.2, we can clearly notice that both scaffolds A and B will

probably show a good oral activity profile. These scaffolds are in complete agreement

with Lipinski’s rule of five. In table 4.3, on the other hand, these two scaffolds deviate

from the last two standards for CNS permeability (parameters with red color). Such

deviation may cause a future problem in CNS accessibility during lead optimization.

From our point of view, we expect that such probable obstacle can be defeated by

reducing the size of polar mass in these scaffolds. We believe that removal of nitrogen

atom from 8-hydroxyquinoline part can enhance the value of {log P - (N+O)} and this

will eventually increase the value of log BB. Such modification in the structure of these

scaffolds will also lower the value of PSA (Polar surface area) and by this way the

modified scaffolds will obey all CNS permeability rules. Based on this assumption, we

have modified the structure of scaffolds A and B by using hydroxynaphthalene moiety

instead of 8-hydroxyquinoline and this gave us two new scaffolds, namely scaffold C and
121

scaffold D. To avoid redundancy, we have listed the physicochemical parameters for

these modified scaffolds in table 4.2 and 4.3 also. Based on these parameters, we can

easily observe that these modified scaffolds (C and D) will probably have a good oral

absorption profile and a satisfactory CNS permeation capacity.

Docking studies in figure 4.3 show that scaffold C exhibits a very similar

configuration to that of scaffold A within MAO-B crystal. In figure 4.4, MAO-B docking

studies for scaffold D show that the thiazolidine ring occupies a reversed orientation as

compared to that of scaffold B. This reversed direction can be attributed to rotation of the

single bond between 4-hydroxynaphthalene moiety and methylidene group (the only

rotatable bond in our design); this rotation will lead to the loss of hydrogen bond between

the oxygen atom attached to position four of 2-thioxo-1, 3-thiazolidine ring and the

sulfhydryl group of Cysteine 172 residue.

Such proposed structural modification (removal of nitrogen atom from 8-

hydroxyquinoline) may not adversely impact MAO-B inhibition capacity of our design. It

is believed that hydroxynaphthalene moiety can quench radical species through single

electron transfer process. This will convert hydroxynaphthalene molecule into a

stabilized radical form which in turn may undergo dimerization (coupling) reaction [189].

In addition some scientific literatures have shown that phenolic compounds can form

complex with iron [190]. Based on these facts, we hope that both scaffolds C and D can

still meet our goals to produce a potent, selective and centrally acting MAO-B inhibitor

with a possible antioxidant activity and a promising iron chelating ability.


122

Table 4.2- The obedience degree to Lipinski’s rule of five. It is important to mention that

the number of rotatable bonds condition in this table was obtained from Veber’s rules for

oral bioavailability of drug candidates [187].

Physicochemical parameter Standard value Scaffolds A&B Scaffolds C&D

Molecular Weight ≤ 500 g/mol 288.34 g/mol 287.36 g/mol

Log P ≤5 2.59 3.43

H-bond donors (OH + NH) ≤5 2 2

H-bond acceptors (O + N) ≤ 10 2 1

Number of rotatable bonds ≤ 10 1 1


123

Table 4.3- The obedience degree to Norinder’s rules (AstraZeneca’s rules) for CNS

permeability. It is important to notice that the third condition for PSA value in this table

was obtained from reference [188].

Physicochemical parameter Standard value Scaffolds A&B Scaffolds C&D

N + O atoms count ≤5 4 3

Log P – (N + O) value > 0 (positive) -1.41 +0.43

Polar surface area (PSA) < 60 – 70 Ų 62.22 Ų 49.33 Ų


124

Scaffold C

Molecular weight =287.36 g/mole

5-[(1-hydroxynaphthalen-2-yl) methylidene]-2-sulfanylidene-1,3-thiazolidin-4-one

Figure 4.3(A)- Chemical structure together with IUPAC name for the modified scaffold

(scaffold C).
125

Figure 4.3(B)- Two dimensional representation for the docking of scaffold C into MAO-

B crystal, this image was generated by using LigPlot+ v.1.4.5 [153]. Hydrogen bonds are

represented by green dashed lines.


126

Scaffold D

Molecular weight =287.36 g/mole

5-[(4-hydroxynaphthalen-1-yl) methylidene]-2-sulfanylidene-1,3-thiazolidin-4-one

Figure 4.4(A)- Chemical structure together with IUPAC name for the modified scaffold

(scaffold D).
127

Figure 4.4(B)- Two dimensional representation for the docking of scaffold D into MAO-

B crystal, this image was generated by using LigPlot+ v.1.4.5 [153].


128

As a final attempt to defeat the blood brain barrier and to improve the overall

kinetic profile of our design, we found that it is more convenient to target the transporters

system within this barrier. Several previous researches have shown that attachment of L-

alanine residue to certain drugs can enhance the CNS accessibility of these drugs. The

addition of such neutral amino acid residue will make the whole resultant molecule a

good substrate for large neutral amino acid transporter (uptake system L). Such strategy

was employed successfully for the selective delivery of several drugs into the brain and

good examples in our case are L-dopa and the experimental drug M10 [107]. The

attachment of L-alanine residue to scaffold A or scaffold B will make these chemicals

more hydrophilic (elevates polar surface area) and this will lower their passive

permeation into tissues other than the brain. Through such addition, we can enhance the

selective delivery of our chemicals into the brain and minimize the unwanted side effects

of our design. We expect that such modification in our design will not negatively impact

MAO-B inhibition effect, iron chelation capacity or radicals quenching potential. The

structural formula and docking images for the two new scaffolds (L-alanine derivatives)

can be seen in figure 4.5. The application of Lipinski’s rule of five shows that these two

new scaffolds (E and F) may be still able to pass gastrointestinal tract as can be seen in

table 4.4.

In order to expand the scope of our possible opportunities, we are hereby

presenting a new design for another possible scaffold. The new design (scaffold G)

combines three essential building blocks and these are: 2-thioxo-1, 3-thiazolidin-4-one

ring (for MAO-B inhibitory effect and antioxidant capacity), catechol moiety (for iron
129

chelation activity and radical quenching potential) and L-alanine residue (for selective

delivery to the brain). We believe that the core of our research is represented in this new

design (scaffold G). The docking image and virtual pharmacokinetic parameters for this

scaffold can be seen in figure 4.6 and table 4.4.


130

Scaffold E

Molecular weight= 375.42 g/mol

2-amino-3-(8-hydroxy-7-{[4-oxo-2-sulfanylidene-1,3-thiazolidin-5-

ylidene]methyl}quinolin-4-yl)propanoic acid

Figure 4.5(A)- Chemical structure together with IUPAC name for the modified scaffold

(scaffold E).
131

Figure 4.5(B)- Two dimensional representation for the docking of scaffold E into MAO-

B crystal, this image was generated by using LigPlot+ v.1.4.5 [153]. Hydrogen bonds are

represented by green dashed lines.


132

Scaffold F

Molecular weight= 375.42 g/mol

2-amino-3-(8-hydroxy-5-{[4-oxo-2-sulfanylidene-1,3-thiazolidin-5-

ylidene]methyl}quinolin-2-yl)propanoic acid

Figure 4.5(C)- Chemical structure together with IUPAC name for the modified scaffold

(scaffold F).
133

Figure 4.5(D)- Two dimensional representation for the docking of scaffold F into MAO-

B crystal, this image was generated by using LigPlot+ v.1.4.5 [153]. Hydrogen bonds are

represented by green dashed lines.


134

Scaffold G

Molecular weight= 340.37 g/mol

2-amino-3-(2,3-dihydroxy-4-{[4-oxo-2-sulfanylidene-1,3-thiazolidin-5-

ylidene]methyl}phenyl)propanoic acid

Figure 4.6(A)- Chemical structure together with IUPAC name for our novel design

(scaffold G).
135

Figure 4.6(B)- Two dimensional representation for the docking of scaffold G into MAO-

B crystal, this image was generated by using LigPlot+ v.1.4.5 [153]. Hydrogen bonds are

represented by green dashed lines.


136

Table 4.4- The application of Lipinski’s rule of five to scaffolds E, F and G. The last two

conditions (number of rotatable bonds and polar surface area) were obtained from

Veber’s rules for oral bioavailability of drug candidates [187].

Physicochemical parameter Standard value Scaffolds E&F Scaffold G

Molecular Weight ≤ 500 g/mol 375.42 g/mol 340.37 g/mol

Log P ≤5 1.7 1.24

H-bond donors (OH + NH) ≤5 4 5

H-bond acceptors (O + N) ≤ 10 3 2

Number of rotatable bonds ≤ 10 4 4

Polar surface area (PSA) ≤ 140 Ų 125.54 Ų 132.88 Ų


137

Our preliminary data indicate that compounds with a close structural formula to

that of both scaffolds A and B are effective in MAO-B inhibition assay; some of these

compounds were able to display an IC50 value within micro Molar range. One of these

compounds, the one that closely resembles scaffold A, was superior in MAO-B inhibition

to Zonisamide. It is important to mention that Zonisamide is a well-known anticonvulsant

agent with antiparkinsonian effect (reversible MAO-B inhibitor); it was used as a positive

control in our MAO-B inhibition assay. Based on these recent data, we are currently

trying to synthesize both scaffolds A and B in our lab. Characterization of these two

scaffolds and refining their structural formula appears to be a promising future plan to

produce and optimize an antiparkinsonian lead compound. The newly proposed structural

modification (generation of scaffolds C, D, E and F) may also expand the scope of our

future plans, goals and opportunities. We believe that scaffold G is a novel design for a

multifunctional antiparkinsonian agent and we hope that such design can see the light in

the nearby future.


References

1. Bove J, Prou D, Perier C, Przedborski S. Toxin-induced models of Parkinson's

disease. NeuroRx 2005;2(3):484-94.

2. Aschner M. Manganese: brain transport and emerging research needs. Environ

Health Perspect 2000;108 Suppl 3:429-32.

3. Shastry BS. Parkinson disease: etiology, pathogenesis and future of gene therapy.

Neurosci Res 2001;41(1):5-12.

4. Sian-Hulsmann J, Mandel S, Youdim MB, Riederer P. The relevance of iron in

the pathogenesis of Parkinson's disease. J Neurochem 2011;118(6):939-57.

5. Lang AE, Lozano AM. Parkinson's disease. First of two parts. N Engl J Med

1998;339(15):1044-53.

6. Coelho M, Ferreira JJ. Late-stage Parkinson disease. Nat Rev Neurol

2012;8(8):435-42.

7. Forno LS. Neuropathology of Parkinson's disease. J Neuropathol Exp Neurol

1996;55(3):259-72.

8. Hertzman C, Wiens M, Bowering D, Snow B, Calne D. Parkinson's disease: a

case-control study of occupational and environmental risk factors. Am J Ind Med

1990;17(3):349-55.

138
139

9. Liou HH, Tsai MC, Chen CJ, Jeng JS, Chang YC, Chen SY, Chen RC.

Environmental risk factors and Parkinson's disease: a case-control study in

Taiwan. Neurology 1997;48(6):1583-8.

10. Abeliovich A, Flint Beal M. Parkinsonism genes: culprits and clues. J Neurochem

2006;99(4):1062-72.

11. Schapira A, Bate G, Kirkpatrick P. Rasagiline. Nat Rev Drug Discov

2005;4(8):625-626.

12. Allain H, Bentue-Ferrer D, Akwa Y. Disease-modifying drugs and Parkinson's

disease. Prog Neurobiol 2008;84(1):25-39.

13. Lew M. Overview of Parkinson's disease. Pharmacotherapy 2007;27(12 Pt

2):155S-160S.

14. Voss T, Ravina B. Neuroprotection in Parkinson's disease: myth or reality? Curr

Neurol Neurosci Rep 2008;8(4):304-9.

15. Chen JJ, Swope DM. Pharmacotherapy for Parkinson's disease. Pharmacotherapy

2007;27(12 Pt 2):161S-173S.

16. Jankovic J, Stacy M. Medical management of levodopa-associated motor

complications in patients with Parkinson's disease. CNS Drugs

2007;21(8):677-92.

17. Rezak M. Current pharmacotherapeutic treatment options in Parkinson's disease.

Dis Mon 2007;53(4):214-22.

18. Lees A. Alternatives to levodopa in the initial treatment of early Parkinson's

disease. Drugs Aging 2005;22(9):731-40.


140

19. Mandel S, Grunblatt E, Riederer P, Gerlach M, Levites Y, Youdim MB.

Neuroprotective strategies in Parkinson's disease : an update on progress. CNS

Drugs 2003;17(10):729-62.

20. Stocchi F, Olanow CW. Neuroprotection in Parkinson's disease: clinical trials.

Ann Neurol 2003;53 Suppl 3:S87-97; discussion S97-9.

21. Zimmermann GR, Lehar J, Keith CT. Multi-target therapeutics: when the whole is

greater than the sum of the parts. Drug Discov Today 2007;12(1-2):34-42.

22. Morphy R, Kay C, Rankovic Z. From magic bullets to designed multiple ligands.

Drug Discov Today 2004;9(15):641-51.

23. Morphy R, Rankovic Z. Designed multiple ligands. An emerging drug discovery

paradigm. J Med Chem 2005;48(21):6523-43.

24. Gal S, Zheng H, Fridkin M, Youdim MB. Novel multifunctional neuroprotective

iron chelator-monoamine oxidase inhibitor drugs for neurodegenerative diseases.

In vivo selective brain monoamine oxidase inhibition and prevention of

MPTP-induced striatal dopamine depletion. J Neurochem 2005;95(1):79-88.

25. Van der Schyf CJ, Gal S, Geldenhuys WJ, Youdim MB. Multifunctional

neuroprotective drugs targeting monoamine oxidase inhibition, iron chelation,

adenosine receptors, and cholinergic and glutamatergic action for

neurodegenerative diseases. Expert Opin Investig Drugs 2006;15(8):873-86.

26. Van der Schyf CJ, Geldenhuys WJ, Youdim MB. Multifunctional neuroprotective

drugs for the treatment of cognitive and movement impaired disorders, including

Alzheimer’s and Parkinson’s diseases. Drugs Future 2006;31:447−60.


141

27. Youdim MB, Buccafusco JJ. CNS Targets for multi-functional drugs in the

treatment of Alzheimer's and Parkinson's diseases. J Neural Transm

2005;112(4):519-37.

28. Youdim MB, Buccafusco JJ. Multi-functional drugs for various CNS targets in

the treatment of neurodegenerative disorders. Trends Pharmacol Sci

2005;26(1):27-35.

29. Di Monte DA, DeLanney LE, Irwin I, Royland JE, Chan P, Jakowec MW,

Langston JW. Monoamine oxidase-dependent metabolism of dopamine in the

striatum and substantia nigra of L-DOPA-treated monkeys. Brain Res

1996;738(1):53-9.

30. Finberg JPM, Wang J, Bankiewicz K, Harvey-White J, Kopin IJ, Goldstein DS.

Increased striatal dopamine production from L-DOPA following selective

inhibition of monoamine oxidase B by R(+)-N-propargyl-1-aminoindan

(rasagiline) in the monkey. In: Finberg JPM, Youdim MBH, Riederer P, Tipton

KF, editors. MAO — The Mother of all Amine Oxidases. Volume 52, Journal of

Neural Transmission. Supplement: Springer Vienna; 1998. p 279-285.

31. Youdim MB, Bakhle YS. Monoamine oxidase: isoforms and inhibitors in

Parkinson's disease and depressive illness. Br J Pharmacol 2006;147 Suppl

1:S287-96.

32. Nicotra A, Pierucci F, Parvez H, Senatori O. Monoamine oxidase expression

during development and aging. Neurotoxicology 2004;25(1-2):155-65.

33. Fowler JS, Volkow ND, Wang GJ, Logan J, Pappas N, Shea C, MacGregor R.
142

Age-related increases in brain monoamine oxidase B in living healthy human

subjects. Neurobiol Aging 1997;18(4):431-5.

34. Karolewicz B, Klimek V, Zhu H, Szebeni K, Nail E, Stockmeier CA, Johnson L,

Ordway GA. Effects of depression, cigarette smoking, and age on monoamine

oxidase B in amygdaloid nuclei. Brain Res 2005;1043(1-2):57-64.

35. Weyler W, Hsu YP, Breakefield XO. Biochemistry and genetics of monoamine

oxidase. Pharmacol Ther 1990;47(3):391-417.

36. Youdim MBH, Edmondson D, Tipton KF. The therapeutic potential of

monoamine oxidase inhibitors. Nat Rev Neurosci 2006;7(4):295-309.

37. Bach AW, Lan NC, Johnson DL, Abell CW, Bembenek ME, Kwan SW, Seeburg

PH, Shih JC. cDNA cloning of human liver monoamine oxidase A and B:

molecular basis of differences in enzymatic properties. Proc Natl Acad Sci U S A

1988;85(13):4934-8.

38. Youdim MBH, Finberg JPM, Tipton KF. In: Trendelenburg U, Weiner N, editors.

Catecholamine II. Handbook of Experimental Pharmacology. Berlin: Springer;

1988. p 127–199

39. Tsang D, Ho KP, Wen HL. Ontogenesis of multiple forms of monoamine oxidase

in rat brain regions and liver. Dev Neurosci 1986;8(4):243-50.

40. Strolin Benedetti M, Dostert P, Tipton KF. Developmental aspects of the

monoamine-degrading enzyme monoamine oxidase. Dev Pharmacol Ther

1992;18(3-4):191-200.

41. O'Carroll AM, Fowler CJ, Phillips JP, Tobbia I, Tipton KF. The deamination of
143

dopamine by human brain monoamine oxidase. Specificity for the two enzyme

forms in seven brain regions. Naunyn Schmiedebergs Arch Pharmacol

1983;322(3):198-202.

42. Fowler JS, MacGregor RR, Wolf AP, Arnett CD, Dewey SL, Schlyer D,

Christman D, Logan J, Smith M, Sachs H and others. Mapping human brain

monoamine oxidase A and B with 11C-labeled suicide inactivators and PET.

Science 1987;235(4787):481-5.

43. Westlund KN, Denney RM, Kochersperger LM, Rose RM, Abell CW. Distinct

monoamine oxidase A and B populations in primate brain. Science

1985;230(4722):181-3.

44. O'Carroll AM, Tipton KF, Sullivan JP, Fowler CJ, Ross SB. Intra- and

extrasynaptosomal deamination of dopamine and noradrenaline by the two forms

of human brain monoamine oxidase. Implications for the neurotoxicity of

N-methyl-4-phenyl-1,2,3,6- tetrahydropyridine in man. Biogenic Amines

1987;4:165–178.

45. Shih JC, Chen K, Ridd MJ. Monoamine oxidase: from genes to behavior. Annu

Rev Neurosci 1999;22:197-217.

46. Kearney EB, Salach JI, Walker WH, Seng RL, Kenney W, Zeszotek E, Singer TP.

The covalently-bound flavin of hepatic monoamine oxidase. 1. Isolation and

sequence of a flavin peptide and evidence for binding at the 8alpha position. Eur J

Biochem 1971;24(2):321-7.

47. Binda C, Newton-Vinson P, Hubalek F, Edmondson DE, Mattevi A. Structure of


144

human monoamine oxidase B, a drug target for the treatment of neurological

disorders. Nat Struct Biol 2002;9(1):22-6.

48. Li M, Hubalek F, Newton-Vinson P, Edmondson DE. High-level expression of

human liver monoamine oxidase A in Pichia pastoris: comparison with the

enzyme expressed in Saccharomyces cerevisiae. Protein Expr Purif

2002;24(1):152-62.

49. Newton-Vinson P, Hubalek F, Edmondson DE. High-level expression of human

liver monoamine oxidase B in Pichia pastoris. Protein Expr Purif

2000;20(2):334-45.

50. Rebrin I, Geha RM, Chen K, Shih JC. Effects of carboxyl-terminal truncations on

the activity and solubility of human monoamine oxidase B. J Biol Chem

2001;276(31):29499-506.

51. Mitoma J, Ito A. Mitochondrial targeting signal of rat liver monoamine oxidase B

is located at its carboxy terminus. J Biochem 1992;111(1):20-4.

52. Edmondson DE, Mattevi A, Binda C, Li M, Hubalek F. Structure and mechanism

of monoamine oxidase. Curr Med Chem 2004;11(15):1983-93.

53. Binda C, Li M, Hubalek F, Restelli N, Edmondson DE, Mattevi A. Insights into

the mode of inhibition of human mitochondrial monoamine oxidase B from

high-resolution crystal structures. Proc Natl Acad Sci U S A

2003;100(17):9750-5.

54. Binda C, Mattevi A, Edmondson DE. Structural properties of human monoamine

oxidases A and B. Int Rev Neurobiol 2011;100:1-11.


145

55. Edmondson DE, Binda C, Mattevi A. The FAD binding sites of human

monoamine oxidases A and B. Neurotoxicology 2004;25(1-2):63-72.

56. Kirksey TJ, Kwan SW, Abell CW. Arginine-42 and threonine-45 are required for

FAD incorporation and catalytic activity in human monoamine oxidase B.

Biochemistry 1998;37(35):12360-6.

57. Frederickson CJ, Kasarskis EJ, Ringo D, Frederickson RE. A quinoline

fluorescence method for visualizing and assaying the histochemically reactive

zinc (bouton zinc) in the brain. J Neurosci Methods 1987;20(2):91-103.

58. Binda C, Mattevi A, Edmondson DE. Structure-function relationships in

flavoenzyme-dependent amine oxidations: a comparison of polyamine oxidase

and monoamine oxidase. J Biol Chem 2002;277(27):23973-6.

59. Miller JR, Edmondson DE. Influence of flavin analogue structure on the catalytic

activities and flavinylation reactions of recombinant human liver monoamine

oxidases A and B. J Biol Chem 1999;274(33):23515-25.

60. Miller JR, Guan N, Hubalek F, Edmondson DE. The FAD binding sites of human

liver monoamine oxidases A and B: investigation of the role of flavin ribityl side

chain hydroxyl groups in the covalent flavinylation reaction and catalytic

activities. Biochim Biophys Acta 2000;1476(1):27-32.

61. Frost JW, Rastetter WH. Biomimetric 8a-functionalization of riboflavin. J Am

Chem Soc 1980;102:7157–9.

62. Wu HF, Chen K, Shih JC. Site-directed mutagenesis of monoamine oxidase A

and B: role of cysteines. Mol Pharmacol 1993;43(6):888-93.


146

63. Nandigama RK, Edmondson DE. Influence of FAD structure on its binding and

activity with the C406A mutant of recombinant human liver monoamine oxidase

A. J Biol Chem 2000;275(27):20527-32.

64. De Colibus L, Li M, Binda C, Lustig A, Edmondson DE, Mattevi A.

Three-dimensional structure of human monoamine oxidase A (MAO A): relation

to the structures of rat MAO A and human MAO B. Proc Natl Acad Sci U S A

2005;102(36):12684-9.

65. Andres AM, Soldevila M, Navarro A, Kidd KK, Oliva B, Bertranpetit J. Positive

selection in MAOA gene is human exclusive: determination of the putative amino

acid change selected in the human lineage. Hum Genet 2004;115(5):377-86.

66. Wang J, Edmondson DE. Do monomeric vs dimeric forms of MAO-A make a

difference? A direct comparison of the catalytic properties of rat and human

MAO-A’s. J Neural Transm 2007;114(6):721-724.

67. Edmondson DE, DeColibus L, Binda C, Li M, Mattevi A. New insights into the

structures and functions of human monoamine oxidases A and B. J Neural

Transm 2007;114(6):703-5.

68. Edmondson DE, Binda C, Mattevi A. Structural insights into the mechanism of

amine oxidation by monoamine oxidases A and B. Arch Biochem Biophys

2007;464(2):269-76.

69. Benkovic SJ, Hammes-Schiffer S. A Perspective on Enzyme Catalysis. Science

2003;301(5637):1196-1202.

70. Ma J, Yoshimura M, Yamashita E, Nakagawa A, Ito A, Tsukihara T. Structure of


147

rat monoamine oxidase A and its specific recognitions for substrates and

inhibitors. J Mol Biol 2004;338(1):103-14.

71. Upadhyay AK, Borbat PP, Wang J, Freed JH, Edmondson DE. Determination of

the oligomeric states of human and rat monoamine oxidases in the outer

mitochondrial membrane and octyl beta-D-glucopyranoside micelles using pulsed

dipolar electron spin resonance spectroscopy. Biochemistry 2008;47(6):1554-66.

72. Binda C, Hubalek F, Li M, Herzig Y, Sterling J, Edmondson DE, Mattevi A.

Crystal structures of monoamine oxidase B in complex with four inhibitors of the

N-propargylaminoindan class. J Med Chem 2004;47(7):1767-74.

73. Husain M, Edmondson DE, Singer TP. Kinetic studies on the catalytic mechanism

of liver monoamine oxidase. Biochemistry 1982;21(3):595-600.

74. Pearce LB, Roth JA. Human brain monoamine oxidase type B: mechanism of

deamination as probed by steady-state methods. Biochemistry 1985;24(8):1821-6.

75. Edmondson DE, Bhattacharyya AK, Walker MC. Spectral and kinetic studies of

imine product formation in the oxidation of p-(N,N-dimethylamino)benzylamine

analogues by monoamine oxidase B. Biochemistry 1993;32(19):5196-202.

76. Ramsay RR. Kinetic mechanism of monoamine oxidase A. Biochemistry

1991;30(18):4624-9.

77. Miller JR, Edmondson DE. Structure-activity relationships in the oxidation of

para-substituted benzylamine analogues by recombinant human liver monoamine

oxidase A. Biochemistry 1999;38(41):13670-83.

78. Walker MC, Edmondson DE. Structure-activity relationships in the oxidation of


148

benzylamine analogues by bovine liver mitochondrial monoamine oxidase B.

Biochemistry 1994;33(23):7088-98.

79. Yu PH, Bailey BA, Durden DA, Boulton AA. Stereospecific deuterium

substitution at the alpha-carbon position of dopamine and its effect on oxidative

deamination catalyzed by MAO-A and MAO-B from different tissues. Biochem

Pharmacol 1986;35(6):1027-36.

80. Belleau B, Moran J. Deuterium isotope effects in relation to the chemical

mechanism of monoamine oxidase. Ann N Y Acad Sci 1963;107:822-39.

81. Ruterjans H, Fleischmann G, Knauf M, Lohr F, Blumel M, Lederer F, Mayhew

SG, Muller F. NMR studies of flavoproteins. Biochem Soc Trans

1996;24(1):116-21.

82. Jonsson T, Edmondson DE, Klinman JP. Hydrogen tunneling in the flavoenzyme

monoamine oxidase B. Biochemistry 1994;33(49):14871-8.

83. Silverman RB. Radical Ideas about Monoamine Oxidase. Accounts of Chemical

Research 1995;28(8):335-342.

84. Silverman RB. Electron Transfer Chemistry of Monoamine Oxidase. In: Mariano

PS, editor. Advances in Electron Transfer Chemistry. Volume 2: JAI Press; 1992.

p 177-213.

85. Miller JR, Edmondson DE, Grissom CB. Mechanistic Probes of Monoamine

Oxidase B Catalysis: Rapid-Scan Stopped Flow and Magnetic Field Independence

of the Reductive Half-Reaction. J Am Chem Soc 1995;117:7830-7831.

86. McEwen CM, Jr., Sasaki G, Jones DC. Human liver mitochondrial monoamine
149

oxidase. II. Determinants of substrate and inhibitor specificities. Biochemistry

1969;8(10):3952-62.

87. Fowler CJ, Wiberg A, Oreland L, Marcusson J, Winblad B. The effect of age on

the activity and molecular properties of human brain monoamine oxidase. J

Neural Transm 1980;49(1-2):1-20.

88. Li M, Binda C, Mattevi A, Edmondson DE. Functional role of the "aromatic

cage" in human monoamine oxidase B: structures and catalytic properties of

Tyr435 mutant proteins. Biochemistry 2006;45(15):4775-84.

89. Trickey P, Basran J, Lian LY, Chen Z, Barton JD, Sutcliffe MJ, Scrutton NS,

Mathews FS. Structural and biochemical characterization of recombinant wild

type and a C30A mutant of trimethylamine dehydrogenase from methylophilus

methylotrophus (sp. W(3)A(1)). Biochemistry 2000;39(26):7678-88.

90. Binda C, Coda A, Angelini R, Federico R, Ascenzi P, Mattevi A. A

30-angstrom-long U-shaped catalytic tunnel in the crystal structure of polyamine

oxidase. Structure 1999;7(3):265-76.

91. Erdem SS, Karahan O, Yildiz I, Yelekci K. A computational study on the

amine-oxidation mechanism of monoamine oxidase: insight into the polar

nucleophilic mechanism. Org Biomol Chem 2006;4(4):646-58.

92. Geldenhuys WJ, Youdim MB, Carroll RT, Van der Schyf CJ. The emergence of

designed multiple ligands for neurodegenerative disorders. Prog Neurobiol

2011;94(4):347-59.

93. Youdim MB, Bar Am O, Yogev-Falach M, Weinreb O, Maruyama W, Naoi M,


150

Amit T. Rasagiline: neurodegeneration, neuroprotection, and mitochondrial

permeability transition. J Neurosci Res 2005;79(1-2):172-9.

94. Olanow CW, Rascol O, Hauser R, Feigin PD, Jankovic J, Lang A, Langston W,

Melamed E, Poewe W, Stocchi F and others. A double-blind, delayed-start trial of

rasagiline in Parkinson's disease. N Engl J Med 2009;361(13):1268-78.

95. Youdim MB, Fridkin M, Zheng H. Bifunctional drug derivatives of MAO-B

inhibitor rasagiline and iron chelator VK-28 as a more effective approach to

treatment of brain ageing and ageing neurodegenerative diseases. Mech Ageing

Dev 2005;126(2):317-26.

96. Zheng H, Gal S, Weiner LM, Bar-Am O, Warshawsky A, Fridkin M, Youdim

MB. Novel multifunctional neuroprotective iron chelator-monoamine oxidase

inhibitor drugs for neurodegenerative diseases: in vitro studies on antioxidant

activity, prevention of lipid peroxide formation and monoamine oxidase

inhibition. J Neurochem 2005;95(1):68-78.

97. Weinreb O, Bar-Am O, Amit T, Chillag-Talmor O, Youdim MB. Neuroprotection

via pro-survival protein kinase C isoforms associated with Bcl-2 family members.

FASEB J 2004;18(12):1471-3.

98. Bar-Am O, Weinreb O, Amit T, Youdim MB. Regulation of Bcl-2 family

proteins, neurotrophic factors, and APP processing in the neurorescue activity of

propargylamine. FASEB J 2005;19(13):1899-901.

99. Bar-Am O, Amit T, Youdim MB. Aminoindan and hydroxyaminoindan,

metabolites of rasagiline and ladostigil, respectively, exert neuroprotective


151

properties in vitro. J Neurochem 2007;103(2):500-8.

100. Maruyama W, Nitta A, Shamoto-Nagai M, Hirata Y, Akao Y, Yodim M,

Furukawa S, Nabeshima T, Naoi M. N-Propargyl-1 (R)-aminoindan, rasagiline,

increases glial cell line-derived neurotrophic factor (GDNF) in neuroblastoma

SH-SY5Y cells through activation of NF-kappaB transcription factor. Neurochem

Int 2004;44(6):393-400.

101. Weinreb O, Amit T, Bar-Am O, Yogev-Falach M, Youdim MB. The

neuroprotective mechanism of action of the multimodal drug ladostigil. Front

Biosci 2008;13:5131-7.

102. Yogev-Falach M, Amit T, Bar-Am O, Weinstock M, Youdim MB. Involvement

of MAP kinase in the regulation of amyloid precursor protein processing by novel

cholinesterase inhibitors derived from rasagiline. FASEB J 2002;16(12):1674-6.

103. Zheng H, Weiner LM, Bar-Am O, Epsztejn S, Cabantchik ZI, Warshawsky A,

Youdim MB, Fridkin M. Design, synthesis, and evaluation of novel bifunctional

iron-chelators as potential agents for neuroprotection in Alzheimer's, Parkinson's,

and other neurodegenerative diseases. Bioorg Med Chem 2005;13(3):773-83.

104. Shachar DB, Kahana N, Kampel V, Warshawsky A, Youdim MB.

Neuroprotection by a novel brain permeable iron chelator, VK-28, against

6-hydroxydopamine lession in rats. Neuropharmacology 2004;46(2):254-63.

105. Mandel S, Weinreb O, Amit T, Youdim MB. Mechanism of neuroprotective

action of the anti-Parkinson drug rasagiline and its derivatives. Brain Res Brain

Res Rev 2005;48(2):379-87.


152

106. Zecca L, Youdim MB, Riederer P, Connor JR, Crichton RR. Iron, brain ageing

and neurodegenerative disorders. Nat Rev Neurosci 2004;5(11):863-73.

107. Zheng H, Youdim MB, Weiner LM, Fridkin M. Novel potential neuroprotective

agents with both iron chelating and amino acid-based derivatives targeting central

nervous system neurons. Biochem Pharmacol 2005;70(11):1642-52.

108. Weinstock M, Poltyrev T, Bejar C, Youdim MB. Effect of TV3326, a novel

monoamine-oxidase cholinesterase inhibitor, in rat models of anxiety and

depression. Psychopharmacology (Berl) 2002;160(3):318-24.

109. Petzer JP, Castagnoli N, Jr., Schwarzschild MA, Chen JF, Van der Schyf CJ.

Dual-target-directed drugs that block monoamine oxidase B and adenosine A(2A)

receptors for Parkinson's disease. Neurotherapeutics 2009;6(1):141-51.

110. Jarvis MF, Williams M. Direct autoradiographic localization of adenosine A2

receptors in the rat brain using the A2-selective agonist, [3H]CGS 21680. Eur J

Pharmacol 1989;168(2):243-6.

111. Schiffmann SN, Jacobs O, Vanderhaeghen JJ. Striatal restricted adenosine A2

receptor (RDC8) is expressed by enkephalin but not by substance P neurons: an in

situ hybridization histochemistry study. J Neurochem 1991;57(3):1062-7.

112. Fink JS, Weaver DR, Rivkees SA, Peterfreund RA, Pollack AE, Adler EM,

Reppert SM. Molecular cloning of the rat A2 adenosine receptor: selective

co-expression with D2 dopamine receptors in rat striatum. Brain Res Mol Brain

Res 1992;14(3):186-95.

113. Ferre S, von Euler G, Johansson B, Fredholm BB, Fuxe K. Stimulation of


153

high-affinity adenosine A2 receptors decreases the affinity of dopamine D2

receptors in rat striatal membranes. Proc Natl Acad Sci U S A

1991;88(16):7238-41.

114. Ferre S, O'Connor WT, Fuxe K, Ungerstedt U. The striopallidal neuron: a main

locus for adenosine-dopamine interactions in the brain. J Neurosci

1993;13(12):5402-6.

115. Xu K, Bastia E, Schwarzschild M. Therapeutic potential of adenosine A2A

receptor antagonists in Parkinson's disease. Pharmacol Ther 2005;105(3):267-310.

116. Pinna A, Wardas J, Simola N, Morelli M. New therapies for the treatment of

Parkinson's disease: adenosine A2A receptor antagonists. Life Sci

2005;77(26):3259-67.

117. Ferre S, Fredholm BB, Morelli M, Popoli P, Fuxe K. Adenosine-dopamine

receptor-receptor interactions as an integrative mechanism in the basal ganglia.

Trends Neurosci 1997;20(10):482-7.

118. Kanda T, Jackson MJ, Smith LA, Pearce RK, Nakamura J, Kase H, Kuwana Y,

Jenner P. Adenosine A2A antagonist: a novel antiparkinsonian agent that does not

provoke dyskinesia in parkinsonian monkeys. Ann Neurol 1998;43(4):507-13.

119. Chen JF, Moratalla R, Impagnatiello F, Grandy DK, Cuellar B, Rubinstein M,

Beilstein MA, Hackett E, Fink JS, Low MJ and others. The role of the D(2)

dopamine receptor (D(2)R) in A(2A) adenosine receptor (A(2A)R)-mediated

behavioral and cellular responses as revealed by A(2A) and D(2) receptor

knockout mice. Proc Natl Acad Sci U S A 2001;98(4):1970-5.


154

120. Schwarzschild MA, Agnati L, Fuxe K, Chen JF, Morelli M. Targeting adenosine

A2A receptors in Parkinson's disease. Trends Neurosci 2006;29(11):647-54.

121. Ikeda K, Kurokawa M, Aoyama S, Kuwana Y. Neuroprotection by adenosine

A2A receptor blockade in experimental models of Parkinson's disease. J

Neurochem 2002;80(2):262-70.

122. Chen JF, Xu K, Petzer JP, Staal R, Xu YH, Beilstein M, Sonsalla PK, Castagnoli

K, Castagnoli N, Jr., Schwarzschild MA. Neuroprotection by caffeine and A(2A)

adenosine receptor inactivation in a model of Parkinson's disease. J Neurosci

2001;21(10):RC143.

123. Ascherio A, Zhang SM, Hernan MA, Kawachi I, Colditz GA, Speizer FE, Willett

WC. Prospective study of caffeine consumption and risk of Parkinson's disease in

men and women. Ann Neurol 2001;50(1):56-63.

124. Ross GW, Abbott RD, Petrovitch H, Morens DM, Grandinetti A, Tung KH,

Tanner CM, Masaki KH, Blanchette PL, Curb JD and others. Association of

coffee and caffeine intake with the risk of Parkinson disease. JAMA

2000;283(20):2674-9.

125. Powers KM, Kay DM, Factor SA, Zabetian CP, Higgins DS, Samii A, Nutt JG,

Griffith A, Leis B, Roberts JW and others. Combined effects of smoking, coffee,

and NSAIDs on Parkinson's disease risk. Mov Disord 2008;23(1):88-95.

126. Nathan DM. Rosiglitazone and cardiotoxicity--weighing the evidence. N Engl J

Med 2007;357(1):64-6.

127. Nissen SE, Wolski K. Effect of rosiglitazone on the risk of myocardial infarction
155

and death from cardiovascular causes. N Engl J Med 2007;356(24):2457-71.

128. Binda C, Aldeco M, Geldenhuys WJ, Tortorici M, Mattevi A, Edmondson DE.

Molecular Insights into Human Monoamine Oxidase B Inhibition by the

Glitazone Anti-Diabetes Drugs. ACS Med Chem Lett 2011;3(1):39-42.

129. Culman J, Zhao Y, Gohlke P, Herdegen T. PPAR-gamma: therapeutic target for

ischemic stroke. Trends Pharmacol Sci 2007;28(5):244-9.

130. Shimazu T, Inoue I, Araki N, Asano Y, Sawada M, Furuya D, Nagoya H,

Greenberg JH. A peroxisome proliferator-activated receptor-gamma agonist

reduces infarct size in transient but not in permanent ischemia. Stroke

2005;36(2):353-9.

131. Luna-Medina R, Cortes-Canteli M, Alonso M, Santos A, Martinez A,

Perez-Castillo A. Regulation of inflammatory response in neural cells in vitro by

thiadiazolidinones derivatives through peroxisome proliferator-activated receptor

gamma activation. J Biol Chem 2005;280(22):21453-62.

132. Combs CK, Johnson DE, Karlo JC, Cannady SB, Landreth GE. Inflammatory

mechanisms in Alzheimer's disease: inhibition of beta-amyloid-stimulated

proinflammatory responses and neurotoxicity by PPARgamma agonists. J

Neurosci 2000;20(2):558-67.

133. Quinn LP, Crook B, Hows ME, Vidgeon-Hart M, Chapman H, Upton N,

Medhurst AD, Virley DJ. The PPARgamma agonist pioglitazone is effective in

the MPTP mouse model of Parkinson's disease through inhibition of monoamine

oxidase B. Br J Pharmacol 2008;154(1):226-33.


156

134. Binda C, Milczek EM, Bonivento D, Wang J, Mattevi A, Edmondson DE. Lights

and shadows on monoamine oxidase inhibition in neuroprotective

pharmacological therapies. Curr Top Med Chem 2011;11(22):2788-96.

135. Grant JA, Gallardo MA, Pickup BT. A fast method of molecular shape

comparison: A simple application of a Gaussian description of molecular shape. J

Comput Chem 1996;17(14):1653-1666.

136. Krajl M. A rapid microfluorimetric determination of monoamine oxidase.

Biochem Pharmacol 1965;14(11):1684-6.

137. Yan Z, Caldwell GW, Zhao B, Reitz AB. A high-throughput monoamine oxidase

inhibition assay using liquid chromatography with tandem mass spectrometry.

Rapid Commun Mass Spectrom 2004;18(8):834-40.

138. Tayeh N, Rungassamy T, Albani JR. Fluorescence spectral resolution of

tryptophan residues in bovine and human serum albumins. J Pharm Biomed Anal

2009;50(2):107-16.

139. Lakowicz JR. Principles of Fluorescence Spectroscopy. New York: Springer;

2006.

140. Sułkowska A. Interaction of drugs with bovine and human serum albumin.

Journal of Molecular Structure 2002;614(1–3):227-232.

141. Freifelder D. Physical Biochemistry: Applications to Biochemistry and Molecular

Biology. San Francisco: W.H.Freeman & Co Ltd; 1976.

142. Bi S, Sun Y, Qiao C, Zhang H, Liu C. Binding of several anti-tumor drugs to

bovine serum albumin: Fluorescence study. Journal of Luminescence


157

2009;129(5):541-547.

143. Di L, Kerns EH, Fan K, McConnell OJ, Carter GT. High throughput artificial

membrane permeability assay for blood-brain barrier. Eur J Med Chem

2003;38(3):223-32.

144. Kansy M, Senner F, Gubernator K. Physicochemical high throughput screening:

parallel artificial membrane permeation assay in the description of passive

absorption processes. J Med Chem 1998;41(7):1007-10.

145. Sugano K, Nabuchi Y, Machida M, Aso Y. Prediction of human intestinal

permeability using artificial membrane permeability. Int J Pharm

2003;257(1-2):245-51.

146. Sinko B, Garrigues TM, Balogh GT, Nagy ZK, Tsinman O, Avdeef A,

Takacs-Novak K. Skin-PAMPA: a new method for fast prediction of skin

penetration. Eur J Pharm Sci 2012;45(5):698-707.

147. Cao G, Alessio HM, Cutler RG. Oxygen-radical absorbance capacity assay for

antioxidants. Free Radic Biol Med 1993;14(3):303-11.

148. Cao G, Verdon CP, Wu AH, Wang H, Prior RL. Automated assay of oxygen

radical absorbance capacity with the COBAS FARA II. Clin Chem 1995;41(12 Pt

1):1738-44.

149. Ou B, Hampsch-Woodill M, Prior RL. Development and validation of an

improved oxygen radical absorbance capacity assay using fluorescein as the

fluorescent probe. J Agric Food Chem 2001;49(10):4619-26.

150. Pettersen EF, Goddard TD, Huang CC, Couch GS, Greenblatt DM, Meng EC,
158

Ferrin TE. UCSF Chimera--a visualization system for exploratory research and

analysis. J Comput Chem 2004;25(13):1605-12.

151. Morris GM, Huey R, Lindstrom W, Sanner MF, Belew RK, Goodsell DS, Olson

AJ. AutoDock4 and AutoDockTools4: Automated docking with selective receptor

flexibility. J Comput Chem 2009;30(16):2785-91.

152. Sanner MF. Python: a programming language for software integration and

development. J Mol Graph Model 1999;17(1):57-61.

153. Wallace AC, Laskowski RA, Thornton JM. LIGPLOT: a program to generate

schematic diagrams of protein-ligand interactions. Protein Eng 1995;8(2):127-34.

154. Schrodinger, LLC. The PyMOL Molecular Graphics System, Version 1.3. 2010.

155. Weinreb O, Amit T, Bar-Am O, Youdim MB. Induction of neurotrophic factors

GDNF and BDNF associated with the mechanism of neurorescue action of

rasagiline and ladostigil: new insights and implications for therapy. Ann N Y

Acad Sci 2007;1122:155-68.

156. Heneka MT, Sastre M, Dumitrescu-Ozimek L, Hanke A, Dewachter I, Kuiperi C,

O'Banion K, Klockgether T, Van Leuven F, Landreth GE. Acute treatment with

the PPARgamma agonist pioglitazone and ibuprofen reduces glial inflammation

and Abeta1-42 levels in APPV717I transgenic mice. Brain 2005;128(Pt

6):1442-53.

157. Yan Q, Zhang J, Liu H, Babu-Khan S, Vassar R, Biere AL, Citron M, Landreth

G. Anti-inflammatory drug therapy alters beta-amyloid processing and deposition

in an animal model of Alzheimer's disease. J Neurosci 2003;23(20):7504-9.


159

158. He XM, Carter DC. Atomic structure and chemistry of human serum albumin.

Nature 1992;358(6383):209-15.

159. Peters T, Jr. Serum albumin. Adv Protein Chem 1985;37:161-245.

160. Crivori P, Cruciani G, Carrupt PA, Testa B. Predicting blood-brain barrier

permeation from three-dimensional molecular structure. J Med Chem

2000;43(11):2204-16.

161. Toulouse A, Sullivan AM. Progress in Parkinson's disease-where do we stand?

Prog Neurobiol 2008;85(4):376-92.

162. Youdim MB, Geldenhuys WJ, Van der Schyf CJ. Why should we use

multifunctional neuroprotective and neurorestorative drugs for Parkinson's

disease? Parkinsonism Relat Disord 2007;13 Suppl 3:S281-91.

163. Markesbery WR, Montine TM, Lovell MA. In: Mattson MP, editor. Pathogenesis

of Neurodegenerative Disorders. New Jersey: Humana Press; 2001. p 21–52.

164. Packer L, Tritschler HJ, Wessel K. Neuroprotection by the metabolic antioxidant

alpha-lipoic acid. Free Radic Biol Med 1997;22(1-2):359-78.

165. Radi R, Turrens JF, Chang LY, Bush KM, Crapo JD, Freeman BA. Detection of

catalase in rat heart mitochondria. J Biol Chem 1991;266(32):22028-34.

166. Milczek EM, Binda C, Rovida S, Mattevi A, Edmondson DE. The 'gating'

residues Ile199 and Tyr326 in human monoamine oxidase B function in substrate

and inhibitor recognition. FEBS J 2011;278(24):4860-9.

167. Hubalek F, Binda C, Khalil A, Li M, Mattevi A, Castagnoli N, Edmondson DE.

Demonstration of isoleucine 199 as a structural determinant for the selective


160

inhibition of human monoamine oxidase B by specific reversible inhibitors. J Biol

Chem 2005;280(16):15761-6.

168. Carter DC, Ho JX. Structure of serum albumin. Adv Protein Chem

1994;45:153-203.

169. Buchholz L, Cai CH, Andress L, Cleton A, Brodfuehrer J, Cohen L. Evaluation of

the human serum albumin column as a discovery screening tool for plasma

protein binding. Eur J Pharm Sci 2002;15(2):209-15.

170. Hodgson J. ADMET--turning chemicals into drugs. Nat Biotechnol

2001;19(8):722-6.

171. Rich RL, Day YS, Morton TA, Myszka DG. High-resolution and high-throughput

protocols for measuring drug/human serum albumin interactions using

BIACORE. Anal Biochem 2001;296(2):197-207.

172. Valko K, Nunhuck S, Bevan C, Abraham MH, Reynolds DP. Fast gradient HPLC

method to determine compounds binding to human serum albumin. Relationships

with octanol/water and immobilized artificial membrane lipophilicity. J Pharm Sci

2003;92(11):2236-48.

173. Kratochwil NA, Huber W, Muller F, Kansy M, Gerber PR. Predicting plasma

protein binding of drugs: a new approach. Biochem Pharmacol

2002;64(9):1355-74.

174. Benet LZ, Hoener BA. Changes in plasma protein binding have little clinical

relevance. Clin Pharmacol Ther 2002;71(3):115-21.

175. Herve F, Urien S, Albengres E, Duche JC, Tillement JP. Drug binding in plasma.
161

A summary of recent trends in the study of drug and hormone binding. Clin

Pharmacokinet 1994;26(1):44-58.

176. Lindup WE, Orme MC. Clinical pharmacology: plasma protein binding of drugs.

Br Med J (Clin Res Ed) 1981;282(6259):212-4.

177. Han XL, Mei P, Liu Y, Xiao Q, Jiang FL, Li R. Binding interaction of quinclorac

with bovine serum albumin: a biophysical study. Spectrochim Acta A Mol

Biomol Spectrosc 2009;74(3):781-7.

178. Pardridge WM. Blood-brain barrier drug targeting: the future of brain drug

development. Mol Interv 2003;3(2):90-105, 51.

179. Minn A, Ghersi-Egea JF, Perrin R, Leininger B, Siest G. Drug metabolizing

enzymes in the brain and cerebral microvessels. Brain Res Brain Res Rev

1991;16(1):65-82.

180. Ghersi-Egea JF, Leninger-Muller B, Suleman G, Siest G, Minn A. Localization of

drug-metabolizing enzyme activities to blood-brain interfaces and

circumventricular organs. J Neurochem 1994;62(3):1089-96.

181. Dutheil F, Dauchy S, Diry M, Sazdovitch V, Cloarec O, Mellottee L, Bieche I,

Ingelman-Sundberg M, Flinois JP, de Waziers I and others.

Xenobiotic-metabolizing enzymes and transporters in the normal human brain:

regional and cellular mapping as a basis for putative roles in cerebral function.

Drug Metab Dispos 2009;37(7):1528-38.

182. Greeff J, Joubert J, Malan SF, van Dyk S. Antioxidant properties of 4-quinolones

and structurally related flavones. Bioorg Med Chem 2012;20(2):809-18.


162

183. Turnquist TD, Sandell EB. Stability constants of iron(III)-8-hydroxyquinoline

complexes. Analytica Chimica Acta 1968;42(0):239-245.

184. Kayyali R, Pannala AS, Khodr H, Hider RC. Comparative radical scavenging

ability of bidentate iron (III) chelators. Biochem Pharmacol 1998;55(8):1327-32.

185. Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and

computational approaches to estimate solubility and permeability in drug

discovery and development settings. Adv Drug Deliv Rev 2001;46(1-3):3-26.

186. Norinder U, Haeberlein M. Computational approaches to the prediction of the

blood-brain distribution. Adv Drug Deliv Rev 2002;54(3):291-313.

187. Veber DF, Johnson SR, Cheng HY, Smith BR, Ward KW, Kopple KD. Molecular

properties that influence the oral bioavailability of drug candidates. J Med Chem

2002;45(12):2615-23.

188. Kelder J, Grootenhuis PD, Bayada DM, Delbressine LP, Ploemen JP. Polar

molecular surface as a dominating determinant for oral absorption and brain

penetration of drugs. Pharm Res 1999;16(10):1514-9.

189. Ahluwalia VK, Aggarwal R. Comprehensive Practical Organic Chemistry:

Preparation and Quantitative Analysis. 2001.

190. Moran JF, Klucas RV, Grayer RJ, Abian J, Becana M. Complexes of iron with

phenolic compounds from soybean nodules and other legume tissues: prooxidant

and antioxidant properties. Free Radic Biol Med 1997;22(5):861-70.

View publication stats

You might also like