Download as pdf
Download as pdf
You are on page 1of 11

Challenges

Neuroprotection by monoamine
oxidase B inhibitors:
a therapeutic strategy
for Parkinson’s disease?{
Rinat Tabakman,{ Shimon Lecht, and Philip Lazarovici*

Summary Recent investigations into the neuroprotective mechan-


Parkinsonism (PD) is a neurodegenerative disorder of the ism of propargylamines indicate that glyceraldehyde-3-
brain resulting in dopamine deficiency caused by the phosphate dehydrogenase (GAPDH), MAO-B and/or
progressive death of dopaminergic neurons. PD is char- other unknown proteins may represent pivotal proteins
acterized by a combination of rigidity, poverty of move- in the survival of the injured neurons. Delineation of the
ment, tremor and postural instability. Selegiline is a mechanism(s) involved in the neuroprotective effects
selective and irreversible propargylamine type B mono- exerted by MAO-B inhibitors may provide the key to
amine oxidase (MAO-B) inhibitor. This drug, which in- preventive novel therapeutic modalities. BioEssays
hibits dopamine metabolism, has been effectively used in 26:80–90, 2004. ß 2003 Wiley Periodicals, Inc.
the treatment of PD. However, its therapeutic effects are
compromised by its many neurotoxic metabolites. To
circumvent this obstacle, a novel MAO-B inhibitor, rasagi-
line, was developed. Paradoxically, the neuroprotective Introduction
mechanism of propargylamines in different neuronal Parkinson’s disease (PD) is a progressive movement disorder
models appears to be independent of MAO-B inhibition. that affects 1–2%of the adult population over 60 years of age.
The main symptoms are tremor at rest, muscular rigidity and a
decrease in the frequency of voluntary movements (hypoki-
Department of Pharmacology, School of Pharmacy, Faculty of nesia).(1) For many years, it has been known that PD synd-
Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel. rome is due to a disorder of the basal ganglia, brain structures
*Correspondence to: Philip Lazarovici, Department of Pharmacology
regulating motor activity and innervated by one of the brain’s
and Experimental Therapeutics, School of Pharmacy, Faculty of
Medicine, The Hebrew University, Jerusalem, 91120, Israel.
major dopaminergic pathways. The neurochemical basis for
E-mail: lazph@md.huji.ac.il the disease was discovered in 1960 by Hornykiewics,(2) who
y
This review is part of a PhD thesis to be submitted to the Hebrew showed that the dopamine content of the substantia nigra (SN)
University of Jerusalem by TR. and corpus striatum in postmortem PD brains was extremely
z
The scientific views and opinions expressed in this article are solely
low (less than 10% of normal). Pathologically, PD is charac-
those of the authors and are not to be construed as having any
commercial interest.
terized by progressive degeneration of pigmented brain stem
DOI 10.1002/bies.10378 nuclei, mostly the pars compacta of the SN, along with the
Published online in Wiley InterScience (www.interscience.wiley.com). formation of characteristic eosinophilic cytoplasmic inclusions
known as Lewy bodies.(3) The disease progresses slowly for
many years. The clinical symptoms are due to the degenera-
tion (death) of dopaminergic neurons in the SN, resulting in a
Abbreviations: PD, Parkinson’s disease; UPDRS, unified Parkinson’s dramatic decline in dopamine level. Diagnosis can be made
disease rating scale; SN, substantia nigra; DA, dopaminergic; MAO-B,
when at least 60% of the dopaminergic SNc neurons are lost.(4)
type B monoamine oxidase; GAPDH, glyceraldehyde-3-phosphate
dehydrogenase; SOD, superoxide dismutase; NADþ, nicotinamide
Many factors are considered to contribute to the pathogenesis
adenine dinucleotide; FAD, flavin adenine dinucleotide; BDNF, brain- of PD: genetic,(5) age-related,(6) enviromental toxins,(7) and
derived neurotrophic factor; NGF, nerve growth factor; GDNF, glial oxidative stress.(8,9) In the past decade, novel hypotheses
derived neurotrophic factor; DMS, desmethylselegiline; MPPþ, 1- have been forwarded suggesting important pathological con-
methyl-4-phenyl pyridinium; MPTP, N-methyl-4-phenyl-1,2,3,6-tetra-
tributions to dopaminergic neuronal cell death: glutamate
hydropyridine; 6-OH-DA, 6-hydroxydopamine; OGD, oxygen–glucose
deprivation; PET, positron emission tomography; fMRI, functional
neurotoxicity,(10) mitochondrial abnormalities, disturbances in
magnetic resonance imaging; IC50, 50% inhibitory concentration; Ki, intracellular calcium homeostasis, altered iron metabolism
inhibition constant; PC12, pheochromocytoma cells. and apoptosis.(9,11 –14) Although the genes responsible for
a few rare familial cases have been uncovered,(15–17) the

80 BioEssays 26.1 BioEssays 26:80–90, ß 2003 Wiley Periodicals, Inc.


Challenges

molecular basis for the more prevalent idiopathic PD remains Therapeutical approaches
unknown. Novel technologies including DNA-microarray(18) to Parkinson’s disease
and single nucleotide polymorphisms(19) are expected to As the cause of the death of the dopaminergic neurons is not
pinpoint the pathogenesis of this disorder. In addition, new known, drugs that can arrest and/or reduce and/or delay the
techniques in neuroimaging such as positron emission tomo- cell death process in dopaminergic neurons are not available.
graphy (PET) and functional magnetic resonance imaging However, significant symptomatic relief has been obtained by
(fMRI) may provide better means of assessing SN function.(20) the use of different dopaminergic drugs that fall into the follow-
In this review, we briefly summarize the emerging knowledge ing categories: dopamine precursors (levodopa); compounds
regarding the complex mechanism of action of type B mono- mimicking (agonists) the action of dopamine (bromocriptine);
amine oxidase (MAO-B) inhibitors, an important family of agents that prevent dopamine degradation (MAO-B inhibitors
drugs used to treat PD. While the search for MAO-B inhibitors such as rasagiline).
has turned up some promising propargylamine candidates, Levodopa, a dopamine precursor that crosses the blood–
their mode of action in ameliorating PD cannot be explained brain barrier, is initially effective in most patients, but often
solely by MAO-B inhibition. Today, the basis for their neuro- loses its efficacy after several years of treatment, the period
protective mechanism of action is being actively pursued. We varying from one patient to another. Complications associated
hypothesize that the beneficial role of MAO-B inhibitors in PD with its use are involuntary movements, which occur in many
is due to a shift from neurotoxicity to neuroprotection of the patients within several years, and an unpredictable ‘‘on–off’’
injured dopaminergic neurons. effect in the course of treatment.(31) Other adverse effects
include queasiness (nausea), a drop in blood pressure
(hypotension), and, occasionally, psychotic symptoms.(31)
Apoptosis and Parkinson’s disease Levodopa treatment requires a certain number of live dopami-
Since 1996, when apoptotic death was discovered in nergic neurons to convert this metabolic precursor into the
dopaminergic cells of Parkinsonian patients,(21) many addi- neurotransmitter dopamine (DA). As a result, the amount of
tional studies have suggested the involvement of an apoptotic synaptic DA is increased and the DA deficiency characteristic
mechanism in this disorder. Apoptosis, a delayed form of cell of PD is corrected. The disadvantage of this therapeutic
death associated with the activation of a ‘‘genetic program’’, is approach is the augmented cell death of large numbers of DA
an important biological process controlling cell number in neurons during PD progression, leading to less DA synthesis
various tissues. This mechanism plays an important role in the during levodopa treatment and rendering it less effective.
development of the nervous system(22) and is also triggered Drugs that mimic DA (agonists) are less effective than
following pathological events such as stroke and neurode- levodopa.(32) They activate postsynaptic DA receptors in the
generative diseases.(14) The SN of PD patients shows an SN and improve motor function independently of DA neuron
increase in caspase activity and in the pro-apoptotic Bax degeneration.
protein, as well as nuclear translocation of glyceraldehyde-3- The first generation of MAO inhibitors, originally synthe-
phosphate dehydrogenase (GAPDH), indicating the activation sized in the 1950s, was used as antidepressants due to their
of apoptotic signals.(23–25) However, it is not clear whether the inhibitory effect on the metabolism of monoamine neurotrans-
degeneration associated with PD involves a fast ‘event’ in mitters. These inhibitors are both irreversible and non-selec-
which a significant number of dopaminergic neurons deterio- tive. However, their use as antidepressants was restricted
rate as a result of unknown factor(s),(26) or a very slow one that because of hepatotoxicity(33) and due to an increase in blood
may last several decades.(27) Furthermore, other postmortem pressure (hypertensive response) following the ingestion of
studies, based on nick-end labeling of fragmented DNA,(28) some foods and drinks containing tyramine,(34) which are nor-
Bcl-2 and Bax apoptotic protein markers,(27) failed to show a mally metabolized in the gastrointestinal tract by MAO.(34,35)
significant contribution of apoptosis to SN dopaminergic As a result of MAO inhibition, the ingested tyramine enters the
neuronal cell death. An additional complication adding to the circulation and is actively taken up by peripheral adrenergic
cell death controversy in PD is the finding that, in postmortem neurons, displacing stored noradrenaline and giving rise to a
tissue, apoptotic markers are present in the glia but not in SN hypertensive response that can be fatal.(36) This side-effect
neurons.(29,30) In general, nigrostriatal dopaminergic neurons pharmacological phenomenon(37) was named the ‘‘cheese
are lost with age. This phenomenon is dramatically acceler- effect’’, since many cheeses are rich in tyramine. The next
ated in Parkinson’s patients. The grounds for the vulnerability discovery was made by Johnston,(38) who showed that the
of the nigrostriatal dopaminergic neurons are unknown and the MAO enzyme exists in two forms: A and B. This led to the
precise mechanism of cell death awaits further investigation. synthesis of a new generation of MAO inhibitors exhibiting
The gene-expression microarray technique is expected to greater selectivity toward the individual isoforms. MAO-A
make a significant contribution to the identification of gene inhibitors are potent antidepressants but, since MAO-A is the
products involved in cell death and leading to PD.(18) major isoform in the intestine, they induce the ‘‘cheese effect’’.

BioEssays 26.1 81
Challenges

The finding that MAO-B inhibitors have weaker antidepressant consists of propargylamine derivatives. These are irreversible
activity, do not induce the ‘‘cheese effect,’’(39) but can amelio- inhibitors, which would ensure continuous dopaminergic
rate PD symptoms, was the basis for modern Parkinson stimulation, considered a major therapeutic goal. Lazabemide,
therapeutics,(39,40) using the MAO-B inhibitor selegiline and a 2-aminoethyl carboxamide derivative, which is a reversible,
rasagiline. Selegiline has been approved for use in the USA as highly selective antioxidant, was developed as a MAO-B
an adjunct to levodopa but not for monotherapy; rasagiline is a inhibitor for treatment of PD.(46) Clinical trials with this com-
new addition to the MAO-B inhibitor group. pound were discontinued in phase III of the clinical study due
to the abnormal liver functions that developed in patients
Monoamine oxidases and inhibitors undergoing this treatment. To date, of the variety of selective
in Parkinson’s disease MAO-B inhibitors investigated, only one, selegiline (l-depre-
MAO (EC 1.4.3.4) oxidatively deaminates monoamine neuro- nyl) (Table 1), has been in clinical use since the mid 1970s.(47)
transmitters (norepinephrine, epinephrine, serotonin and Rasagiline (N-propargyl-1-R-aminoindan) (Table 1), a novel,
dopamine), as well as exogeneous amines (e.g., tyramine). potent, selective, and irreversible inhibitor of MAO-B, absorb-
The end products of the enzymatic reaction are aldehydes and ed after oral administration,(48,49) is currently being evaluated
hydrogen peroxide, both toxic to DA neurons. The physio- in phase III clinical trial.(40) In contrast to selegiline, rasagiline is
logical role of MAO is to terminate the action of several not metabolized to potentially toxic amphetaminic metabolites
neurotransmitters and to detoxify exogenous monoamines. and its major metabolite, 1-R-aminoindan, has exhibited
Two MAO subtypes, MAO-A and MAO-B, were defined in beneficial effects in animal models of PD.(50) Because of its
1968,(38) based on their differential sensitivity to irreversible selectivity towards MAO-B, rasagiline does not trigger the
inhibitors. 20 years later, the corresponding cDNAs of these ‘‘cheese effect’’,(51) and was found to improve motor and
two subtypes were cloned. Sequence comparison indicated cognitive function in PD animal models.(50) A recent clinical
72.6% amino acid homology.(41,42) The genes encoding the trial demonstrated the beneficial influence of rasagiline mono-
two subtypes have been localized to human chromosome therapy in early stages of Parkinson’s disease.(40) To evaluate
Xp11.23-Xp11.4.(43) The MAO-A and MAO-B genes are homo- the safety and efficacy of rasagiline, a multicenter clinical trial
logous at the level of intron–exon organization and in co-factor was carried out in which a total of 404 patients in early stages of
FAD-binding site, and both enzymes are located at the outer PD were included. The participants were randomly assigned to
mitochondrial membrane.(44) Along with the similarities, MAO- groups receiving rasagiline at a dose of 1–2 mg per day or
A and MAO-B display differences in their numbers of amino corresponding placebo. A one-week escalation period was
acids, 527 and 520, and molecular weights 59.7 kDa and followed by a 25-week maintenance period. The efficacy of
58.8 kDa, respectively. The in vivo substrate affinity in humans treatment was evaluated according to the Unified Parkinson’s
also differs: MAO-A metabolizes mainly serotonin, norepi- Disease Rating Scale (UPDRS); rasagiline monotherapy at
nephrine and epinephrine, whereas MAO-B metabolizes both doses proved effective.(40)
dopamine and phenylethylamine.(44) Both MAO-B and MAO- Although inhibition of MAO-B is of prime importance in
A are widely distributed in the human body, the ratio between controlling PD symptoms, it appears that this inhibition under-
them varying in different tissues. The highest value (>69) is lies many additional effects in the brain. An interesting analogy
found in platelets; lower ratios were detected in the human can be drawn between PD patients chronically administered
brain, the highest of which (5.5) was found in the SN.(45) The MAO-B inhibitors and knockout mice lacking the gene encod-
selectivity toward different endogenous substrates, together ing MAO-B. Chen et al. demonstrated significant upregulation
with tissue-specific localization, determines the clinical poten- (30%) of D2 receptors in the striatum and the functional
tial of each isoform. Inhibition of MAO activity in the brain supersensitivity of D1 receptors in the mouse nucleus accum-
increases the synaptic level of the neurotransmitters seroto- bens.(52) This upregulation resembles similar regulation of the
nin, noradrenaline and dopamine. This mechanism was dopamine receptors observed in PD patients chronically
exploited to treat depression, first with nonselective MAO treated with MAO-B inhibitors. It has been suggested that
inhibitors and, more recently, with selective MAO-A inhibitors. MAO-B makes a smaller contribution than MAO-A to dopamine
Today however, inhibitors of MAO-A are not the drugs of first metabolism in the mouse, as compared with man,(53) making it
choice in the treatment of depression, mainly because of the difficult to conclude that similar changes occur in MAO-B
development of novel antidepressants that act by different inhibitor-treated patients. However, subjects in whom the
pharmacological mechanisms, without eliciting the cheese MAO-B gene is deleted (atypical Norrie disease) have normal
effect. levels of DA metabolites in their plasma.(54) The parallelism
Many selective MAO-B inhibitors, potentially applicable to between MAO-B knockout mice and humans with a deleted
PD treatment, have been synthesized. They are categorized MAO-B gene, suggests a similarity in the lack of change in
according to chemical structure, each including several DA metabolites levels This additional circumstantial evidence
derivatives (Table 1). The most-potent and selective group leads us to postulate that upon treatment with MAO-B

82 BioEssays 26.1
Challenges

Table 1. Classification of MAO-B inhibitors of clinical relevance, according to chemical structure, potency,
mechanism of action and selectivity
MAO-B inhibition Mechanism Selectivitya
Chemical group Chemical structure (IC50 nM) of inhibition MAOB/MAOA References

Propargylamine derivatives 6 Irreversible 233 95


Selegiline

Rasagiline 30 Irreversible 100 95

Allylamine derivatives 100 Irreversible 50 95

2-aminoethyl carboxamide 37 Reversible 26568 95


derivatives
Lazabemide

N-allenic indolalkylamine 25–25000 Irreversible >1 96


derivatives

4-substituted 120–260b Irreversible — 97


cubylcarbinyl amines

a
Ratio between MAO-B and MAO-A (IC50). High value indicates greater selectivity towards MAO-B.
b
Ki.

inhibitors, DA receptors undergo alterations seen in the MAO- strategy in the last 15 years has been based on the DA
B knockout mice. However, changes in dopamine receptors precursor levodopa, which raises the DA level. The adverse
are not the sole effect of the MAO-B inhibitors. Recent studies effects of the treatment, the fact that this compound does
show the potential for an additional beneficial activity: not arrest dopaminergic cell death and disease progression
neuroprotection. Cumulative in vivo and in vitro evidence and the possible contribution of apoptotic cell death to
points to the neuroprotective properties of propargylamine Parkinson’s disease, prompted the search for new neuropro-
MAO-B inhibitors, such as selegiline and rasagiline.(55–57) tective approaches. A neuroprotective therapeutic or disease-
modifying modality may be defined as an intervention that
MAO-B inhibitors and neuroprotection delays or prevents neuronal cell death and thus affects
in different neurotoxic models disease progression.
Since PD progresses very slowly and over many years, its Selegiline aroused considerable interest as a potential
diagnosis poses a tremendous challenge. Because the trigger neuroprotective drug. As summarized in Tables 2 and 3,
for dopaminergic cell death is unknown, the therapeutic preclinical studies point to the neuroprotective effect of the

BioEssays 26.1 83
Challenges

Table 2. Neuroprotective in vitro effects of MAO-B inhibitors selegiline and rasagiline


Neurotoxicity Neuroprotective
MAO-B Inhibitor Insult Model mechanism mechanism References

Selegiline Excitotoxicity Mesencephalic Depolarization and Yes 98


dopaminergic, primary calcium overload Independent of MAO-B
neurons
Hypocampal glia and Oxidative stress Yes, release of NGF 99
neurons
þ
MPP Dopaminergic neurons Oxidative stress Yes 100
mesencephalic and
striatal cells
BSO L-buthionine-(S,R)- Mesencephalic neurons Glutathione depletion Yes 101
sulfoximine Independent of MAO-B
Serum and NGF NGF-partially Oxidative stress and Yes 102
withdrawal differentiated rat growth factor Independent of
PC12 cells starvation MAO-B, induction
of transcription
Natural cell death Mesencephalic neurons Apoptosis and Yes 49
combined with serum oxidative stress Serum-enhanced 103
deprivation 104
Serum deprivation and E1A-NR3 immortalized Oxidative stress and Yesa 105
hypoxia retinal neurons hypoxia
SIN-1 Neuroblastoma Oxidative stress Yes 77
SH-SY5Y cells
Rasagiline Natural cell death Mesencephalic neurons Apoptosis and Yes 49
combined with serum oxidative stress Serum-enhanced 103
deprivation
SIN-1 Neuroblastoma Oxidative stress Yesb 75
SH-SY5Y cells 76
77
6-OHDA, SIN-1 Neuroblastoma SH-SY5Y Oxidative stress Yesb 76
cells Independent of MAO-B
N-methyl(R)salsolinol, Neuroblastoma Oxidative stress Yesb,c 106
6-OHDA, peroxynitrite SH-SY5Y cells Independent of MAO-B
N-methyl(R)salsolinol SH-SY5Y cells Oxidative stress Yesd 107
overexpressing Bcl-2
e
Serum and NGF Partially differentiated Free radical-induced Yes 48
withdrawal PC12 cells apoptosis
Oxidative stress Independent of MAO-B
OGD NGF-differentiated Oxygen-glucose Yes 57
PC12 cells deprivation Independent of MAO-B 60

a
Regulation of apoptosis-related gene expression.
b
Stabilization of mitochondrial potential.
c
Suppresion of caspases and DNA fragmentation.
d
Prevention of nuclear accumulation of GAPDH.
e
Increased gene expression of anti-apoptotic targets (proteins).

propargylamine MAO-B inhibitors selegiline and rasagiline. sult,(57,60) prevents deficits in behavioral parameters following
This is illustrated by in vitro studies using a variety of hypoxia in adult and senescent rats,(61) protects from experi-
dopaminergic and nondopaminergic neurons taken from mental focal ischemia in the rat,(62) and from closed head injury
different species and exposed to a range of neurotoxic insults in the mouse.(63) The last three are in vivo models. Although the
(excitotoxicity, oxidative stress, depolarization, growth factor neuroprotective in vitro effect of selegiline, and even more so
withdrawal and oxygen glucose deprivation). For example, of rasagiline, is clear-cut, the lack of reliable pathophysiological
rasagiline prolongs the survival of cultured primary human and models for the disease has led to some skepticism among the
rat DA neurons under serum-free conditions,(58) reduces Parkinson medical community. The PD models using neuronal
glutamate toxicity in hippocampal neurons,(59) protects nerve cultures treated with 1-methyl-4-phenyl pyridinium (MPPþ, a
growth factor (NGF)-differentiated pheochromocytoma PC12 metabolite of the neurotoxin N-methyl-4-phenyl-1,2,3,6-tetra-
cells from oxygen-glucose-deprivation (OGD-ischemia) in- hydropyridine (MPTP)) and 6-hydroxydopamine (6-OH-DA)

84 BioEssays 26.1
Challenges

Table 3. Neuroprotective in vivo effects of MAO-B inhibitors selegiline and rasagiline


Neurotoxicity Neuroprotective
MAO-B Inhibitor Insult Model mechanism mechanism References

Selegiline Permanent or transient Rat Focal or whole Yes 99


occlusion of cerebral artery Mouse brain ischemia 108
Gerbil 109
Selegiline Unilateral MPPþ neurotoxicity Sprague-Dawley rat Oxidative stress Yes 110
Independent of MAO-B
Rasagiline, Selegiline MPTP Monkey Oxidative stress Yes 111
Rasagiline Permanent middle cerebral Wistar rat Brain ischemia Yes Independent of MAO-B 62
artery occlusion
Rasagiline Closed head injury Mouse Mechanical brain Yes 48
trauma

to trigger cell death, are the most common, and less cultures lacking MAO-B on the one hand, and the absence
disputable. They are the main models used for screening of any neuroprotective effect by clorgyline, a MAO-A inhibitor,
and developing novel drugs for PD treatment, although the on the other, also support the lack of correlation between
animals injected with the neurotoxin MPTP do not exhibit Lewy neuroprotection and MAO inhibition.(57) Furthermore, the
bodies, a hallmark of PD. propargylamine concentration required for neuroprotection
While a variety of neurological deficits are triggered by the both in vitro and in vivo is 1 mM, whereas the IC50 or Ki value for
above-mentioned insults in in vivo animal models, mainly the in vitro MAO-B inhibition is in the range of 6–30 nM. As shown
MPTP model partially mimics the PD syndrome. In rats fed in Tables 2 and 3, the major insults evoking neurotoxicity and,
selegiline, the lifespan of the animals was increased, in parallel in many cases, leading to neuronal apoptosis, are depolariza-
with enhanced catecholaminergic activity in the brain.(64,65) tion, calcium overload and oxidative stress. Conceptually,
This phenomenon was unrelated to MAO-B inhibition and therefore, the neuroprotective effect of propargylamines might
was a topic of controversy. The neuroprotective effects of the be grounded in the prevention of cell death or the activation of
MAO-B inhibitors selegiline and rasagiline have also been pro-survival pathways minimizing cellular stress.
investigated in mice, gerbils and monkeys (Table 3), in brain- Rasagiline and selegiline exert their antioxidant effect
ischemia, head-trauma, and under MPTP toxicity. As opposed by upregulating the enzymes involved.(68) Selegiline(69) and
to the clearly neuroprotective effects of propargylamines in rasagiline,(70) promote free radical scavenging by activating
in vitro cellular systems and in vivo experimental animal superoxide dismutase (SOD1 and SOD2) and catalase or by
models, clinical studies with selegiline have failed to distin- increasing the SOD protein level,(71) upon chronic adminis-
guish between the contribution of MAO-B inhibition and tration to rats. In vitro experiments in our laboratory using cyclic
MAO-B induced neuroprotection to the amelioration of clinical voltammetry further substantiated the antioxidant properties of
symptoms.(66) Although selegiline delayed the need for levo- these compounds. Selegiline displays antioxidant effects
dopa, its effect on disease progression has not been proved. in vivo, which are probably not related to MAO-B inhibition
As for rasagiline, a placebo (control) study revealed that since selegiline reduced the free radical level at a pM con-
patients with early PD who were treated with rasagiline for centrations too low to inhibit MAO-B activity.(72) The neuro-
12 months, showed less decline in neurological symptoms protective effect of selegiline could also be mediated indirectly
(UPDRS score) than patients whose rasagiline treatment via antagonistic modulation of the polyamine’s binding site on
was delayed for 6 months. These clinical results cannot be the NMDA receptor, thereby reducing NMDA-receptor-gener-
explained by the purely symptomatic effect of rasagiline,(67) ated excitotoxicity.(73) Moreover, rasagiline and selegiline act
and they may represent a neuroprotective effect. directly as anti-apoptotic agents, probably by interfering with
the cellular apoptotic cascade (pro-survival) (Table 2). Several
Insights into the neuroprotection mechanism: studies have shown that treatment of cells with rasagiline
lack of correlation between neuroprotection causes upregulation of putative anti-apoptotic and antioxidant
and MAO-B inhibition proteins such as Bcl-2, SOD, glutathione and BCLXL.(74) It has
The neuroprotective effect of propargylamines, demonstrated been suggested that rasagiline protects cells from apoptosis
in different insult models, both in vitro and in vivo, suggests that by stabilizing the mitochondrial membrane potential(75,76)
it is not due to MAO inhibition (Tables 2 and 3). The finding that since such stabilization showed a causal relationship to inhibi-
propargylamines induce neuroprotection in neuronal cell tion of caspase activity and prevention of DNA fragmentation.

BioEssays 26.1 85
Challenges

In any event, the propargyl group of rasagiline and selegiline apoptotic process in the cells. It is very tempting to suggest
is essential to MAO-B inhibition and the anti-apoptotic that the active site of these proteins possesses an FAD-
effect.(77) binding domain such as in MAO-B.(83) Recently, the structure
of human MAO-B was determined by crystalization at 3Å
Molecular neuroprotective mechanism resolution. Electron density analysis revealed that pargyline, a
of MAO-B inhibitors selegiline analog, binds covalently to N5 of the flavine
The precise target and details of the molecular mechanisms nucleotide.(83) Therefore, it is conceivable that the insertion
involved in the neuroprotective effect exerted by propargy- of propargylamines into the FAD pocket(83) of certain proteins
lamines are not known. A major obstacle in mechanistic with a structure similar to that of MAO-B will be followed by
studies on selegiline is that the molecule is largely (/80%) covalent binding to FAD or NAD, as in MAO-B, resulting in
converted into amphetamines, some of which display neuro- neuroprotection.
toxic activity.(78) Cumulative evidence suggests that brain Hydrogen peroxide (H2O2), which is a by-product of the
injury following amphetamine and methamphetamine admin- enzymatic reaction catalyzed by MAO-B, has been shown to
istration is due to increased free radical formation and mito- inhibit GAPDH. This inactivates the glycolytic and mitochon-
chondrial damage, leading to a failure in cellular energy drial pathways of ADP phosphorylation, resulting in a drop in
metabolism followed by secondary excitotoxicity-induced intracellular ATP level and cell death. It was shown that MAO-
seizures.(78) The neuroprotective activity of selegiline derives B inhibition by propargylamines confers neuroprotection
from a minor metabolite, desmethylselegiline (DMS), which from H2O2-induced injury due to GAPDH damage.(84) It
was shown to be neuroprotective in vitro.(79) However, the was recently demonstrated that the level of GAPDH and its
amphetamine-like major metabolites may cause cell da- nuclear accumulation increase in apoptotic models of serum
mage(80) and thus could reverse the potential beneficial withdrawal, growth factor deprivation and MPPþ-induced
effects of DMS. Indeed, the addition of L-methamphetamine neurotoxicity, pointing to the enzyme’s essential role in apop-
to oxygen–glucose-deprived PC-12 cells substantially in- tosis.(85,86) Furthermore, propargylamines prevented both
creased cell death.(57) When both L-methamphetamine and these phenomena, suggesting that this activity rather than
selegiline were added to PC-12 cells exposed to OGD, the MAO-B inhibition, may contribute to the neuroprotective
protective effect of the latter was reduced.(57) In contrast to effect.(85) Another important finding was the subcellular redu-
selegiline, whose neuroprotective activity derives from its ction in GAPDH activity in Alzheimer’s disease and Hunting-
minor metabolite, DMS, that of rasagiline is mediated solely by ton’s disease.(87) The investigators posited that intracellular
the parent drug(57,81) and its major metabolite, 1-R-amino- formation of complexes between GAPDH and proteins chara-
indan, is not toxic.(57) cteristic of neurodegenerative disorders, such as a-synuclein
An interesting attempt to identify the target of the propargy- in PD, b-amyloid in Alzheimer’s or huntingtin in Huntington’s,
lamine anti-apoptotic effect was made by Kragten et al. in may represent an emerging cellular phenotype of neurode-
1998.(82) They synthesized a selegiline derivative (CGP 3466) generative disorders The major challenges in this field will be
that is neuroprotective but does not inhibit MAO and is not to further clarify the apoptotic role of GAPDH, the pathophy-
metabolized to amphetamines. Using affinity binding, labeling siological meaning of GAPDH complexes, to identify novel
and BIAcore technology (in which CGP 3466 was covalently anti-apoptotic and/or pro-survival FAD- or NAD-binding
bound to the surface of a flow cell with a CM5 sensor chip(82)), proteins and to delineate the interaction between these novel
they detected in rat brain lysates four protein bands of enzymes and propargylamines in neuroprotection.
38,43,50 and 299 kDa, all putative endogeneous binding sites
for CGP 3466. One of these proteins (38 kDa) was identified Neuroprotection strategy perspectives
as glyceraldehyde-3-phosphate dehydrogenase (GAPDH). In the present review, we have focused on the neuroprotection
Using GAPDH antisense oligonucleotides, the investigators of MAO-B inhibitors, their chemical characterization and their
established the importance of this enzyme in mediating the cellular and molecular mechanisms of action. It is impossible in
neuroprotective effect of CGP 3466. Although the precise so brief a review to cite all the earlier work recently summarized
role of GAPDH in neuronal apoptosis and in the neuroprotec- by Magyar and Vizi.(88) We have included a few examples to
tive mechanism of propargylamines awaits further investiga- illustrate the newly emerging apoptotic concept of dopami-
tion, GAPDH is the first selective target reported for these nergic neuronal degeneration and have described selected
compounds. GAPDH binds nicotinamide adenine dinucleotide studies on the neuroprotective contribution of MAO-B inhi-
(NADþ)(82) at a site structurally similar to that of the flavin bitors such as propargylamines. The majority of in vitro and
adenine dinucleotide (FAD)-binding enzymes, to which MAO- in vivo PD studies attribute the neurodegeneration of SN
B belongs. Therefore, we propose that propargylamines bind dopaminergic neurons to a long series of insults such as:
to GAPDH or other proteins with a tertiary structure similar to mitochondrial impairment and oxidative stress, Ca2þ overload,
that of the active site of MAO-B, thereby initiating an anti- iron metabolism, neurotoxins, excitotoxicity, neurotrophins,

86 BioEssays 26.1
Challenges

oxygen and glucose deprivation, depolarization and inflam- agonists could prove beneficial in the treatment of this
mation. It is conceivable that the neuroprotective effect disorder. Development of MAO-B inhibitors with neurotro-
of propargylamines is due to a shift in the neurotoxicity- phin-like activity may be a future strategy in PD therapy. This
neuroprotection balance towards survival (Fig. 1). MAO-B, possibility is supported by the recent finding that rasagiline
GAPDH and other FAD/NAD-binding proteins represent increases GDNF production and release by neuroblastoma
pivotal targets of the balance, as reflected by the neuropro- cells.(92) Despite the uncertainties and difficulties attendant
tective effect of selegiline and rasagiline. Therefore, a pharma- upon neuroprotective therapy in PD, clinical trials with innovat-
ceutical approach towards neuroprotection would be to ive neuroprotective agents must proceed for neuroprotection
synthesize novel MAO-B/GAPDH inhibitors. Such inhibitors to become a reality.
should help to launch mechanistic studies aimed at elucidating In addition to the use of low molecular weight drugs for PD
the molecular interactions of these compounds in anti-apo- treatment, modern therapeutic tactics focus on cell and gene
ptotic and/or survival signaling pathways. A crucial need in PD therapy. Cell therapy includes implantation in the SN of em-
drug development is the establishment of new in vitro and bryonic or neuronal stem cells, which can synthesize and
in vivo models of the disease. Once these models are avail- release dopamine to correct the PD syndrome.(93) Gene
able, it will be necessary to identify neuronal dopaminergic therapy in PD should aim both at supplementing the low SN
apoptotic pathways, as well as survival pathways. One of the dopamine level by introducing the genes encoding dopamine-
most remarkable changes recently observed in the nigro- synthesizing enzymes into non-dopaminergic cells in the
striatal region of the PD brain is the decreased level of neuro- striatum, and at supporting the survival of dopaminergic
trophins supporting dopaminergic neuron survival, such neurons by preventing apoptosis through the introduction of
as brain-derived neurotrophic factor (BDNF), NGF(89) and genes blocking this process.(94)
glial-derived neurotrophic factor (GDNF).(90) Therefore, re- We anticipate that, in coming years, the cellular pathophy-
combinant neurotrophins or compounds increasing the local siology of Parkinson’s disease will be clarified, allowing the
production of neurotrophins,(91) and/or neurotrophin receptor- design of new drugs and novel therapeutic approaches.

Figure 1. Schematic depicting the balance be-


tween neurotoxicity and neuroprotection in Par-
kinson’s disease. Upper part: Normal population of
SN dopaminergic neurons, consisting mainly of
live neurons (blue) and a few dead neurons (red).
Enzymes (green) (targeted by propargylamines)
pivot the balance between cell survival (#) and cell
death ("). Insults shift the balance towards
neurotoxicity; rasagiline and selegiline tip the
balance towards neuroprotection.

BioEssays 26.1 87
Challenges

References 25. Tatton NA. Increased caspase 3 and Bax immunoreactivity accompany
1. Sethi KD. Clinical aspects of Parkinson’s disease. Curr Opin Neurol nuclear GAPDH translocation and neuronal apoptosis in Parkinson’s
2002;15:457–460. disease. Exp Neurol 2000;166:29–43.
2. Hornykiewicz O, Kish SJ. Biochemical pathophysiology of Parkinson’s 26. Clarke G, Collins RA, Leavitt BR, Andrews DF, Hayden MR, Lumsden CJ,
disease. Adv Neurol 1987;45:19–34. McInnes RR. A one-hit model of cell death in inherited neuronal
3. Jellinger K. Pathology of Parkinson’s syndrome. In: Calne DB, editor. degenerations. Nature 2000;406:195–199.
Handbook of Experimental Pharmacology. Berlin: Springer-Verlag; 1989. 27. Wullner U, Kornhuber J, Weller M, Schulz JB, Loschmann PA, Riederer P,
p 47–112. Klockgether T. Cell death and apoptosis regulating proteins in
4. Bernheimer H, Birkmayer W, Hornykiewicz O, Jellinger K, Seitelberger F. Parkinson’s disease—a cautionary note. Acta Neuropathol 1999;97:
Brain dopamine and the syndromes of Parkinson and Huntington. Clini- 408–412.
cal, morphological and neurochemical correlations. J Neurol Sci 1973; 28. Kosel S, Egensperger R, von Eitzen U, Mehraein P, Graeber MB. On the
20:415–455. question of apoptosis in the parkinsonian substantia nigra. Acta
5. Riess O, Kuhn W, Krüger R. Genetic influence on the development of Neuropathol (Berl) 1997;93:105–108.
Parkinson’s disease. J Neurol 2000;247:69–74. 29. Jellinger KA, Stadelmann CH. The enigma of cell death in neurodegen-
6. Yahr MD, Bergman KJ. Advances in Neurology. New York: Raven Press; erative disorders. J Neural Transm Suppl 2000;60:21–36.
1986. p 317–321. 30. Banati RB, Daniel SE, Blunt SB. Glial pathology but absence of apoptotic
7. Tanner CM. The role of environmental toxins in the etiology of Parkinson’s nigral neurons in long-standing Parkinson’s disease. Mov Disord 1998;
disease. TINS 1989;12:49–54. 13:221–227.
8. Fahn S, Cohen G. The oxidant stress hypothesis in Parkinson’s disease. 31. Rajput AH, Fenton ME, Birdi S, Macaulay R, George D, Rozdilsky B, Ang
Evidence supporting it. Ann Neurol 1992;32:804–812. LC, Senthilselvan A, Hornykiewicz O. Clinical-pathological study of
9. Gerlach M, Riederer P, Youdim MBH. Molecular mechanism for levodopa complications. Mov Disord 2002;17:289–296.
neurodegeneration: synergism between reactive oxygen species, 32. Hristova AH, Koller WC. Early Parkinson’s disease: what is the best
calcium and excitotoxic amino acids. In: Battistin L, Scarlato G, Caraceni approach to treatment. Drugs Aging 2000;17:165–181.
T, Ruggieri S, editors. Advances in Neurology. Philadelphia: Lippincott- 33. Yu PH, Tipton KF. Deuterium isotope effect of phenelzine on the inhibition
Raven; 1996. p 177–194. of rat liver mitochondrial monoamine oxidase activity. Biochem Pharma-
10. Headley PM, Grillner S. Excitatory amino acid neurotoxicity and synaptic col 1989;38:4245–4251.
transmission: the evidence for a physiological function. In: Lodge D, 34. Davis DS, Tasuhara H, Boobis AR, George CF. The effects of reversible
Collingridge G, editors. The pharmacology of excitatory amino acids: A inhibitors of monoamine oxidase on tyramine deamination in dog intes-
TIPS special report. Cambridge: Elsevier; 1990. p 30–36. tine. In: Tipton KF, Dostert P, Strolin-Benedetti M, editors. Monoamine
11. De Erausquin GA, Costa E, Hanbauer I. Calcium homoestasis, free Oxidase and Disease. London: Academic Press; 1984. p 443–448.
radical formation, and trophic factor dependence mechanisms in 35. Hasan F, McCrodden JM, Kennedy NP, Tipton KF. The involvement of
Parkinson’s disease. Pharmacol Rev 1994;46:467–482. intestinal monoamine oxidase in the transport and metabolism of
12. Beal MF. Ageing, energy, and oxidative stress in neurodegenerative tyramine. J Neural Transm 1988;26(Suppl):1–9.
diseases. Ann Neurol 1995;38:357–366. 36. Blackwell B. Hypertensive crisis due to monoamine oxidase inhibitors.
13. Gerlach M, Ben-Shachar D, Riederer P, Youdim MBH. Altered brain Lancet 1963;ii:849–851.
metabolism of iron as a cause of neurodegenerative diseases? 37. Blackwell B, Marley E, Price J. Hypertensive interactions between
J Neurochem 1994;63:793–807. monoamine oxidase inhibitors and food stuffs. Br J Psychiatry 1967;113:
14. Thompson CB. Apoptosis in the pathogenesis and treatment of disease. 349–365.
Science 1995;267:1456–1462. 38. Johnston JP. Some observations upon a new inhibitor of monoamine
15. Kitada T, Asakawa S, Hattori N, Matsumine H, Yamamura Y, Minoshima oxidase in the brain tissue. Biochem Pharmacol 1968;17:1285–1297.
S, Yokochi M, Mizuno Y, Shimizu N. Mutations in the parkin gene cause 39. Parkinson Study Group. Effects of deprenyl on the progression of
autosomal recessive juvenile parkinsonism. Nature 1998;392:605– disability in early Parkinson’s disease. New Engl J Med 1989;321:1364–
608. 1371.
16. Leroy E, et al. The ubiquitin pathway in Parkinson’s disease. Nature 40. Parkinson Study Group. A controlled trial of rasagiline in early Parkinson’s
1998;392:451–452. disease: the TEMPO Study. Arch Neurol 2002;59:1937–1943.
17. Polymeropoulos MH, et al. Mutation in the alpha-synuclein gene 41. Bach AW, Lan NC, Johnson DL, Abell CW, Bembenek ME, Kwan SW,
identified in families with Parkinson’s disease. Science 1997;276:2045– Seeburg PH, Shih JC. cDNA cloning of human liver monoamine oxidase
2047. A and B: molecular basis of differences in enzymatic properties. Proc
18. Mandel S, Weinreb O, Youdim MB. Using cDNA microarray to assess Natl Acad Sci USA 1988;85:4934–4938.
Parkinson’s disease models and the effects of neuroprotective drugs. 42. Hsu YP, Weyler W, Chen S, Sims KB, Rinehart WB, Utterback MC, Powell
TIPS 2003;24:184–191. JF, Breakefield XO. Structural features of human monoamine oxidase A
19. Momose Y, Murata M, Kobayashi K, Tachikawa M, Nakabayashi Y, elucidated from cDNA and peptide sequences. J Neurochem 1988;51:
Kanazawa I, Toda T. Association studies of multiple candidate genes for 1321–1324.
Parkinson’s disease using single nucleotide polymorphisms. Ann Neurol 43. Chen ZY, et al. Structure of the human gene for monoamine oxidase type
2002;51:133–136. A. Nucleic Acids Res 1991;19:4537–4541.
20. Ceballos-Baumann AO. Functional imaging in Parkinson’s disease: acti- 44. Shih JC, Chen K, Ridd MJ. Monoamine Oxidase: From Genes to
vation studies with PET, fMRI and SPECT. J Neurol 2003;250(Suppl 1): Behavior. Annu Rev Neurosci 1999;22:197–217.
I15–I23. 45. Hall TR, Uruena G. Pharmacology and physiology of monoamine oxidase
21. Mochizuki H, Goto K, Mon H, Mizuno Y. Histochemical detection of activity in vertebrates—a comparative study. Comp Biochem Physiol B
apoptosis in Parkinson’s disease. J Neurol Sci 1996;137:120–123. 1983;76:393–397.
22. Oppenheim RW, Flavell RA, Vinsant S, Prevette D, Kuan CY, Rakic P. 46. Parkinson Study Group. Effect of lazabemide on the progression of
Programmed cell death of developing mammalian neurons after genetic disability in early Parkinson’s disease. Ann Neurol 1996;40:99–107.
deletion of caspases. J Neurosci 2001;21:4752–4760. 47. Birkmayer W, Riederer P, Youdim MB, Linauer W. The potentiation of the
23. Hartmann A, et al. Caspase-3: A vulnerability factor and final effector in anti-akinetic effect after L-dopa treatment by an inhibitor of MAO-B,
apoptotic death of dopaminergic neurons in Parkinson’s disease. Proc Deprenyl. J Neural Transm 1975;36:303–326.
Natl Acad Sci USA 2000;97:2875–2880. 48. Finberg JP, Lamensdorf I, Commissiong JW, Youdim MB. Pharmacology
24. Mogi M, Togari A, Kondo T, Mizuno Y, Komure O, Kuno S, Ichinose H, and neuroprotective properties of rasagiline. J Neural Transm 1996;48:
Nagatsu T. Caspase activities and tumor necrosis factor receptor R1 95–101.
(p55) level are elevated in the substantia nigra from parkinsonian brain. 49. Youdim MBH, Wadia A, Tatton W, Weinstock M. The anti-Parkinson
J Neural Transm 2000;107:335–341. drug rasagiline and its cholinesterase inhibitor derivatives exert

88 BioEssays 26.1
Challenges

neuroprotection unrelated to MAO inhibition in cell culture and in vivo. 71. Lai CT, Zuo DM, Yu PH. Is brain superoxide dismutase activity increased
Ann NY Acad Sci 2001;939:450–458. following chronic treatment with 1-deprenyl? J Neural Transm Suppl
50. Speiser Z, Levy R, Cohen S. Effects of N-propargyl-1-(R)aminoindan 1994;41:221–229.
(rasagiline) in models of motor and cognition disorders. J Neural Transm 72. Wu RM, Chiuech CC, Pert A, Murphy DL. Apparent antioxidant effect of
1998;Suppl 52:287–300. L-deprenyl on hydroxyl radical formation and nigral injury elicited by
51. Youdim MB, Finberg JP. Pharmacological actions of l-deprenyl (selegi- MPPþ in vivo. Eur J Pharmacol 1993;243:241–248.
line) and other selective monoamine oxidase B inhibitors. Clin Pharmacol 73. Gerlach M, Desser H, Youdim MB, Riederer P. New horizons in molecular
Ther 1994;56:725–733. mechanisms underlying Parkinson’s disease and in our understanding of
52. Chen L, He M, Sibille E, Thompson A, Sarnyai Z, Baker H, Shippenberg the neuroprotective effects of selegiline. J Neural Transm Suppl 1996;48:
T, Toth M. Adaptive changes in postsynaptic dopamine receptors 7–21.
despite unaltered dopamine dynamics in mice lacking monoamine 74. Naoi M, Maruyama W, Youdium MBH. Molecular mechanism underlying
oxidase B J Neurochem 1999;73:647–655. neuroprotection by rasagiline. J Neurochem 2002;81:4.
53. Garrick NA, Murphy DL. Species differences in the deamination of 75. Maruyama W, Yamamoto T, Kitani K, Carrillo MC, Youdim M, Naoi M.
dopamine and other substrates for monoamine oxidase in brain. Mechanism underlying anti-apoptotic activity of a (-) deprenyl-related
Psychopharmacology 1980;72:27–33. propargylamine, rasagiline. Mech Ageing Dev 2000;116:181–191.
54. Lenders JWM, et al. Specific genetic deficiencies of the A and B 76. Maruyama W, Takahashi T, Youdim M, Naoi M. The anti-Parkinson drug,
isoenzymes of monoamine oxidase are characterized by distinct neuro- rasagiline, prevents apoptotic DNA damage induced by peroxynitrite in
chemical and clinical phenotypes. J Clin Invest 1996;97:1010–1019. human dopaminergic neuroblastoma SH-SY5Y cells. J Neural Transm
55. Finnegan KT, Skratt JJ, Irwin I, DeLanney LE, Langston JW. Protection 2002;109:467–481.
against DSP-4-induced neurotoxicity by deprenyl is not related to its 77. Maruyama W, Naoi M. Neuroprotection by (-)-deprenyl and related
inhibition of MAO B. Eur J Pharmacol 1990;184:119–126. compounds. Mech Ageing Dev 1999;111:189–200.
56. Yu PH, Davis BA, Fang J, Boulton AA. Neuroprotective effects of some 78. Bowyer JF, Peterson SL. Neuronal degeneration in the forebrain
monoamine oxidase-B inhibitors against DSP-4-induced noradrenaline produced by amphetamine, methamphetamine and fenfluramine. In:
depletion in the mouse hippocampus. J Neurochem 1994;62:697–704. Lester DS, Slikker W Jr, Lazarovici P, editors. Cellular and Molecular
57. Abu-Raya S, Tabakman R, Blaugrund E, Trembovler V, Lazarovici P. Mechanisms of Toxin Action: Site-Selective Neurotoxicity. London: Taylor
Neuroprotective and neurotoxic effects of monoamine oxidase-B and Francis; 2002. p 207–232.
inhibitors and derived metabolites under ischemia in PC12 cells. Eur J 79. Mytilineou C, Radcliffe PM, Olanow CW. L-(-)-desmethylselegiline, a
Pharmacol 2002;434:109–116. metabolite of selegiline [L-(-)-deprenyl], protects mesencephalic dopa-
58. Goggi J, Theofilopoulos S, Riaz SS, Jauniaux E, Stern GM, Bradford HF. mine neurons from excitoxicity in vitro. J Neurochem 1997;68:434–
The neuronal survival effects of rasagiline and deprenyl on fetal human 436.
and rat ventral mesencephalic neurons in culture. Neuroreport 80. Oh C, Murray B, Bhattacharya N, Holland D, Tatton WG. (-)-Deprenyl
2000;11:3937–3941. alters the survival of adult murine facial motoneurons after axotomy:
59. Yoles E, Shwartz M. N-Propargyl-1 (R)-aminoindan (TVP-1012), a increases in vulnerable C57BL strain but decreases in motor neuron
putative neuroprotective agent, enhances in vitro neuronal survival after degeneration mutants. J Neurosci Res 1994;38:64–74.
glutamate toxicity. Proc Am Soc Neurosci 1995;21:562. 81. Maruyama W, Youdim MBH, Naoi M. Antiapoptotic properties of
60. Abu-Raya S, Blaugrund E, Trembovler V, Shilderman-Bloch E, Shohami rasagiline N-propargylamine-1(R)-aminoindan, and its optical (S)-iso-
E, Lazarovici P. Rasagiline, a monoamine oxidase-B inhibitor, protects mer, TV1022. Ann NY Acad Sci 2001;939:320–329.
NGF-differentiated PC12 cells against oxygen-glucose deprivation. 82. Kragten E, Lalande I, Zimmermann K, Roggo S, Schindler P, Müller D,
J Neurosci Res 1999;58:456–463. Van Oostrum J, Waldmeier P, Fürst P. Glyceraldehyde-3-phosphate
61. Speiser Z, Katzir O, Rehavi M, Zabarski T, Cohen S. Sparing by dehydrogenase, the putative target of the anti-apoptotic compounds
rasagiline (TVP-1012) of cholinergic functions and behavior in the CGP 3466 and R-(-)-deprenyl. J Biol Chem 1998;273:5821–5828.
postnatal anoxia rat. Pharmacol Biochem Behav 1998;60:387–393. 83. Binda C, Newton-Vinson P, Hubalek F, Edmondson DE, Mattevi A.
62. Speiser Z, Mayk A, Eliash S, Cohen S. Studies with rasagiline, a MAO-B Structure of human monoamine oxidase B, a drug target for the treatment
inhibitor, in experimental focal ischemia in the rat. J Neural Transm of neurological disorders. Nat Struct Biol 2002;9:22–26.
1999;106:593–606. 84. Hyslop PA, Hinshaw DB, Halsey WA Jr, Schraufstatter IU, Sauerheber
63. Huang W, Chen Y, Shohami E, Weinstock M. Neuroprotective effect of RD, Spragg RG, Jackson JH, Cochrane CG. Mechanisms of oxidant-
rasagiline, a selective monoamine oxidase-B inhibitor, against closed mediated cell injury. The glycolytic and mitochondrial pathways of ADP
head injury in the mouse. Eur J Pharmacol 1999;366:127–135. phosphorylation are major intracellular targets inactivated by hydrogen
64. Knoll J, Miklya I. Enhanced catecholaminergic and serotoninergic peroxide. J Biol Chem 1988;263:1665–1675.
activity in rat brain from weaning to sexual maturity: rationale for 85. Carlile GW, Chalmers-Redman RM, Tatton NA, Pong A, Borden KE,
prophylactic (-)deprenyl (selegiline) medication. Life Sci 1995;56:611– Tatton WG. Reduced apoptosis after nerve growth factor and serum
620. withdrawal: conversion of tetrameric glyceraldehyde-3-phosphate dehy-
65. Knoll J. The pharmacology of selegiline ((-)deprenyl). New aspects. Acta drogenase to a dimer. Mol Pharmacol 2000;57:2–12.
Neurol Scand Suppl 1989;126:83–91. 86. Fukuhara Y, Takeshima T, Kashiwaya Y, Shimoda K, Ishitani R,
66. Parkinson Study Group. Effect of tocopherol and deprenyl on the Nakashima K. GAPDH knockdown rescues mesencephalic dopaminer-
progression of disability in early Parkinson’s disease. New Engl J Med gic neurons from MPPþ-induced apoptosis. Neuroreport 2001;12: 2049–
1989;328:176–183. 2052.
67. Siderowf A, and the Parkinson Study Group. Earlier treatment with 87. Mazzola JL, Sirover MA. Alteration of intracellular structure and function
rasagiline may attenuate (UPDRS) progression of PD. Movement of glyceraldehyde-3-phosphate dehydrogenase: a common phenotype
Disorders 2001;16:981. of neurodegenerative disorders? Neurotoxicology 2002;23:603–609.
68. Kitani K, Kanai S, Ivy GO, Carrillo MC. Pharmacological modifications of 88. Magyar K, Vizi ES. Milestones in monoamine oxidase research: dis-
endogenous antioxidant enzymes with special reference to the effects of covery of (-)-deprenyl. Budapest: Medicina Publishing House Co; 2000.
deprenyl: a possible antioxidant strategy. Mech Ageing Dev 1999;111: 89. Mogi M, Togari A, Kondo T, Mizuno Y, Komure O, Kuno S, Ichinose H,
211–221. Nagatsu T. Brain-derived growth factor and nerve growth factor
69. Kitani K, Miyasaka K, Kanai S, Carrillo MC, Ivy GO. Upregulation of concentrations are decreased in the substantia nigra in Parkinson’s
antioxidant enzyme activities by deprenyl. Implications for life span disease. Neurosci Lett 1999;270:45–48.
extension. Ann N Y Acad Sci 1996;786:391–409. 90. Brundin P. GDNF treatment in Parkinson’s disease: time for controlled
70. Carrillo MC, Kanai S, Nokubo M, Kitani K. (-) deprenyl induces activities clinical trials? Brain 2002;125:2149–2151.
of both superoxide dismutase and catalase but not of glutathione 91. Yamaguchi K, Sasano A, Urakami T, Tsuji T, Kondo K. Stimulation of
peroxidase in the striatum of young male rats. Life Sci 1991;48:517–521. nerve growth factor production by pyrroloquinoline quinone and its

BioEssays 26.1 89
Challenges

derivatives in vitro and in vivo. Biosci Biotechnol Biochem 1993;57: 103. Finberg JP, Takeshima T, Johnston JM, Commissiong JW. Increased
1231–1233. survival of dopaminergic neurons by rasagiline, a monoamine oxidase B
92. Maruyama W. Neuroprotection by propargylamines in PD Suppression of inhibitor. NeuroReport 1998;9:703–707.
apoptosis and induction of prosurvival genes Neurotoxicol and Teratol 104. Roy E, Bedard PJ. Deprenyl increases survival of rat foetal nigral neurons
2002;4:675–682. in culture. NeuroReport 1993;4:1183–1186.
93. Zigova T, Snyder EY, Sanberg PR. Neural Stem Cells for Brain and Spinal 105. Xu L, Ma J, Seigel GM, Ma J. L-deprenyl, blocking apoptosis and
Cord Repair. Totowa: Humana Press; 2003. regulating gene expression in cultured retinal neurons. Biochem
94. Nagatsu T. Parkinson’s disease: changes in apoptosis-related factors Pharmacol 1999;58:1183–1190.
suggesting possible gene therapy. J Neural Transm 2002;109:731–745. 106. Maruyama W, Akao Y, Youdim MB, Naoi M. Neurotoxins induce
95. Bentue-Ferrer D, Menard G, Allain H. Monoamine oxidase B inhibitors: apoptosis in dopamine neurons: protection by N-propargylamine-1(R)-
current status and future potential. CNS drugs 1996;6:217–236. and (S)-aminoindan, rasagiline and TV1022. In: Riederer P, Calne DB,
96. Perez V, Marco JL, Fernandez-Alvarez E, Unzeta M. Kinetic studies of N- Horowski R, Mizuno Y, Olanow CW, Poewe W, Youdim MBH, editors.
allenic analogues of tryptamine as monoamine oxidase inhibitors. J Advances in Research on Neurodegeneration. Wien: Springer-Verlag;
Pharm Pharmacol 1996;48:718–722. 2000. p 171–186.
97. Zhou JP, Li J, Upadhyaya S, Eaton PE, Silverman RB. 4-substituted 107. Maruyama W, Akao Y, Youdim MB, Davis BA, Naoi M. Transfection-
cubylcarbinylamines: a new class of mechanism-based monoamine enforced Bcl-2 overexpression and an anti-Parkinson drug, rasagiline,
oxidase B inactivators. J Med Chem 1997;40:1165–1168. prevent nuclear accumulation of glyceraldehyde-3-phosphate dehydro-
98. Mytilineou C, Radcliffe PM, Olanow CW. L-(-)-Desmethylselegiline, a genase induced by an endogenous dopaminergic neurotoxin, N-methyl
metabolite of selegiline [L-(-)-Deprenyl], protects mesencephalic dopa- (R) salsolinol. J Neurochem 2001;78:727–735.
mine neurons from excitotoxicity in vitro. J Neurochem 1997;68:434–436. 108. Lahtinen H, Koistinaho J, Kauppinen R, Haapalinna A, Keinänen R,
99. Semkova I, Wolz P, Schilling M, Krieglstein J. Selegiline enhances NGF Sivenius J. Selegiline treatment after transient global ischemia in gerbils
synthesis and protects central nervous system neurons from excitotoxic enhances the survival of CA1 pyramidal cells in the hippocampus. Brain
and ischemic damage. Eur J Pharmacol 1996;315:19–30. Res 1997;757:260–267.
100. Koutsilieri E, Chen TS, Rausch WD, Riederer P. Selegiline is neuropro- 109. Knollema S, Aukema W, Hom H, Korf J, Ter Horst GJ. L-deprenyl
tective in primary brain cultures treated with 1-methyl-4-phenylpyridi- reduces brain damage in rats exposed to transient hypoxia-ischemia.
nium. Europ J Pharmacol 1996;306:181–186. Stroke 1995;26:1883–1887.
101. Mytilineou C, Leonardi EK, Radcliffe P, Heinonen EH, Han SK, Werner P, 110. Wu RM, Chen RC, Chiueh CC. Effect of MAO-B inhibitors on MPPþ
Cohen G, Olanow CW. Deprenyl and desmethylselegiline protect toxicity in vivo. Ann NY Acad Sci 2000;899:255–261.
mesencephalic neurons from toxicity induced by glutathione depletion. 111. Kupsch A, Sautter J, Gotz ME, Breithaupt W, Schwarz J, Youdim MBH,
J Pharmacol Exp Therap 1998;284:700–706. Riederer P, Gerlach M, Oertel WH. Monoamine oxidase-inhibition and
102. Tatton WG, Ju WY, Holland DP, Tai C, Kwan M. (-)-Deprenyl reduces MPTP-induced neurotoxicity in the non-human primate: comparison of
PC12 cell apoptosis by inducing new protein synthesis. J Neurochem rasagiline (TVP 1012) with selegiline. J Neural Transm 2001;108:985–
1994;63:1572–1575. 1009.

90 BioEssays 26.1

You might also like