<<

Journal of Neural Transmission https://doi.org/10.1007/s00702-018-1853-9

NEUROLOGY AND PRECLINICAL NEUROLOGICAL STUDIES - REVIEW ARTICLE

Pharmacological aspects of the neuroprotective efects of irreversible MAO‑B inhibitors, and , in Parkinson’s disease

Éva Szökő1 · Tamás Tábi1 · Peter Riederer2 · László Vécsei3,4 · Kálmán Magyar1

Received: 4 January 2018 / Accepted: 31 January 2018 © Springer-Verlag GmbH Austria, part of Springer Nature 2018

Abstract The era of MAO-B inhibitors dates back more than 50 years. It began with Kálmán Magyar’s outstanding discovery of the selective inhibitor, selegiline. This compound is still regarded as the gold standard of MAO-B inhibition, although newer drugs have also been introduced to the feld. It was revealed early on that selective, even irreversible inhibition of MAO-B is free from the severe side efect of the non-selective MAO inhibitors, the potentiation of , resulting in the so-called ‘cheese efect’. Since MAO-B is involved mainly in the degradation of , the inhibitors lack any antidepressant efect; however, they became frst-line for the therapy of Parkinson’s disease based on their dopamine-sparing activity. Extensive studies with selegiline indicated its complex pharmacological activity profle with MAO-B-independent mechanisms involved. Some of these benefcial efects, such as neuroprotective and antiapoptotic properties, were connected to its propargylamine structure. The second MAO-B inhibitor approved for the treatment of Parkinson’s disease, rasagiline also possesses this structural element and shows similar pharmacological characteristics. The preclinical studies performed with selegiline and rasagiline are summarized in this review.

Keywords MAO-B inhibition · Selegiline · Rasagiline · Neuroprotection

Introduction inhibitor in the 1960s (Knoll et al. 1965). The R-(−)-isomer of deprenyl, later called selegiline, was identifed as the The inhibitors of the catecholamine-degrading , more potent MAO inhibitor (Magyar et al. 1967). In 1968, (MAO), have long established thera- Johnston discovered that the enzyme is heterogeneous and peutic use. Deprenyl was frst synthesized as a psychic ener- the isoform inhibited more potently by clorgyline was named gizer (Varga and Tringer 1967), tested for antidepressant MAO-A (Johnston 1968). Magyar and Knoll subsequently efects and later on characterized as an irreversible MAO found that selegiline is also a selective inhibitor, preferen- tially afecting the other isoform called MAO-B (Knoll and Magyar 1972). Because of diferent substrate specifcity and In memory of Professor Kálmán Magyar (1933–2017) tissue distribution of the two isoenzymes, their physiologi- Kálmán Magyar : Deceased. cal roles difer considerably. The well-known antidepressant efect of the previously used non-selective MAO inhibitors * László Vécsei was found to be the consequence of blocking MAO-A activ- [email protected]‑szeged.hu ity. The irreversible inhibition of MAO-A, however, is also 1 Department of Pharmacodynamics, Semmelweis University, responsible for their severe adverse efect, the life-threaten- Nagyvárad tér 4, Budapest 1089, Hungary ing hypertensive crisis after the consumption of tyramine- 2 Center of Mental Health, Department of Psychiatry, rich meals. This was named ‘cheese efect’ because of the Psychosomatics and Psychotherapy, University Hospital high tyramine content of cheese products. Selegiline, in its Würzburg, Magarete‑Höppel‑Platz 1, 97080 Würzburg, selective MAO-B inhibitory dose, is free from this danger- Germany ous side efect (Knoll et al. 1968) but also lacks antidepres- 3 Department of Neurology, University of Szeged, sant activity. As it preferentially reduces the degradation Semmelweis u. 6, Szeged 6725, Hungary of dopamine in the central nervous system (Riederer et al. 4 MTA-SZTE Neuroscience Research Group, Semmelweis 1978), it gained importance in the treatment of Parkinson’s u. 6, Szeged 6725, Hungary

Vol.:(0123456789)1 3 É. Szökő et al. disease from 1975 (Birkmayer et al. 1975, 1977; Parkinson In following studies, the efcacy of selegiline against the Study Group 1993). active toxin ­MPP+ was analyzed in various experimental The distribution of MAO isoenzymes in the brain was designs. First, Mytilineou and Cohen reported that selegiline extensively studied in post-mortem tissues by Riederer and pretreatment prevented the dopamine depletion induced by co-workers. MAO-B was shown predominantly in the glia ­MPP+ in rat embryonic midbrain neuronal culture (Mytili- (Konradi et al. 1989) and its activity was raised in aged neou and Cohen 1985). On the other hand, this efect was patients (Kornhuber et al. 1989). In post-mortem brain stud- not confrmed in the study by Vaglini et al. (1996). The ies, increased dopamine (Riederer and Youdim 1986), and results of the in vivo animal experiments were also con- especially phenylethylamine levels, (Riederer et al. 1984), troversial; failure, partial efect, and prevention of deple- were found in the nigrostriatal system of selegiline-treated tion by selegiline were reported alike (Bradbury et al. 1985; parkinsonian patients. Based on these fndings, it was sug- Mihatsch et al. 1988; Vizuete et al. 1993). In these studies, gested that the catecholamine-releasing efect of the elevated the ­MPP+-induced toxicity was followed by striatal dopa- phenylethylamine may contribute to the dopamine-sparing mine depletion, but this did not necessarily correspond to activity of selegiline (Reynolds et al. 1978; Youdim and the cell loss. Riederer 1993). MPP+ was identifed as a mitochondrial toxin in dopa- Since it was found that selegiline was able to reduce oxi- minergic neurons (Nicklas et al. 1985; Heikkila et al. 1985). dative stress (Cohen and Spina 1989) and prevent the dopa- It inhibits the activity of Complex I of the electron transport mine-depleting efect of the neurotoxin 1-methyl-4-phenyl- chain (Nicklas et al. 1987) which results in compromised 1,2,3,6-tetrahydropyridine (MPTP) (Langston et al. 1984; energy production (Mizuno et al. 1987) and increased oxi- Cohen et al. 1984), further studies were initiated to clarify dative stress (Cleeter et al. 1992). Similar alterations can its pharmacological properties. The results of these experi- be induced by the decreased Complex I expression found ments indicated possible MAO-B inhibition-independent in post-mortem Parkinsonian brain (Mizuno et al. 1989; neuroprotective, antiapoptotic, neurorescue activities, and Schapira et al. 1989; Reichmann and Riederer 1989) along the importance of the propargylamine moiety in these with its increased iron content (Sian-Hülsmann et al. 2011) efects. About 30 years later, rasagiline, another MAO-B causing signifcant oxidative damage. An additional source inhibitor with propargylamine structure, was introduced into of enhanced oxygen free radical generation is the auto-oxi- the therapy of Parkinson’s disease. The pharmacological dation of the enormous amount of dopamine released after studies aiming to understand the neuroprotective, antiapop- toxin-induced cell damage. The suggested components of totic and neurorescue efects of these two drugs are reviewed the protective efect of MAO-B inhibitors against MPTP and discussed in this paper. toxicity are summarized in Fig. 1. To obtain further information about the supposed protec- tive efect of selegiline, the involvement of its suggested properties (Cohen and Spina 1989; Gotz et al. Protective efect of selegiline 1995) was studied. Although inhibition of MAO-B itself against neurotoxins reduces hydrogen peroxide production, other components of its antioxidant activity were also hypothesized. As early The dopaminergic neurotoxin, MPTP, was discovered acci- as 1989 Knoll reported that chronic selegiline treatment of dentally when rapid onset and severe Parkinson’s syndrome aged rats enhanced the activity of superoxide dismutase developed in a drug addict using an in-house synthesized in the striatum (Knoll 1989). This was later confrmed by analogue. Later on, the nigrostriatal damage Carillo et al. in young animals, as well. Furthermore, in induced by the compound was also confrmed (Davis et al. addition to superoxide dismutase, increased catalase activ- 1979). It was soon found that MAO-B inhibitors, ity was also observed (Carrillo et al. 1991). In another study, and selegiline, prevented MPTP toxicity, suggesting the increased antioxidant enzyme activity after selegiline treat- involvement of MAO-B in the conversion of pro-toxin ment could only be measured in the striatum of aged rats. MPTP to its active form, 1-methyl-4-phenyl-pyridinium Besides the antioxidant , the level of (MPP­ +) (Langston et al. 1984; Cohen et al. 1984). Subse- was found to be elevated. No efect was detected in the cor- quently, it was also shown that MPP­ + generated by MAO-B tex or hippocampus or any brain regions of young animals can enter into and damage dopaminergic neurons and is (Takahata et al. 2006). In in vitro cell culture experiments, responsible for MPTP toxicity. Besides MAO inhibitors, it was shown that selegiline treatment, similar to nerve dopamine uptake blockers also provided protection, indi- growth factor (NGF), dose-dependently induced superox- cating the prominent role of this transporter in the selec- ide dismutase mRNA expression. Selegiline also potentiated tive accumulation of the toxin (Fuller and Hemrick-Luecke the efect of NGF, which suggests its action on superoxide 1985). dismutase mRNA induction is NGF-independent (Li et al.

1 3 Pharmacological aspects of the neuroprotective efects of irreversible MAO‑B inhibitors,…

Fig. 1 Targets of selegiline and rasagiline in MPTP toxicity

1998). This is in line with the fndings of Thifault et al., inhibitor, MDL 72974, failed to prevent DSP-4 toxicity, indi- who found selegiline inhibited oxygen consumption of iso- cating another mechanism in the protective efect of sele- lated mitochondria and suggested that this impairment can giline (Finnegan et al. 1990). As DSP-4 accumulates in the induce an adaptive increase in superoxide dismutase activity noradrenergic terminals by uptake transporters, their inhibi- (Thifault et al. 1997). However, the antioxidant property tion by selegiline and/or its metabolites was also proposed due to the induction of antioxidant enzymes develops slowly (Magyar et al. 1996, 1998; Magyar and Haberle 1999). How- following chronic treatment and thus cannot fully explain ever, the metabolites, R-(−)- and R-(−)- the rapid neuroprotective efect of selegiline. Its acute efect amphetamine, possessing stronger uptake inhibitory activity, on ­MPP+-induced hydroxyl radical formation was reported showed weaker protection compared to selegiline against without signifcant infuence on dopamine overfow, sug- DSP-4 induced noradrenaline depletion in the hippocam- gesting the crucial role of direct antioxidant activity (Wu pus. Uptake inhibition thus cannot be the sole mechanism et al. 1993). This was supported by the similar efect of responsible for the protective efect (Haberle et al. 2001). other hydroxyl radical scavengers and (Wu et al. Similarly, selegiline was also found to be protective against 1996; Khaldy et al. 2000). Reduced lactate release, a marker 6-hydroxy-dopamine and 3,4-methylenedioxy-methamphet- of hypoxia, from ­MPP+-damaged neurons treated with sele- , other toxins causing biogenic amine depletion, but giline is also indicative of its protective efect (Matsubara the exact mechanism of action has not yet been clarifed et al. 2001). Selegiline could also protect nigral neurons (Salonen et al. 1996; Sprague and Nichols 1995). against other types of oxidative stress-inducing agents, like Although these toxins are very useful in modelling some rotenone and iron overload (Saravanan et al. 2006; Budni aspects of Parkinson’s disease, their efect is far from the et al. 2007; de Lima et al. 2005), further supporting the pathophysiology of the disorder. Later, other neuronal importance of its antioxidant property. insults, probably more closely related to the pathomecha- Selegiline was found to provide protection against other nism of neurodegenerative disorders, were used to study the monoamine-depleting neurotoxins, as well. DSP-4 (N-(2- protective efect of selegiline. chloroethyl)-N-ethyl-2-bromobenzylamine hydrochloride) is selectively accumulated in noradrenergic nerve terminals and thus specifcally reduces noradrenaline levels in several Protective efect of selegiline in models brain regions. Furthermore, it also decreases dopamine-beta- of neuronal degeneration hydroxylase activity in the brain, consistent with the degen- eration of noradrenergic neurons (Ross 1976). The protective NMDA receptor-mediated excitotoxicity is one of the mech- efect of the MAO-B inhibitor selegiline and the non-selec- anisms participating in neuronal death. Selegiline and its tive inhibitor pargyline against DSP-4-induced noradrena- metabolite desmethylselegiline prevented NMDA-induced line depletion was reported, while the MAO-A inhibitor neurotoxicity in cultured dopaminergic mesencephalic neu- clorgyline was found inefective (Gibson 1987). This sug- rons. Desmethylselegiline was actually found to be more gested the role of MAO-B inhibition in protection. Later, potent in this test than its parent compound, suggesting however, the structurally unrelated and selective MAO-B the contribution of this metabolite to the neuroprotective

1 3 É. Szökő et al. efect of selegiline (Mytilineou et al. 1997a, b). Protection reduced cell loss to about 15% (Tatton and Greenwood against NMDA toxicity was also demonstrated in hippocam- 1991). The rescue efect of selegiline was also observed in pal slices, and at the same time, it was also shown that the primary mesencephalic neuronal and glial co-culture. Sele- drug did not interfere with the NMDA-dependent long-term giline was administered 24 h after ­MPP+ insult and increased potentiation process (Niittykoski et al. 2003). In an in vivo the dopamine level, TH positive cell and astrocyte count in experiment, retinal damage was induced by intravitreal the cultures (Koutsilieri et al. 1994, 1996). Rescue efect was NMDA injection. Parenteral, but not local, selegiline admin- shown on non-dopaminergic neurons after axotomy of facial istration was found to be protective, while local desmethylse- motor neurons as well. The authors suggested that selegiline legiline was also efective. This fnding further indicates the could compensate the loss of target-derived trophic support possible contribution of the metabolite desmethylselegiline (Salo and Tatton 1992). In their further detailed study on to the neuroprotective efect (Takahata et al. 2003). mutant mice with inherited motor neuron degeneration, sele- It was also shown that selegiline can protect mitochondria giline failed to show protection (Oh et al. 1994). The neuro- against pro-oxidant-induced damage. It prevented swelling, rescue efect was also observed in a rat model of traumatic collapse of membrane potential and cytochrome c release. brain injury, where selegiline improved cognitive functions Direct interaction and inhibition of some components of the although motor functions remained unchanged. Consistent mitochondrial permeability transition pore were proposed with the symptomatic efect, the injury-induced loss of DBH (De Marchi et al. 2003). positive cells in the hippocampus was reduced (Zhu et al. Protein aggregation and the formation of deposits are 2000). The kainite-induced hippocampal cell loss and the hallmarks of several neurodegenerative disorders. In Par- consequent behavioral changes were also afected by sele- kinson’s disease, α-synuclein is the main component of giline treatment. As the improvement remained after cessa- the characteristic Lewy bodies. In an in vitro experiment, tion of selegiline dosing, the neurorescue rather than symp- selegiline was shown to modify the aggregation process tomatic efect is confrmed (Gelowitz and Paterson 1999). resulting in the formation of large non-toxic aggregates The infuence of selegiline on trophic factor production (Braga et al. 2011). The structure of selegiline-bound alpha- was extensively examined. One of the frst reports showed synuclein is probably more compact. A similar efect of its that subchronic selegiline treatment potentiated the lesion- metabolite, methamphetamine was also reported (Kakish induced production of basic fbroblast growth factor (bFGF) et al. 2015). In a mouse study, selegiline reversed the cogni- and glial fbrillary acidic protein (GFAP) in rat neostriatum tive defcit induced by amyloid beta-peptide in both acute (Biagini et al. 1994). Potentiation of cAMP-induced bFGF and subchronic dosing schedules (Pazini et al. 2013). mRNA elevation in rat cortical astrocytes was also shown Ischemia–reperfusion is a well-established model of neu- (Riva et al. 1997). The increased production of other neu- ronal injury and degeneration that is most closely related to rotrophic factors; NGF, brain-derived neurotrophic factor brain infarction in stroke. Chronic selegiline pretreatment (BDNF), and glia-derived neurotrophic factor (GDNF), in effectively reduced the infarct size after transient brain cultured mouse astrocytes 24 h after high-dose selegiline ischemia (Knollema et al. 1995). Besides the infarct size, and desmethylselegiline treatment was also demonstrated the markers of apoptosis, cleaved caspase and DNA frag- (Mizuta et al. 2000). In addition to increasing the release of mentation, were also signifcantly decreased (Unal et al. neurotrophic factors from astrocytes, its possible efect on 2001). Ischemia-induced lipid peroxidation and memory activated microglia-related neuronal damage was also pro- defcit were also alleviated (Maia et al. 2004). Combina- posed and studied in a model system. Neuroblastoma cells tion treatment with selegiline and Ginkgo biloba extract was were treated with conditioned media of activated monocytic more efective in preventing ischemia-induced neuronal loss THP-1 cells. Selegiline treatment of the monocytes, but not (Kwon et al. 2004). However, pretreatment applied shortly the neuroblasts, dose-dependently prevented the damage of before or after global forebrain ischemia was inefective in the latter. Other examined MAO inhibitors were inefective, preventing cell death (Ballabriga et al. 1997). confrming the suggested MAO-independent mechanism (Klegeris and McGeer 2000). Selegiline-induced enhanced NGF production and the consequent protection against exci- Neurorescue activity of selegiline totoxic and ischemic damage of neurons were reported in a thorough study. In the in vitro cell culture experiment, Selegiline studies gained a new impetus from the reports increased NGF production was demonstrated in response to of Tatton and co-workers that it efectively rescued dying a rather low (10 pM–1 nM) selegiline concentration. Moreo- neurons in delayed administration protocols. MPTP admin- ver, increased production of NGF was also shown in the istration to mice induced about 40% decrease in the - cerebral cortex of rats after intraperitoneal administration hydroxylase positive cell count in the substantia nigra over of selegiline accompanying the protection against ischemic 20 days. Selegiline treatment started 72 h after the toxin insult (Semkova et al. 1996).

1 3 Pharmacological aspects of the neuroprotective efects of irreversible MAO‑B inhibitors,…

Furthermore, in cultured dopaminergic neurons, it was these model cells (Suuronen et al. 2000). Contrary to this also shown that neurite length was similarly increased by fnding, selegiline in submicromolar concentration prevented selegiline and BDNF, although selegiline increased the apoptotic cell death induced by either serum deprivation average neuritic branch length, while the formation of new or hypoxia in cultured retinal neurons (Xu et al. 1999). In branches was increased by BDNF (Kontkanen and Castren another study, a similar low selegiline dose could delay 1999). Consistent with its neurotrophic factor inducing and/ the serum deprivation evoked excessive apoptosis in two or mimicking efect, selegiline induced the diferentiation of cell lines of neuroectodermal origin by 2 days, various stem cells into neuron phenotype. In these in vitro while its enantiomer pair, S-(+)-deprenyl was inefective. experiments, selegiline was typically found efective in a In this study high, millimolar, doses of selegiline and des- low concentration (around 10­ −8 M) that cannot consider- methylselegiline were shown to increase apoptotic cell death ably inhibit MAO-B activity. Increased neurotrophic factor even without serum withdrawal (Szende et al. 2000). These expression and diferentiation to neurons were demonstrated findings confirmed the bell-shaped dose dependence of in embryonic stem cells (Esmaeili et al. 2006), bone-marrow the antiapoptotic efect, indicating the importance of care- stromal cells (Ghorbanian et al. 2010), embryonal carci- ful experimental design and reconsideration of the dosing noma stem cells (Bakhshalizadeh et al. 2011), and neural schedule of selegiline (Magyar et al. 2004; Magyar 2011). stem cells (Hassanzadeh et al. 2015) following selegiline The antiapoptotic efect of selegiline and related prop- treatment. argyl compounds was also demonstrated against various neurotoxins in in vitro cell culture experiments. It partially protected the MPP­ +-treated SK-N-SH neuroblastoma cells Antiapoptotic activity of selegiline with attenuation of toxin-induced alterations of several apop- tosis markers, including cytochrome c release and caspase After revealing that selegiline can induce the synthesis of 3 activation (Sharma et al. 2003; Chetsawang et al. 2008). neurotrophic factors and antioxidant enzymes, it was sug- In SH-SY5Y neuroblastoma cells, selegiline pretreatment gested that its neuroprotective-neurorescue efect is based protected the cells against the apoptotic efect of the hypoth- on a complex action on the regulation of cell survival esized endogenous toxin, N-methyl-R-salsolinol detected by mechanisms (Tatton and Chalmers-Redman 1996; Magyar a reduction in DNA fragmentation. In addition to the toxin, et al. 2006). In diferentiated PC12 cells, selegiline and its apoptosis induced by reactive oxygen and nitrogen species metabolite desmethylselegiline modifed the serum and NGF was also studied (Naoi et al. 2000). It is worth mention- withdrawal-induced expression of several genes. In addition ing that in these experiments on neuroblastoma cells, the to the previously reported increase in antioxidant enzymes, efective selegiline concentration was much higher, in the superoxide dismutase and glutathione peroxidase, altered micromolar range. This discrepancy might be explained by expression of other apoptosis-related proteins was also the possibly difering nature of diferentiated and undifer- observed. It prevented the elevation of c-JUN and GAPDH entiated cells. and the reduction of BCL-2 levels, as well as the mitochon- In recent studies, the apoptosis-preventing efect of sele- drial translocation of BAX. All these changes may result in giline was also reported in various animal experiments. an antiapoptotic efect by maintaining mitochondrial mem- Pretreatment with selegiline attenuated the 3-nitropropi- brane potential and preventing the opening of permeability onic acid-induced striatal and cortical apoptosis demon- transition pores (Tatton and Chalmers-Redman 1996). On strated by reduced caspase 3 activation and altered BCL-2/ cerebellar granule cells, selegiline and another propargyl BAX ratio. Furthermore, the toxin-induced behavioral compound prevented the impaired mitochondrial function changes were also improved (Wahdan et al. 2017). Admin- related to apoptosis without afecting low potassium-induced istering selegiline following MPTP lesion in mice induced apoptotic cell death. These fndings further confrmed that an antiapoptotic change in BCL-2 and BAX expression selegiline selectively inhibits the mitochondrial route of besides increased trophic factor production. At the same apoptosis (Paterson et al. 1998). The efect of selegiline and time, the drug rescued the toxin-induced gait defcit (Zhao the related propargylamines was dose dependent; the maxi- et al. 2013). When administered after focal brain ischemia, mal antiapoptotic efect developed at a concentration level selegiline reduced the infarct size, induced the antiapop- of 10­ −9 M. Further concentration increase was accompa- totic genes, and promoted NOTCH-JAGGED signaling in nied by a decline in the efect (Tatton et al. 2002). This low the astrocytes. It was accompanied by reduced peri-infarct concentration of selegiline was previously found efective edema, likely due to preserving brain microcirculation (Nar- in preventing okadaic acid-induced apoptotic response in dai et al. 2015). The proposed mechanisms contributing to cultured hippocampal and cerebellar granule cells, as well the antiapoptotic activity of selegiline are shown in Fig. 2. as in Neuro-2a neuroblastoma cells. However, selegiline did The cytoprotective, antioxidant, and antiapoptotic efects not provide protective activity against serum withdrawal in of selegiline are not restricted to damaged neuronal cells.

1 3 É. Szökő et al.

Fig. 2 Putative targets of selegiline and rasagiline involved in their efects independent of MAO-B enzyme inhibition

In cultured airway epithelial cells, cigarette smoke extract- also improved the sexual activity (Knoll 1988; Knoll et al. induced oxidative stress and inflammation were dose- 1989) and learning performance of aged animals (Cara- dependently reversed by selegiline in the ­10−8–10−6 M georgiou et al. 2003). As potential mechanisms responsible range. The authors suggested that the antioxidant property of for longevity induction, its antioxidant, cytoprotective, and the drug is also responsible for its anti-infammatory efect neuronal activity stimulating, so-called enhancer efect on through preventing the activation of infammation-related monoaminergic transmissions, are suggested (Knoll and transcription factors Nrf2 and NF-κB (Cui et al. 2017). Miklya 1994; Kaur et al. 2003; Singh et al. 2012; Kitani Chronic low-dose selegiline was shown to increase the total et al. 2002). scavenger capacity of plasma and prevent the fat-rich diet- During the studies on the neuroprotective efect of sele- induced increase in the fat content (Bekesi et al. 2012). giline, it emerged that the pharmacophore includes the pro- Renal cells could also be rescued from hypoxia-induced pargylamine moiety. This was supported by the observed apoptotic death in an animal study (Toronyi et al. 2002). activity of its desmethyl metabolite. However, desmethyl- In a rabbit model of cardiac failure, 8 weeks of selegiline selegiline was shown to be a minor metabolite, representing treatment improved left ventricular function by rescuing less than 1% of the administered dose (Szökő et al. 2004b). myocytes from apoptosis (Qin et al. 2003). Other aromatic and aliphatic propargyl compounds with or The benefcial efect of selegiline on aging has also without MAO inhibitory activity were also examined. Their been reported in several papers since the frst one pub- efects in in vitro and in vivo antiapoptotic and neuroprotec- lished by Birkmayer et al. (1985). Chronic treatment with tive tests were similar to those of selegiline (Paterson et al. selegiline considerably increased the lifespan of rats and 1998; Waldmeier et al. 2000a, b).

1 3 Pharmacological aspects of the neuroprotective efects of irreversible MAO‑B inhibitors,…

Neuroprotective and neurorescue efects overexpressing alpha-synuclein, the use of paraquat-induced of rasagiline signifcant oxidative stress and caspase 3 activation that was prevented by rasagiline (Chau et al. 2010). In another Besides selegiline, several propargyl compounds were syn- in vitro model, rasagiline attenuated microglia activation- thesized and studied in the 1970s for their MAO inhibitory related oxidative stress and pro-inflammatory cytokine efect. Among these, an aminoindane structure, J-508, was release, features that may contribute to its neuroprotective found to be about ten times more potent an inhibitor of properties (Trudler et al. 2014). MAO-B, although its selectivity for MAO-B over MAO-A The mechanism of neuroprotective-antiapoptotic efect of inhibition was not better than that of selegiline (Magyar rasagiline was extensively studied in cell culture experiments et al. 1979; Magyar 1994). The desmethyl derivative of this mainly using SH-SY5Y dopaminergic neuroblastoma cells. compound, rasagiline, was developed later as the second Induction of neurotrophic factors, mainly GDNF (Maruyama MAO-B inhibitor approved for the treatment of Parkinson’s et al. 2002a, 2004) and antiapoptotic BCL protein family disease. Its neuroprotective and neurorescue efects were members (Akao et al. 2002b; Inaba-Hasegawa et al. 2012) also extensively studied. were reported. Probably, these contribute to protection of Initially, in vitro and in vivo tests, similar to those previ- mitochondrial integrity and prevention of mitochondrial per- ously done with selegiline, were performed with rasagiline meability transition pore opening which were also demon- to demonstrate its neuroprotective, antioxidant, antiapop- strated (Maruyama et al. 2000b, 2002b; Wu et al. 2016). The totic, and neurorescue efects. Similar to selegiline, it was importance of mitochondrial protection in the antiapoptotic found efective against various neurotoxins. It increased the efect was also confrmed in cerebellar granule cells, where survival of dopaminergic neurons in the substantia nigra and rasagiline prevented cytosine beta-d-arabinofuranoside, the stereotypical behavior induced by 6-hydroxydopamine l-buthionine-(S,R)-sulfoximine, or glutamate but not low (Blandini et al. 2004). In in vitro experiments on SH-SY5Y potassium or serum deprivation-induced apoptosis (Bonneh- cells, the inhibition of toxin-induced mitochondrial impair- Barkay et al. 2005). ment-related apoptosis was demonstrated as a mechanism Prevention of mitochondrial depolarization and the open- of the protective efect. Similar results were reported when ing of permeability transition pores by rasagiline were also peroxynitrite donating SIN-1 or N-methyl-(R)-salsolinol reported using isolated mitochondria suggesting a direct were used to induce neuronal damage (Maruyama et al. mitochondrial target (Akao et al. 2002a). Prevention of 2000a, 2001b). Rasagiline showed neurorescue activity nuclear translocation of GAPDH was also demonstrated and when administered chronically to post-MPTP-lesioned mice. the enzyme was suggested as another possible target for pro- It increased dopaminergic cell survival in the midbrain by pargylamine compounds (Maruyama et al. 2001a; Ou et al. upregulation of the tyrosine kinase receptor (Trk)–phos- 2009; Waldmeier et al. 2000b). Chronic rasagiline treatment phatidylinositol 3 (PI3) kinase–Akt pathway (Weinreb et al. was shown to increase the expression and activation of pro- 2006; Sagi et al. 2007). It also reduced the degeneration of tein kinase C and, at the same time, to benefcially afect neurons in the paraventricular nucleus of the hypothalamus amyloid processing (Bar-Am et al. 2004). In a cell culture in spontaneously hypertensive rats in a dose-dependent man- study, this efect was shown dependent on alpha-secretase ner. This efect was accompanied by a reduction in blood activity and protein kinase C and ERK kinase signaling. A pressure. The antihypertensive medications, captopril and similar efect of the non-MAO inhibitor S-enantiomer of , were inefective in preventing neurodegenera- rasagiline was also demonstrated, indicating that the protec- tion, suggesting direct neuroprotection by rasagiline as a tion is unrelated to MAO-B inhibition but dependent on the primary efect (Eliash et al. 2005). propargylamine structure (Yogev-Falach et al. 2003). The induction of catalase and superoxide dismutase (anti- oxidant enzymes) activities by chronic rasagiline treatment in substantia nigra and striatum, but not in hippocampus, Safnamide, the reversible MAO‑B inhibitor was demonstrated. Interestingly, the activity of the enzymes also increased in peripheral tissues, , and heart (Car- Recently, a third MAO-B inhibitor, safnamide was also rillo et al. 2000). Similar to selegiline, it was proven efective introduced into the therapy. It has a different chemical against ischemic insults. In NGF-diferentiated PC12 cells, it structure and pharmacological properties compared to sele- protected against oxygen–glucose deprivation-induced cell giline and rasagiline. In isolated rat brain mitochondria, it death (Abu-Raya et al. 1999). In an in vivo animal study, it potently and reversibly inhibits MAO-B with an IC50 value reduced the infarct size and improved the neurological and of 98 nM, while its potency on MAO-A is about 5000 times cognitive functions when administered after middle cerebral lower (Caccia et al. 2006). In line with these in vitro results, artery occlusion (Speiser et al. 2007). In a cell-culture model it provides complete MAO-B inhibition in 0.6 mg/kg single dose without afecting MAO-A activity in humans (Marzo

1 3 É. Szökő et al. et al. 2004). Besides MAO-B inhibition, it was found to survival of tyrosine-hydroxylase positive neurons was selec- block voltage-dependent sodium and N-type calcium chan- tively improved by both MAO-B inhibitors at the relatively nels, although in much higher concentrations. In in vitro high concentration of 1–10 µM. The MAO-A inhibitor experiments, the diference between IC50 values of chan- clorgyline and R-(−)-methamphetamine, had no efect on nels blocking and MAO-B inhibition was 2–3 orders of cell viability in this concentration range. The survival of magnitude. As a consequence of its efect on ion channels, GABAergic neurons was not afected by any examined com- it also inhibits the release of glutamate (Caccia et al. 2006; pounds (Finberg et al. 1998; Goggi et al. 2000). Rasagiline Fariello 2007). and selegiline similarly inhibited Ca­ 2+ efux from mito- Neuroprotection by was also studied. It chondria after opening of the mitochondrial permeability prevented the MPTP-induced toxicity consistent with its transition pores and attenuated superoxide generation (Wu MAO-B inhibitory efect and its weak dopamine uptake et al. 2015). Using hippocampal slices, it was observed that inhibitor activity (Fariello 2007) may also contribute. Four glutamate-mediated excitotoxicity was attenuated by both hours after MPTP insult, it failed to afect dopamine deple- rasagiline and selegiline; however, more detailed analysis tion; however, some sparing efect on dopaminergic neu- revealed diferent mechanisms, i.e., modulation of diferent rons was observed. In pretreatment, it also alleviated neu- glutamate receptor responses (Dimpfel and Hofmann 2011). ron loss induced by kainate. The neurodegeneration induced In an in vivo study of focal cerebral ischemia, rasagiline in by ischemia was also reduced by both pre- and post-insult 3 mg/kg dose, but not in 1 mg/kg, accelerated the sympto- treatment (Caccia et al. 2006). These protective efects were matic recovery, while the fnal neurological scores were not reported at high safnamide doses, indicating the probable improved. At the same time, it reduced the infarct size. Sele- role of its channel blocking property and the consequential giline in the same dose did not show these efects. Higher inhibition of glutamate release. Clinical properties of safna- doses were not examined; thus, the inefectiveness may have mide along with those of the irreversible MAO-B inhibitors been caused by the well-known potency diference between were recently reviewed (Dezsi and Vecsei 2017). the two drugs (Speiser et al. 1999). In another in vivo model There is continuous interest in studying reversible of neurodegeneration using ubiquitin–proteasome system MAO-B inhibitors. In addition to safnamide, several other inhibitor lactacystin, selegiline and rasagiline pretreatment natural and synthetic compounds were reported as reversible were efective alike. When treatment started after the insult, MAO-B inhibitors and examined in preclinical and early selegiline was found less efective (Zhu et al. 2008). clinical studies. Sembragiline, a compound structurally Some of the studies also aimed to evaluate the infuence related to safnamide, is a highly selective MAO-B inhibi- of their metabolites on the protective efect. Some selegiline tor with long lasting efect (Borroni et al. 2017). Its pos- metabolites keep the propargylamine structure. The neuro- sible benefcial efect in moderate Alzheimer’s disease has protective efect of desmethylselegiline was observed in recently been evaluated in a phase 2 without various tests, including demonstration of its benefcial efect conclusive results (Nave et al. 2017). In preclinical stud- against NMDA toxicity (Mytilineou et al. 1997b; Takahata ies, not only further biaryl compounds (Yeon et al. 2018), et al. 2003) and in inducing the production of neurotrophic but new chemical structures, like furanochalcones (Suresh factors (Mizuta et al. 2000) and antiapoptotic proteins (Tat- et al. 2018) are also being investigated. Among natural ton and Chalmers-Redman 1996). Its N-oxide metabolite compounds, chrysin was reported having MAO-B inhibi- was only identifed later (Katagi et al. 2001; Tabi et al. tory activity, besides several other pharmacological efects 2003; Szökő et al. 2004b) because of shortcomings in the resembling to those of selegiline and rasagiline (Guo et al. previously used analytical method, gas chromatography. The 2016). neuroprotective efect of selegiline-N-oxide against DSP-4 induced noradrenaline depletion was found to be compara- ble with that of the parent compound (Szökő et al. 2004a). Comparison of the efects of selegiline The main non-propargyl metabolites of selegiline are and rasagiline R-(−)-methamphetamine and R-(−)-amphetamine (Szoko et al. 1999; Tabi et al. 2003; Shin 1997), while rasagiline is The majority of the reported efects of rasagiline are similar converted to 1-R-aminoindan (Siddiqui and Plosker 2005). to those previously published with selegiline. Some direct Since S-enantiomers of amphetamines have considerable comparative studies were performed, as well. Prevention psychomotorstimulatory and neurotoxic efects, it was sug- of MPTP-induced neurotoxicity was compared in monkeys gested that the metabolites of selegiline may counteract its and their efects were found similar in terms of biochemi- neuroprotective efect. The R-amphetamines, however, are cal, histological, and behavioral means. As MAO-B inhibi- much weaker releasers of catecholamines and are formed tion signifcantly contributes to the reduced MPTP toxicity, in a rather low concentration during selegiline this result could be expected (Kupsch et al. 2001). In vitro (Magyar and Tothfalusi 1984; Magyar et al. 2004). In an

1 3 Pharmacological aspects of the neuroprotective efects of irreversible MAO‑B inhibitors,… in vitro cell culture study, a high concentration of meth- Conclusions amphetamine did not prevent against serum and NGF withdrawal-induced death of PC12 cells and reduced the Although several pharmacological experimental data in protection by selegiline or rasagiline (Bar Am et al. 2004). support of the neuroprotective efect of both irreversible However, during therapeutic use of selegiline, much lower MAO-B inhibitors were published, the results of in vitro concentrations of amphetamines are formed (Heinonen et al. experiments could hardly be translated to a clinical efect. 1989), so the counteraction of the selegiline efect is not Even the preclinical data are inconsistent regarding the very probable. Data on the efect of 1-R-aminoindane are efective concentrations of the drugs and the suggested role controversial as well. It was found efective in a rather high of their metabolites, which may be the sources of some concentration of 1 μM in a model of serum and NGF with- discrepancies between the fndings of in vitro and in vivo drawal-induced PC12 cell death (Bar Am et al. 2004) and studies. Some of the preclinical results suggest diferences dexamethasone-induced apoptosis of SH-SY5Y neuroblas- between the protective efect of selegiline and rasagiline. toma and 1242-MG glioblastoma cells (Tazik et al. 2009). However, the recent meta-analysis of randomized clinical However, in another test using cerebellar granule cells, it trials (Marconi and Zwingers 2014), drug utilization analysis was not found efective against various cell insults in the of delaying levodopa prescription in patients on selegiline or wide concentration range studied (Bonneh-Barkay et al. rasagiline treatment (Peretz et al. 2016), and a head-to-head 2005). Based on these results, the importance of the propar- 3-year retrospective case–control study (Cereda et al. 2017) gylamine structure in the neuroprotective efect of MAO-B concluded similar efcacy and neuroprotective potential of inhibitors is rather accepted. This is further confrmed by the two drugs. the results that N-propargylamine itself showed neuroprotec- tive activity against serum withdrawal-induced apoptosis of Acknowledgements Supported by GINOP 2.3.2-15-2016- PC12 cells (Weinreb et al. 2005). 00034.2017.-1.2.1-NKP-2017-00002. The linguistic correction was made by Jennifer Tusz (Edmonton, Canada).

Evaluation of neuroprotective efect in clinical studies References

There were several clinical trials designed to assess the dis- Abu-Raya S, Blaugrund E, Trembovler V, Shilderman-Bloch E, Sho- ease-modifying, neuroprotective efect of the MAO-B inhib- hami E, Lazarovici P (1999) Rasagiline, a monoamine oxidase-B inhibitor, protects NGF-diferentiated PC12 cells against oxygen- itors in Parkinson’s disease but without any fnal conclusion glucose deprivation. J Neurosci Res 58(3):456–463 (for review and detailed discussion, see Riederer and Laux Akao Y, Maruyama W, Shimizu S, Yi H, Nakagawa Y, Shamoto- 2011). In the DATATOP study, according to the putative Nagai M, Youdim MB, Tsujimoto Y, Naoi M (2002a) Mito- antioxidant activity, the efect of selegiline and chondrial permeability transition mediates apoptosis induced by N-methyl(R)salsolinol, an endogenous neurotoxin, and is was evaluated in patients in early phases of the disease with- inhibited by Bcl-2 and rasagiline, N-propargyl-1(R)-aminoindan. out taking other anti-parkinson treatment. The motor scores J Neurochem 82(4):913–923 (UPDRS) and daily activity (ADL) were measured and the Akao Y, Maruyama W, Yi H, Shamoto-Nagai M, Youdim MB, Naoi N R need for initiating levodopa for symptoms control registered. M (2002b) An anti-Parkinson’s disease drug, -propargyl-1( )- aminoindan (rasagiline), enhances expression of anti-apoptotic Selegiline treatment improved the scores and delayed the bcl-2 in human dopaminergic SH-SY5Y cells. Neurosci Lett initiation of levodopa treatment by about 9 months. Tocoph- 326(2):105–108 erol was not more efective than the placebo. After 2 months Bakhshalizadeh S, Esmaeili F, Houshmand F, Shirzad H, Saedi M of selegiline withdrawal, the motor symptoms declined and (2011) Efects of selegiline, a inhibitor, on diferentiation of P19 embryonal carcinoma stem cells, into the neuroprotective efect of the drug could not be proven neuron-like cells. In Vitro Cell Dev Biol Anim 47(8):550–557. (Parkinson Study Group 1993). In the case of rasagiline, a https​://doi.org/10.1007/s1162​6-011-9442-3 diferently designed study was performed and revealed that Ballabriga J, Pellise A, Ferrer I (1997) l-Deprenyl does not reduce Meri- the beneft of early initiated rasagiline treatment was main- brain damage in global forebrain ischemia in adult gerbils ( ones ungiculatus). J Neurol Sci 148(1):1–5 tained during the continued treatment period. However, a Bar Am O, Amit T, Youdim MB (2004) Contrasting neuropro- signifcant diference was only found in the 1 mg/day rasa- tective and neurotoxic actions of respective metabolites of giline dose treatment and not at the higher, 2 mg/day dose anti-Parkinson drugs rasagiline and selegiline. Neurosci Lett (Parkinson Study Group 2004). In spite of several re-eval- 355(3):169–172 Bar-Am O, Yogev-Falach M, Amit T, Sagi Y, Youdim MB (2004) uations and post hoc analysis of the clinical trial data, the Regulation of protein kinase C by the anti-Parkinson drug, symptomatic and disease-modifying efect of neither drug MAO-B inhibitor, rasagiline and its derivatives, in vivo. could be unequivocally distinguished. J Neurochem 89(5):1119–1125. https​://doi.org/10.111 1/j.1471-4159.2004.02425​.x

1 3 É. Szökő et al.

Bekesi G, Tulassay Z, Lengyel G, Schaf Z, Szombath D, Stark J, Cereda E, Cilia R, Canesi M, Tesei S, Mariani CB, Zecchinelli AL, Marczell I, Nagy-Repas P, Adler I, Dinya E, Racz K, Magyar Pezzoli G (2017) Efcacy of rasagiline and selegiline in Parkin- K (2012) The efect of selegiline on total scavenger capacity son’s disease: a head-to-head 3-year retrospective case–control and liver fat content: a preliminary study in an animal model. J study. J Neurol 264(6):1254–1263. https://doi.org/10.1007/s0041​ ​ Neural Transm (Vienna) 119(1):25–30. https​://doi.org/10.1007/ 5-017-8523-y s0070​2-011-0666-x Chau KY, Cooper JM, Schapira AH (2010) Rasagiline protects against Biagini G, Frasoldati A, Fuxe K, Agnati LF (1994) The concept of alpha-synuclein induced sensitivity to oxidative stress in dopa- astrocyte-kinetic drug in the treatment of neurodegenerative minergic cells. Neurochem Int 57(5):525–529. https​://doi. diseases: evidence for l-deprenyl-induced activation of reactive org/10.1016/j.neuin​t.2010.06.017 astrocytes. Neurochem Int 25(1):17–22 Chetsawang B, Kooncumchoo P, Govitrapong P, Ebadi M (2008) Birkmayer W, Riederer P, Youdim MB, Linauer W (1975) The potenti- 1-Methyl-4-phenyl-pyridinium ion-induced oxidative stress, ation of the anti akinetic efect after l-dopa treatment by an inhib- c-Jun phosphorylation and DNA fragmentation factor-45 cleav- itor of MAO-B, Deprenil. J Neural Transm 36(3–4):303–326 age in SK-N-SH cells are averted by selegiline. Neurochem Int Birkmayer W, Riederer P, Ambrozi L, Youdim MB (1977) Implica- 53(6–8):283–288. https​://doi.org/10.1016/j.neuin​t.2008.08.007 tions of combined treatment with ‘Madopar’ and l-deprenil in Cleeter MW, Cooper JM, Schapira AH (1992) Irreversible inhibition of Parkinson’s disease. A long-term study. Lancet 1(8009):439–443 mitochondrial complex I by 1-methyl-4-phenylpyridinium: evi- Birkmayer W, Knoll J, Riederer P, Youdim MB, Hars V, Marton J dence for free radical involvement. J Neurochem 58(2):786–789 (1985) Increased life expectancy resulting from addition of Cohen G, Spina MB (1989) Deprenyl suppresses the oxidant stress l-deprenyl to Madopar treatment in Parkinson’s disease: a associated with increased dopamine turnover. Ann Neurol longterm study. J Neural Transm 64(2):113–127 26(5):689–690. https​://doi.org/10.1002/ana.41026​0518 Blandini F, Armentero MT, Fancellu R, Blaugrund E, Nappi G Cohen G, Pasik P, Cohen B, Leist A, Mytilineou C, Yahr MD (1984) (2004) Neuroprotective efect of rasagiline in a rodent model Pargyline and deprenyl prevent the neurotoxicity of 1-methyl- of Parkinson’s disease. Exp Neurol 187(2):455–459. https​://doi. 4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in monkeys. Eur J org/10.1016/j.expne​urol.2004.03.005 Pharmacol 106(1):209–210 Bonneh-Barkay D, Ziv N, Finberg JP (2005) Characterization of the Cui Y, Liu KW, Liang Y, Ip MS, Mak JC (2017) Inhibition of mono- neuroprotective activity of rasagiline in cerebellar granule cells. amine oxidase-B by selegiline reduces cigarette smoke-induced Neuropharmacology 48(3):406–416. https​://doi.org/10.1016/j. oxidative stress and infammation in airway epithelial cells. Toxi- neuro​pharm​.2004.10.016 col Lett 268:44–50. https://doi.org/10.1016/j.toxle​ t.2017.01.005​ Borroni E, Bohrmann B, Grueninger F, Prinssen E, Nave S, Loetscher Davis GC, Williams AC, Markey SP, Ebert MH, Caine ED, Reichert H, Chinta SJ, Rajagopalan S, Rane A, Siddiqui A, Ellenbroek CM, Kopin IJ (1979) Chronic secondary to intra- B, Messer J, Pähler A, Andersen JK, Wyler R, Cesura AM venous injection of meperidine analogues. Psychiatry Res (2017) Sembragiline: a novel, selective monoamine oxidase 1(3):249–254 type B inhibitor for the treatment of Alzheimer’s disease. J de Lima MN, Laranja DC, Caldana F, Grazziotin MM, Garcia VA, Pharmacol Exp Ther 362(3):413–423. https​://doi.org/10.1124/ Dal-Pizzol F, Bromberg E, Schroder N (2005) Selegiline protects jpet.117.24165​3 against recognition memory impairment induced by neonatal iron Bradbury AJ, Costall B, Jenner PG, Kelly ME, Marsden CD, Naylor treatment. Exp Neurol 196(1):177–183. https://doi.org/10.1016/j.​ RJ (1985) The neurotoxic actions of 1-methyl-4-phenylpyridine expne​urol.2005.07.017 (MPP+) are not prevented by deprenyl treatment. Neurosci Lett De Marchi U, Pietrangeli P, Marcocci L, Mondovi B, Toninello A 58(2):177–181 (2003) l-Deprenyl as an inhibitor of menadione-induced per- Braga CA, Follmer C, Palhano FL, Khattar E, Freitas MS, Romao meability transition in liver mitochondria. Biochem Pharmacol L, Di Giovanni S, Lashuel HA, Silva JL, Foguel D (2011) The 66(9):1749–1754 anti-Parkinsonian drug selegiline delays the nucleation phase of Dezsi L, Vecsei L (2017) Monoamine oxidase B inhibitors in Parkin- alpha-synuclein aggregation leading to the formation of nontoxic son’s disease. CNS Neurol Disord Drug Targets 16(4):425–439. species. J Mol Biol 405(1):254–273. https​://doi.org/10.1016/j. https​://doi.org/10.2174/18715​27316​66617​01241​65222​ jmb.2010.10.027 Dimpfel W, Hofmann JA (2011) Efects of rasagiline, its metabolite Budni P, de Lima MN, Polydoro M, Moreira JC, Schroder N, Dal- aminoindan and selegiline on glutamate receptor mediated sig- Pizzol F (2007) Antioxidant efects of selegiline in oxidative nalling in the rat hippocampus slice in vitro. BMC Pharmacol stress induced by iron neonatal treatment in rats. Neurochem 11:2. https​://doi.org/10.1186/1471-2210-11-2 Res 32(6):965–972. https://doi.org/10.1007/s1106​ 4-006-9249-x​ Eliash S, Shteter N, Eilam R (2005) Neuroprotective efect of rasa- Caccia C, Maj R, Calabresi M, Maestroni S, Faravelli L, Curatolo L, giline, a monoamine oxidase-B inhibitor, on spontaneous Salvati P, Fariello RG (2006) Safnamide: from molecular targets cell degeneration in a rat model. J Neural Transm (Vienna) to a new anti-Parkinson drug. Neurology 67(7 Suppl 2):S18–S23 112(8):991–1003. https​://doi.org/10.1007/s0070​2-004-0254-4 Carageorgiou H, Zarros A, Tsakiris S (2003) Selegiline long-term Esmaeili F, Tiraihi T, Movahedin M, Mowla SJ (2006) Selegiline efects on brain acetylcholinesterase, (Na­ +, K­ +)-ATPase activi- induces neuronal phenotype and neurotrophins expression in ties, antioxidant status and learning performance of aged rats. embryonic stem cells. Rejuvenation Res 9(4):475–484. https://​ Pharmacol Res 48(3):245–251 doi.org/10.1089/rej.2006.9.475 Carrillo MC, Kanai S, Nokubo M, Kitani K (1991) (−) deprenyl Fariello RG (2007) Safnamide. Neurotherapeutics 4(1):110–116. https​ induces activities of both superoxide dismutase and catalase but ://doi.org/10.1016/j.nurt.2006.11.011 not of glutathione peroxidase in the striatum of young male rats. Finberg JP, Takeshima T, Johnston JM, Commissiong JW (1998) Life Sci 48(6):517–521 Increased survival of dopaminergic neurons by rasagiline, a Carrillo MC, Minami C, Kitani K, Maruyama W, Ohashi K, Yamamoto monoamine oxidase B inhibitor. NeuroReport 9(4):703–707 T, Naoi M, Kanai S, Youdim MB (2000) Enhancing efect of Finnegan KT, Skratt JJ, Irwin I, DeLanney LE, Langston JW (1990) rasagiline on superoxide dismutase and catalase activities in the Protection against DSP-4-induced neurotoxicity by deprenyl dopaminergic system in the rat. Life Sci 67(5):577–585 is not related to its inhibition of MAO B. Eur J Pharmacol 184(1):119–126

1 3 Pharmacological aspects of the neuroprotective efects of irreversible MAO‑B inhibitors,…

Fuller RW, Hemrick-Luecke SK (1985) Mechanisms of MPTP Khaldy H, Escames G, Leon J, Vives F, Luna JD, Acuna-Castroviejo D (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) neurotoxicity (2000) Comparative efects of , l-deprenyl, Trolox and to striatal dopamine neurons in mice. Prog Neuropsychophar- ascorbate in the suppression of hydroxyl radical formation dur- macol Biol Psychiatry 9(5–6):687–690 ing dopamine autoxidation in vitro. J Pineal Res 29(2):100–107 Gelowitz DL, Paterson IA (1999) Neuronal sparing and behavioral Kitani K, Minami C, Isobe K, Maehara K, Kanai S, Ivy GO, Car- efects of the antiapoptotic drug, (−)deprenyl, following kainic rillo MC (2002) Why (−)deprenyl prolongs survivals of experi- acid administration. Pharmacol Biochem Behav 62(2):255–262 mental animals: increase of anti-oxidant enzymes in brain and Ghorbanian MT, Tiraihi T, Mesbah-Namin SA, Fathollahi Y (2010) other body tissues as well as mobilization of various humoral Selegiline is an efcient and potent inducer for bone marrow factors may lead to systemic anti-aging efects. Mech Ageing stromal cell diferentiation into neuronal phenotype. Neurol Dev 123(8):1087–1100 Res 32(2):185–193. https://doi.org/10.1179/17431​ 3209x​ 40901​ ​ Klegeris A, McGeer PL (2000) R-(−)-Deprenyl inhibits monocytic 6 THP-1 cell neurotoxicity independently of monoamine oxidase Gibson CJ (1987) Inhibition of MAO B, but not MAO A, blocks DSP-4 inhibition. Exp Neurol 166(2):458–464. https://doi.org/10.1006/​ toxicity on central NE neurons. Eur J Pharmacol 141(1):135–138 exnr.2000.7517 Goggi J, Theoflopoulos S, Riaz SS, Jauniaux E, Stern GM, Bradford Knoll J (1988) The striatal dopamine dependency of life span in male HF (2000) The neuronal survival efects of rasagiline and depre- rats. Longevity study with (−)deprenyl. Mech Ageing Dev nyl on fetal human and rat ventral mesencephalic neurones in 46(1–3):237–262 culture. NeuroReport 11(18):3937–3941 Knoll J (1989) The pharmacology of selegiline ((−)deprenyl). New Gotz ME, Dirr A, Burger R, Rausch WD, Riederer P (1995) High dose aspects. Acta Neurol Scand Suppl 126:83–91 selegiline augments striatal ubiquinol in mouse: an indication of Knoll J, Magyar K (1972) Some puzzling pharmacological efects of decreased oxidative stress or of interference with mitochondrial monoamine oxidase inhibitors. Adv Biochem Psychopharmacol respiration? A pilot study. J Neural Transm Suppl 46:149–156 5:393–408 Guo B, Zheng C, Cai W, Cheng J, Wang H, Li H, Sun Y, Cui W, Wang Knoll J, Miklya I (1994) Multiple, small dose administration of (−) Y, Han Y, Lee SM, Zhang Z (2016) Multifunction of chrysin in deprenyl enhances catecholaminergic activity and diminishes Parkinson’s model: anti-neuronal apoptosis, neuroprotection via serotoninergic activity in the brain and these efects are unrelated activation of MEF2D, and inhibition of monoamine oxidase-B. to MAO-B inhibition. Arch Int Pharmacodyn Ther 328(1):1–15 J Agric Food Chem 64(26):5324–5333. https://doi.org/10.1021/​ Knoll J, Ecseri Z, Kelemen K, Nievel J, Knoll B (1965) Phenyliso- acs.jafc.6b017​07 propylmethylpropinylamine (E-250), a new spectrum psychic Haberle D, Szoko E, Halasz AS, Magyar K (2001) The efect of low energizer. Arch Int Pharmacodyn Ther 155(1):154–164 oral doses of (−)-deprenyl and its metabolites on DSP-4 toxic- Knoll J, Vizi ES, Somogyi G (1968) Phenylisopropyl-methyl-propi- ity. J Neural Transm (Vienna) 108(11):1239–1247. https​://doi. nylamine (E-250), a monoaminoxidase inhibitor antagonizing org/10.1007/s0070​20100​002 efects of tyramine. Arzn-Forsch 18:109–112 Hassanzadeh K, Nikzaban M, Moloudi MR, Izadpanah E (2015) Efect Knoll J, Dallo J, Yen TT (1989) Striatal dopamine, sexual activity and of selegiline on neural stem cells diferentiation: a possible role lifespan. Longevity of rats treated with (−)deprenyl. Life Sci for neurotrophic factors. Iran J Basic Med Sci 18(6):549–554 45(6):525–531 Heikkila RE, Nicklas WJ, Vyas I, Duvoisin RC (1985) Dopaminergic Knollema S, Aukema W, Hom H, Korf J, ter Horst GJ (1995) l-depre- toxicity of rotenone and the 1-methyl-4-phenylpyridinium ion nyl reduces brain damage in rats exposed to transient hypoxia- after their stereotaxic administration to rats: implication for the ischemia. Stroke 26(10):1883–1887 mechanism of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine tox- Konradi C, Kornhuber J, Froelich L, Fritze J, Heinsen H, Beckmann icity. Neurosci Lett 62(3):389–394 H, Schulz E, Riederer P (1989) Demonstration of monoamine Heinonen EH, Myllyla V, Sotaniemi K, Lamintausta R, Salonen JS, oxidase-A and -B in the human brainstem by a histochemical Anttila M, Savijarvi M, Kotila M, Rinne UK (1989) Pharmacoki- technique. Neuroscience 33(2):383–400 netics and metabolism of selegiline. Acta Neurol Scand Suppl Kontkanen O, Castren E (1999) Trophic efects of selegiline on cul- 126:93–99 tured dopaminergic neurons. Brain Res 829(1–2):190–192 Inaba-Hasegawa K, Akao Y, Maruyama W, Naoi M (2012) Type A Kornhuber J, Konradi C, Mack-Burkhardt F, Riederer P, Heinsen H, monoamine oxidase is associated with induction of neuropro- Beckmann H (1989) Ontogenesis of monoamine oxidase-A and tective Bcl-2 by rasagiline, an inhibitor of type B monoamine -B in the human brain frontal cortex. Brain Res 499(1):81–86 oxidase. J Neural Transm (Vienna) 119(4):405–414. https​://doi. Koutsilieri E, O’Callaghan JF, Chen TS, Riederer P, Rausch WD org/10.1007/s0070​2-011-0730-6 (1994) Selegiline enhances survival and neurite outgrowth Johnston JP (1968) Some observations upon a new inhibitor of of MPP(+)-treated dopaminergic neurons. Eur J Pharmacol monoamine oxidase in brain tissue. Biochem Pharmacol 269(3):R3–R4 17(7):1285–1297 Koutsilieri E, Chen TS, Rausch WD, Riederer P (1996) Selegiline is Kakish J, Tavassoly O, Lee JS (2015) Rasagiline, a suicide inhibitor of neuroprotective in primary brain cultures treated with 1-methyl- monoamine oxidases, binds reversibly to alpha-synuclein. ACS 4-phenylpyridinium. Eur J Pharmacol 306(1–3):181–186 Chem Neurosci 6(2):347–355. https​://doi.org/10.1021/cn500​ Kupsch A, Sautter J, Gotz ME, Breithaupt W, Schwarz J, Youdim MB, 2914 Riederer P, Gerlach M, Oertel WH (2001) Monoamine oxidase- Katagi M, Tatsuno M, Miki A, Nishikawa M, Nakajima K, Tsuchihashi inhibition and MPTP-induced neurotoxicity in the non-human H (2001) Simultaneous determination of selegiline-N-oxide, a primate: comparison of rasagiline (TVP 1012) with selegiline. new indicator for selegiline administration, and other metabo- J Neural Transm (Vienna) 108(8–9):985–1009. https​://doi. lites in urine by high-performance liquid chromatography-elec- org/10.1007/s0070​20170​018 trospray ionization mass spectrometry. J Chromatogr B Biomed Kwon YS, Ann HS, Nabeshima T, Shin EJ, Kim WK, Jhoo JH, Jhoo Sci Appl 759(1):125–133 WK, Wie MB, Kim YS, Jang KJ, Kim HC (2004) Selegiline Kaur J, Singh S, Sharma D, Singh R (2003) Neurostimulatory and potentiates the efects of EGb 761 in response to ischemic antioxidative efects of l-deprenyl in aged rat brain regions. brain injury. Neurochem Int 45(1):157–170. https​://doi. Biogerontology 4(2):105–111 org/10.1016/j.neuin​t.2003.10.005

1 3 É. Szökő et al.

Langston JW, Irwin I, Langston EB, Forno LS (1984) Pargyline induced by peroxynitrite in human dopaminergic neuroblastoma prevents MPTP-induced parkinsonism in primates. Science SH-SY5Y cells. J Neural Transm (Vienna) 109(4):467–481. https​ 225(4669):1480–1482 ://doi.org/10.1007/s0070​20200​038 Li XM, Juorio AV, Qi J, Boulton AA (1998) L-deprenyl potentiates Maruyama W, Nitta A, Shamoto-Nagai M, Hirata Y, Akao Y, Yodim NGF-induced changes in superoxide dismutase mRNA in PC12 M, Furukawa S, Nabeshima T, Naoi M (2004) N-Propargyl-1 cells. J Neurosci Res 53(2):235–238. https://doi.org/10.1002/ (R)-aminoindan, rasagiline, increases glial cell line-derived (sici)1097-4547(19980715)53:2<235::aid-jnr12>3.0.co;2-5 neurotrophic factor (GDNF) in neuroblastoma SH-SY5Y cells Magyar K (1994) Behaviour of (−)-deprenyl and its analogues. J through activation of NF-kappaB transcription factor. Neuro- Neural Transm Suppl 41:167–175 chem Int 44(6):393–400 Magyar K (2011) The pharmacology of selegiline. Int Rev Neu- Marzo A, Dal Bo L, Monti NC, Crivelli F, Ismaili S, Caccia C, Cat- robiol 100:65–84. https​://doi.org/10.1016/b978-0-12-38646​ taneo C, Fariello RG (2004) and pharmaco- 7-3.00004​-2 dynamics of safnamide, a neuroprotectant with antiparkinsonian Magyar K, Haberle D (1999) Neuroprotective and neuronal rescue and anticonvulsant activity. Pharmacol Res 50(1):77–85. https://​ efects of selegiline: review. Neurobiology (Bp) 7(2):175–190 doi.org/10.1016/j.phrs.2003.12.004 Magyar K, Tothfalusi L (1984) Pharmacokinetic aspects of deprenyl Matsubara K, Senda T, Uezono T, Awaya T, Ogawa S, Chiba K, efects. Pol J Pharmacol Pharm 36(4):373–384 Shimizu K, Hayase N, Kimura K (2001) l-Deprenyl prevents Magyar K, Vizi ES, Ecseri Z, Knoll J (1967) Comparative pharma- the cell hypoxia induced by dopaminergic neurotoxins, MPP(+) cological analysis of the optical isomers of phenyl-isopropyl- and beta-carbolinium: a microdialysis study in rats. Neurosci methyl-propinylamine (E-250). Acta Physiol Acad Sci Hung Lett 302(2–3):65–68 32(4):377–387 Mihatsch W, Russ H, Przuntek H (1988) Intracerebroventricular Magyar K, Ecseri Z, Bernáth G, Sátory É, Knoll J (1979) Sturc- administration of 1-methyl-4-phenylpyridinium ion in mice: ture-activity relationship of selective inhibitors of MAO-B. effects of simultaneously administered , depre- In: Magyar K (ed) Monoamine oxidases and their selective nyl, and 1-t-butyl-4,4-diphenylpiperidine. J Neural Transm inhibition. Pergamon Press, Budapest, pp 11–21 71(3):177–188 Magyar K, Szende B, Lengyel J, Tekes K (1996) The pharmacology Mizuno Y, Suzuki K, Sone N, Saitoh T (1987) Inhibition of ATP syn- of B-type selective monoamine oxidase inhibitors; milestones thesis by 1-methyl-4-phenylpyridinium ion (MPP+) in isolated in (−)-deprenyl research. J Neural Transm Suppl 48:29–43 mitochondria from mouse brains. Neurosci Lett 81(1–2):204–208 Magyar K, Szende B, Lengyel J, Tarczali J, Szatmary I (1998) The Mizuno Y, Ohta S, Tanaka M, Takamiya S, Suzuki K, Sato T, Oya H, neuroprotective and neuronal rescue efects of (−)-deprenyl. J Ozawa T, Kagawa Y (1989) Defciencies in complex I subunits Neural Transm Suppl 52:109–123 of the respiratory chain in Parkinson’s disease. Biochem Biophys Magyar K, Palf M, Tabi T, Kalasz H, Szende B, Szoko E (2004) Res Commun 163(3):1450–1455 Pharmacological aspects of (−)-deprenyl. Curr Med Chem Mizuta I, Ohta M, Ohta K, Nishimura M, Mizuta E, Hayashi K, Kuno 11(15):2017–2031 S (2000) Selegiline and desmethylselegiline stimulate NGF, Magyar K, Palf M, Jenei V, Szoko E (2006) Deprenyl: from chemi- BDNF, and GDNF synthesis in cultured mouse astrocytes. cal synthesis to neuroprotection. J Neural Transm Suppl Biochem Biophys Res Commun 279(3):751–755. https​://doi. 71:143–156 org/10.1006/bbrc.2000.4037 Maia FD, Pitombeira BS, Araujo DT, Cunha GM, Viana GS (2004) Mytilineou C, Cohen G (1985) Deprenyl protects dopamine neurons l-Deprenyl prevents lipid peroxidation and memory defcits from the neurotoxic efect of 1-methyl-4-phenylpyridinium ion. produced by cerebral ischemia in rats. Cell Mol Neurobiol J Neurochem 45(6):1951–1953 24(1):87–100 Mytilineou C, Radcliffe P, Leonardi EK, Werner P, Olanow CW Marconi S, Zwingers T (2014) Comparative efcacy of selegiline ver- (1997a) l-deprenyl protects mesencephalic dopamine neurons sus rasagiline in the treatment of early Parkinson’s disease. Eur from glutamate receptor-mediated toxicity in vitro. J Neurochem Rev Med Pharmacol Sci 18(13):1879–1882 68(1):33–39 Maruyama W, Akao Y, Youdim MB, Naoi M (2000a) Neurotoxins Mytilineou C, Radcliffe PM, Olanow CW (1997b) l-(−)- induce apoptosis in dopamine neurons: protection by N-propar- desmethylselegiline, a metabolite of selegiline [l-(−)-deprenyl], gylamine-1(R)- and (S)-aminoindan, rasagiline and TV1022. J protects mesencephalic dopamine neurons from excitotoxicity Neural Transm Suppl 60:171–186 in vitro. J Neurochem 68(1):434–436 Maruyama W, Yamamoto T, Kitani K, Carrillo MC, Youdim M, Naoi Naoi M, Maruyama W, Yagi K, Youdim M (2000) Anti-apoptotic M (2000b) Mechanism underlying anti-apoptotic activity of a (−) function of l-(−)deprenyl (Selegiline) and related compounds. deprenyl-related propargylamine, rasagiline. Mech Ageing Dev Neurobiology (Bp) 8(1):69–80 116(2–3):181–191 Nardai S, Dobolyi A, Pal G, Skopal J, Pinter N, Lakatos K, Merkely Maruyama W, Akao Y, Youdim MB, Davis BA, Naoi M (2001a) B, Nagy Z (2015) Selegiline promotes NOTCH-JAGGED sign- Transfection-enforced Bcl-2 overexpression and an anti-Parkin- aling in astrocytes of the peri-infarct region and improves the son drug, rasagiline, prevent nuclear accumulation of glyceral- functional integrity of the neurovascular unit in a rat model of dehyde-3-phosphate dehydrogenase induced by an endogenous focal ischemia. Restor Neurol Neurosci 33(1):1–14. https​://doi. dopaminergic neurotoxin, N-methyl(R)salsolinol. J Neurochem org/10.3233/rnn-14042​0 78(4):727–735 Nave S, Doody RS, Boada M, Grimmer T, Savola JM, Delmar P, Maruyama W, Youdim MB, Naoi M (2001b) Antiapoptotic properties Pauly-Evers M, Nikolcheva T, Czech C, Borroni E, Ricci B, of rasagiline, N-propargylamine-1(R)-aminoindan, and its optical Dukart J, Mannino M, Carey T, Moran E, Gilaberte I, Muel- (S)-isomer, TV1022. Ann N Y Acad Sci 939:320–329 hardt NM, Gerlach I, Santarelli L, Ostrowitzki S, Fontoura P Maruyama W, Akao Y, Carrillo MC, Kitani K, Youdium MB, Naoi (2017) Sembragiline in moderate Alzheimer’s disease: results M (2002a) Neuroprotection by propargylamines in Parkinson’s of a randomized, double-blind, placebo-controlled phase II trial disease: suppression of apoptosis and induction of prosurvival (MAyfOwer RoAD). J Alzheimers Dis 58(4):1217–1228. https​ genes. Neurotoxicol Teratol 24(5):675–682 ://doi.org/10.3233/JAD-16130​9 Maruyama W, Takahashi T, Youdim M, Naoi M (2002b) The anti- Nicklas WJ, Vyas I, Heikkila RE (1985) Inhibition of NADH-linked Parkinson drug, rasagiline, prevents apoptotic DNA damage oxidation in brain mitochondria by 1-methyl-4-phenyl-pyridine,

1 3 Pharmacological aspects of the neuroprotective efects of irreversible MAO‑B inhibitors,…

a metabolite of the neurotoxin, 1-methyl-4-phenyl-1,2,5,6-tet- Riva MA, Molteni R, Racagni G (1997) l-deprenyl potentiates rahydropyridine. Life Sci 36(26):2503–2508 cAMP-induced elevation of FGF-2 mRNA levels in rat corti- Nicklas WJ, Youngster SK, Kindt MV, Heikkila RE (1987) MPTP, cal astrocytes. NeuroReport 8(9–10):2165–2168 MPP+ and mitochondrial function. Life Sci 40(8):721–729 Ross SB (1976) Long-term efects of N-2-chlorethyl-N-ethyl-2-bro- Niittykoski M, Haapalinna A, Sirvio J (2003) Selegiline reduces mobenzylamine hydrochloride on noradrenergic neurones in N-methyl-d-aspartic acid induced perturbation of neurotransmis- the rat brain and heart. Br J Pharmacol 58(4):521–527 sion but it leaves NMDA receptor dependent long-term poten- Sagi Y, Mandel S, Amit T, Youdim MB (2007) Activation of tyrosine tiation intact in the hippocampus. J Neural Transm (Vienna) kinase receptor signaling pathway by rasagiline facilitates neu- 110(11):1225–1240. https://doi.org/10.1007/s0070​ 2-003-0035-5​ rorescue and restoration of nigrostriatal dopamine neurons in Oh C, Murray B, Bhattacharya N, Holland D, Tatton WG (1994) post-MPTP-induced parkinsonism. Neurobiol Dis 25(1):35–44. (−)-Deprenyl alters the survival of adult murine facial motoneu- https​://doi.org/10.1016/j.nbd.2006.07.020 rons after axotomy: increases in vulnerable C57BL strain but Salo PT, Tatton WG (1992) Deprenyl reduces the death of motoneu- decreases in motor neuron degeneration mutants. J Neurosci Res rons caused by axotomy. J Neurosci Res 31(2):394–400. https​ 38(1):64–74. https​://doi.org/10.1002/jnr.49038​0109 ://doi.org/10.1002/jnr.49031​0223 Ou XM, Lu D, Johnson C, Chen K, Youdim MB, Rajkowska G, Shih JC Salonen T, Haapalinna A, Heinonen E, Suhonen J, Hervonen A (2009) Glyceraldehyde-3-phosphate dehydrogenase-monoamine (1996) Monoamine oxidase B inhibitor selegiline protects oxidase B-mediated cell death-induced by is prevented young and aged rat peripheral sympathetic neurons against by rasagiline and 1-R-aminoindan. Neurotox Res 16(2):148–159. 6-hydroxydopamine-induced neurotoxicity. Acta Neuropathol https​://doi.org/10.1007/s1264​0-009-9064-7 91(5):466–474 Parkinson Study Group (1993) Efects of tocopherol and deprenyl on Saravanan KS, Sindhu KM, Senthilkumar KS, Mohanakumar KP the progression of disability in early Parkinson’s disease. N Engl (2006) l-deprenyl protects against rotenone-induced, oxida- J Med 328(3):176–183. https​://doi.org/10.1056/nejm1​99301​ tive stress-mediated dopaminergic neurodegeneration in rats. 21328​0305 Neurochem Int 49(1):28–40. https​://doi.org/10.1016/j.neuin​ Parkinson Study Group (2004) A controlled, randomized, delayed- t.2005.12.016 start study of rasagiline in early Parkinson disease. Arch Neurol Schapira AH, Cooper JM, Dexter D, Jenner P, Clark JB, Marsden CD 61(4):561–566. https​://doi.org/10.1001/archn​eur.61.4.561 (1989) Mitochondrial complex I defciency in Parkinson’s dis- Paterson IA, Zhang D, Warrington RC, Boulton AA (1998) R-deprenyl ease. Lancet 1(8649):1269 and R-2-heptyl-N-methylpropargylamine prevent apoptosis in Semkova I, Wolz P, Schilling M, Krieglstein J (1996) Selegiline cerebellar granule neurons induced by cytosine arabinoside but enhances NGF synthesis and protects central nervous system not low extracellular potassium. J Neurochem 70(2):515–523 neurons from excitotoxic and ischemic damage. Eur J Pharma- Pazini AM, Gomes GM, Villarinho JG, da Cunha C, Pinheiro F, col 315(1):19–30 Ferreira AP, Mello CF, Ferreira J, Rubin MA (2013) Sele- Sharma SK, Carlson EC, Ebadi M (2003) Neuroprotective actions giline reverses abeta(2)(5)(−)(3)(5)-induced cognitive defcit of Selegiline in inhibiting 1-methyl, 4-phenyl, pyridinium ion in male mice. Neurochem Res 38(11):2287–2294. https​://doi. (MPP+)-induced apoptosis in SK-N-SH neurons. J Neurocytol org/10.1007/s1106​4-013-1137-6 32(4):329–343. https​://doi.org/10.1023/B:NEUR.00000​11327​ Peretz C, Segev H, Rozani V, Gurevich T, El-Ad B, Tsamir J, Giladi .23739​.1b N (2016) Comparison of selegiline and rasagiline therapies Shin HS (1997) Metabolism of selegiline in humans. Identifcation, in Parkinson disease: a real-life study. Clin Neuropharmacol , and stereochemistry of urine metabolites. Drug Metab 39(5):227–231. https://doi.org/10.1097/wnf.00000​ 00000​ 00016​ 7​ Dispos 25(6):657–662 Qin F, Shite J, Mao W, Liang CS (2003) Selegiline attenuates car- Sian-Hülsmann J, Mandel S, Youdim MB, Riederer P (2011) The diac oxidative stress and apoptosis in heart failure: associa- relevance of iron in the pathogenesis of Parkinson’s disease. J tion with improvement of cardiac function. Eur J Pharmacol Neurochem. 118(6):939–957 461(2–3):149–158 Siddiqui MA, Plosker GL (2005) Rasagiline. Drugs Aging 22(1):83–91 Reichmann H, Riederer P. (1989). Biochemische Analyse der Atmung- Singh R, Mishra M, Singh S, Sharma D (2012) Efect of l-deprenyl skomplexkette verschiedener Hirnregionen von Patienten mit treatment on electrical activity, Na­ +, K­ + ATPase, and protein M. Parkinson. Symposium zu Morbus Parkinson und anderen kinase C activities in hippocampal subfelds (CA1 and CA3) of Basalganglienerkrankungen. Ministerium für Forschung und aged rat brain. Indian J Exp Biol 50(2):101–109 Technologie (BMBF)m Bad Kissingen, p 44 Speiser Z, Mayk A, Eliash S, Cohen S (1999) Studies with rasagil- Reynolds GP, Riederer P, Sandler M, Jellinger K, Seemann D (1978) ine, a MAO-B inhibitor, in experimental focal ischemia in the Amphetamine and 2-phenylethylamine in post-mortem Parkin- rat. J Neural Transm (Vienna) 106(7–8):593–606. https​://doi. sonian brain after (−)deprenyl administration. J Neural Transm org/10.1007/s0070​20050​182 43(3–4):271–277 Speiser Z, Mayk A, Litinetsky L, Fine T, Nyska A, Blaugrund E, Riederer P, Laux G (2011) MAO-inhibitors in Parkinson’s disease. Exp Cohen S (2007) Rasagiline is neuroprotective in an experimen- Neurobiol 20(1):1–17. https​://doi.org/10.5607/en.2011.20.1.1 tal model of brain ischemia in the rat. J Neural Transm (Vienna) Riederer P, Youdim MB (1986) Monoamine oxidase activity and mon- 114(5):595–605. https​://doi.org/10.1007/s0070​2-006-0612-5 oamine metabolism in brains of parkinsonian patients treated Sprague JE, Nichols DE (1995) The monoamine oxidase-B inhibitor with l-deprenyl. J Neurochem 46(5):1359–1365 l-deprenyl protects against 3,4-methylenedioxymethamphet- Riederer P, Youdim MB, Rausch WD, Birkmayer W, Jellinger K, See- amine-induced lipid peroxidation and long-term serotonergic mann D (1978) On the mode of action of l-deprenyl in the human defcits. J Pharmacol Exp Ther 273(2):667–673 central nervous system. J Neural Transm 43(3–4):217–226 Suresh J, Baek SC, Ramakrishnan SP, Kim H, Mathew B (2018) Dis- Riederer P, Jellinger K, Seemann D (1984) Monoamine oxidase and covery of potent and reversible MAO-B inhibitors as furano- parkinsonism. In: Tipton KF, Dostert P, Strolin-Benedetti M chalcones. Int J Biol Macromol 108:660–664. https​://doi. (eds) Monoamine oxidase and disease. Academic Press, Lon- org/10.1016/j.ijbio​mac.2017.11.159 don, pp 403–415 Suuronen T, Kolehmainen P, Salminen A (2000) Protective efect of l-deprenyl against apoptosis induced by okadaic acid in cultured neuronal cells. Biochem Pharmacol 59(12):1589–1595

1 3 É. Szökő et al.

Szende B, Magyar K, Szegedi Z (2000) Apoptotic and antiapoptotic Varga E, Tringer L (1967) Clinical trial of a new type promptly act- efect of (−)deprenyl and (−)-desmethyl-deprenyl on human ing psychoenergetic agent (phenyl-isopropyl-methylpropinyl- cell lines. Neurobiology (Bp) 8(3–4):249–255 HCl, “E-250”). Acta Med Acad Sci Hung 23(3):289–295 Szoko E, Kalasz H, Magyar K (1999) Biotransformation of deprenyl Vizuete ML, Stefen V, Ayala A, Cano J, Machado A (1993) Protec- enantiomers. Eur J Drug Metab Pharmacokinet 24(4):315–319 tive efect of deprenyl against 1-methyl-4-phenylpyridinium Szökő É, Tábi T, Halász AS, Pálf M, Kalász H (2004a) Identifcation neurotoxicity in rat striatum. Neurosci Lett 152(1–2):113–116 of the eneatiomer form of deprenyl metabolites and deprenyl- Wahdan SA, Tadros MG, Khalifa AE (2017) Antioxidant and antia- N-oxide in rat urine. In: Török TL, Klebovich I (eds) Mono- poptotic actions of selegiline protect against 3-NP-induced amine oxidase inhibitors and their role in neurotransmission neurotoxicity in rats. Naunyn Schmiedebergs Arch Pharmacol. (). Medicina, Budapest, pp 41–54 https​://doi.org/10.1007/s0021​0-017-1392-1 Szökő É, Tábi T, Halász AS, Pálf M, Kalász H, Magyar K (2004b) Waldmeier PC, Boulton AA, Cools AR, Kato AC, Tatton WG Chiral characterization and quantifcation of deprenyl-N-oxide (2000a) Neurorescuing efects of the GAPDH ligand CGP and other deprenyl metabolites in rat urine by capillary elec- 3466B. J Neural Transm Suppl 60:197–214 trophoresis. Chromatographia 60:S245–S251 Waldmeier PC, Spooren WP, Hengerer B (2000b) CGP 3466 protects Tabi T, Magyar K, Szoko E (2003) Chiral characterization of dopaminergic neurons in lesion models of Parkinson’s disease. deprenyl-N-oxide and other deprenyl metabolites by capillary Naunyn Schmiedebergs Arch Pharmacol 362(6):526–537 electrophoresis using a dual cyclodextrin system in rat urine. Weinreb O, Amit T, Bar-Am O, Chillag-Talmor O, Youdim MB Electrophoresis 24(15):2665–2673. https​://doi.org/10.1002/ (2005) Novel neuroprotective mechanism of action of rasa- elps.20030​5468 giline is associated with its propargyl moiety: interaction of Takahata K, Katsuki H, Kobayashi Y, Muraoka S, Yoneda F, Kume T, Bcl-2 family members with PKC pathway. Ann N Y Acad Sci Kashii S, Honda Y, Akaike A (2003) Protective efects of sele- 1053:348–355. https​://doi.org/10.1196/annal​s.1344.030 giline and desmethylselegiline against N-methyl-d-aspartate- Weinreb O, Amit T, Bar-Am O, Sagi Y, Mandel S, Youdim MB induced rat retinal damage. Eur J Pharmacol 458(1–2):81–89 (2006) Involvement of multiple survival signal transduction Takahata K, Shimazu S, Katsuki H, Yoneda F, Akaike A (2006) pathways in the neuroprotective, neurorescue and APP process- Efects of selegiline on antioxidant systems in the nigrostria- ing activity of rasagiline and its propargyl moiety. J Neural tum in rat. J Neural Transm (Vienna) 113(2):151–158. https://​ Transm Suppl 70:457–465 doi.org/10.1007/s0070​2-005-0309-1 Wu RM, Chiueh CC, Pert A, Murphy DL (1993) Apparent anti- Tatton WG, Chalmers-Redman RM (1996) Modulation of gene oxidant efect of l-deprenyl on hydroxyl radical formation expression rather than monoamine oxidase inhibition: and nigral injury elicited by MPP+ in vivo. Eur J Pharmacol (−)-deprenyl-related compounds in controlling neurodegen- 243(3):241–247 eration. Neurology 47(6 Suppl 3):S171–S183 Wu RM, Murphy DL, Chiueh CC (1996) Suppression of hydroxyl Tatton WG, Greenwood CE (1991) Rescue of dying neurons: a new radical formation and protection of nigral neurons by l-depre- action for deprenyl in MPTP parkinsonism. J Neurosci Res nyl (selegiline). Ann N Y Acad Sci 786:379–390 30(4):666–672. https​://doi.org/10.1002/jnr.49030​0410 Wu Y, Kazumura K, Maruyama W, Osawa T, Naoi M (2015) Rasagil- Tatton WG, Chalmers-Redman RM, Ju WJ, Mammen M, Carlile GW, ine and selegiline suppress calcium efux from mitochondria Pong AW, Tatton NA (2002) Propargylamines induce antia- by PK11195-induced opening of mitochondrial permeability poptotic new protein synthesis in serum- and nerve growth transition pore: a novel anti-apoptotic function for neuropro- factor (NGF)-withdrawn, NGF-diferentiated PC-12 cells. J tection. J Neural Transm (Vienna) 122(10):1399–1407. https​ Pharmacol Exp Ther 301(2):753–764 ://doi.org/10.1007/s0070​2-015-1398-0 Tazik S, Johnson S, Lu D, Johnson C, Youdim MB, Stockmeier CA, Wu Y, Shamoto-Nagai M, Maruyama W, Osawa T, Naoi M (2016) Ou XM (2009) Comparative neuroprotective efects of rasagil- Rasagiline prevents cyclosporine A-sensitive superoxide ine and aminoindan with selegiline on dexamethasone-induced fashes induced by PK11195, the initial signal of mitochon- brain cell apoptosis. Neurotox Res 15(3):284–290. https://doi.​ drial membrane permeabilization and apoptosis. J Neural org/10.1007/s1264​0-009-9030-4 Transm (Vienna) 123(5):491–494. https​://doi.org/10.1007/ Thiffault C, Quirion R, Poirier J (1997) The effect of l-depre- s0070​2-016-1531-8 nyl, d-deprenyl and MDL72974 on mitochondrial respira- Xu L, Ma J, Seigel GM, Ma JX (1999) l-Deprenyl, blocking apopto- tion: a possible mechanism leading to an adaptive increase sis and regulating gene expression in cultured retinal neurons. in superoxide dismutase activity. Brain Res Mol Brain Res Biochem Pharmacol 58(7):1183–1190 49(1–2):127–136 Yeon SK, Choi JW, Park JH, Lee YR, Kim HJ, Shin SJ, Jang BK, Toronyi E, Hamar J, Magyar K, Szende B (2002) Antiapoptotic efect Kim S, Bahn YS, Han G, Lee YS, Pae AN, Park KD (2018) of (−)-deprenyl in rat kidney after ischemia–reperfusion. Med Synthesis and evaluation of biaryl derivatives for structural Sci Monit 8(2):BR65–BR68 characterization of selective monoamine oxidase B inhibi- Trudler D, Weinreb O, Mandel SA, Youdim MB, Frenkel D (2014) tors toward Parkinson’s disease therapy. Bioorg Med Chem DJ-1 defciency triggers microglia sensitivity to dopamine 26(1):232–244. https​://doi.org/10.1016/j.bmc.2017.11.036 toward a pro-inflammatory phenotype that is attenuated Yogev-Falach M, Amit T, Bar-Am O, Youdim MB (2003) The by rasagiline. J Neurochem 129(3):434–447. https​://doi. importance of propargylamine moiety in the anti-Parkinson org/10.1111/jnc.12633​ drug rasagiline and its derivatives in MAPK-dependent amy- Unal I, Gursoy-Ozdemir Y, Bolay H, Soylemezoglu F, Saribas O, loid precursor protein processing. FASEB J 17(15):2325–2327. Dalkara T (2001) Chronic daily administration of selegiline https​://doi.org/10.1096/f.03-0078f​ e and EGb 761 increases brain’s resistance to ischemia in mice. Youdim MB, Riederer P (1993) Dopamine metabolism and neu- Brain Res 917(2):174–181 rotransmission in primate brain in relationship to monoam- Vaglini F, Pardini C, Cavalletti M, Maggio R, Corsini GU (1996) ine oxidase A and B inhibition. J Neural Transm Gen Sect l-deprenyl fails to protect mesencephalic dopamine neurons 91(2–3):181–195 and PC12 cells from the neurotoxic efect of 1-methyl-4-phe- Zhao Q, Cai D, Bai Y (2013) Selegiline rescues gait defcits and nylpyridinium ion. Brain Res 741(1–2):68–74 the loss of dopaminergic neurons in a subacute MPTP mouse

1 3 Pharmacological aspects of the neuroprotective efects of irreversible MAO‑B inhibitors,…

model of Parkinson’s disease. Int J Mol Med 32(4):883–891. Zhu W, Xie W, Pan T, Jankovic J, Li J, Youdim MB, Le W (2008) https​://doi.org/10.3892/ijmm.2013.1450 Comparison of neuroprotective and neurorestorative capabilities Zhu J, Hamm RJ, Reeves TM, Povlishock JT, Phillips LL (2000) of rasagiline and selegiline against lactacystin-induced nigros- Postinjury administration of l-deprenyl improves cognitive triatal dopaminergic degeneration. J Neurochem 105(5):1970– function and enhances neuroplasticity after traumatic brain 1978. https​://doi.org/10.1111/j.1471-4159.2008.05330​.x injury. Exp Neurol 166(1):136–152. https​://doi.org/10.1006/ exnr.2000.7484

1 3