Review Article

Melatoninergic System in Parkinson’s Disease: From Neuroprotection to the Management of Motor and Nonmotor Symptoms

Table 2

Summary of studies presenting neuroprotective effects of melatonin in in vivo and in vitro models of PD.

ToxinSubjectsExperimental approachMain findingsRef.

6-OHDAMale Wistar ratsUnilateral injection of 6-OHDA (8 g) into the right SNc. Treatment with melatonin (1 and 10 mg/kg, i.p.) before apomorphine administration.Melatonin treatment inhibited apomorphine-induced rotational behaviour.[131]
PC12 cellsPreincubation (3 h) with melatonin (10−7 and 10−9 M). Incubation with 6-OHDA (25, 50, 100, and 250 M).Melatonin prevented the loss of cell viability and apoptosis induced by 6-OHDA. Melatonin also protected the reduction of mRNAs of antioxidant enzymes evoked by 6-OHDA.[132]
Male Sprague-Dawley ratsUnilateral injection of 6-OHDA (20 g in 5 L) into the right striatum. Melatonin (3 and 10 mg/kg, i.p.) was administrated 1 h before and immediately and 1 and 2 h after 6-OHDA injection. After that, the animals received a daily administration of melatonin in the next 3 days.Melatonin treatment recovered the 6-OHDA-induced changes in striatal MDA and DA levels and TH activity.[133]
PC12 cellsPreincubation (3 h) with melatonin 10−7 M. Incubation with 25, 50, and 100 M of 6-OHDA.Melatonin protected cells from apoptosis and necrotic lesions induced by 6-OHDA.[134]
Male Sprague-Dawley ratsUnilateral injection of 6-OHDA (8 g in 2 L) into the right SNC. Melatonin treatment (50 ± 7.5 g/h, s.c.) started immediately after 6-OHDA injection and it was maintained for 7 days.Melatonin treatment prevented apomorphine-induced rotational behaviour and loss of complex I activity induced by 6-OHDA.[135]
Male Wistar ratsUnilateral injection of 6-OHDA into the right striatum (two injections of 12 g in 1 g of saline). Posttreatment (1 h) with melatonin (2, 5, 10, and 25 mg/kg, i.p.), daily for 7 days.Melatonin prevented 6-OHDA-induced depletion of striatal DA and serotonin levels. Melatonin blocked the apomorphine-induced rotational behaviour.[136]
SK-N-SH cellsPreincubation (1 h) with melatonin (0.1, 0.5, 1.0, and 2.0 mM). Incubation with 6-OHDA (100 M) for 24 h.Melatonin protected against 6-OHDA-induced loss of cellular viability and increased activity of c-Jun-N terminal kinase signalling cascade.[137]
Male Sprague-Dawley ratsUnilateral injection of 6-OHDA (8.75 g) into striatum. After lesion, animals received melatonin (0.4 or 4 g/mL) in drinking water for 10 weeks.Melatonin 4 g/mL recovered motor deficits and normalized TH immunoreactivity and GDNF mRNA levels.[138]
Female Sprague-Dawley ratsPretreatment with melatonin (0.5 mg/kg, i.p.) for 7 days. On day 8, animals received an unilateral injection of 6-OHDA (8 g) into the lateral striatum.Melatonin treatment prevented motor deficits (observed in the apomorphine-induced rotational behaviour, staircase test, disengage time, stepping test, initiation time, and postural balance test) induced by 6-OHDA administration.[139]
Male Wistar ratsUnilateral injection of 6-OHDA (8 g) into the right medial forebrain bundle (MFB). Melatonin treatment (10 mg/kg, p.o.) began four days after 6-OHDA injection and continued for 30 days.Melatonin treatment improved motor performance without evoking dyskinesia. Melatonin also protected TH-positive neurons and neuronal ultrastructure of striatum.[140]
Male Wistar ratsUnilateral injection of 6-OHDA (12 g) into the right MFB. Melatonin (10 mg/kg/day, i.p.) was administrated 23 days before and 7 days after (pre- and posttreatment) or only 7 days after (posttreatment) the injection of 6-OHDA.Melatonin decreased COX and caspase-3 activity and PGE2 levels and increased Bcl-2 levels that have been altered by 6-OHDA injection. Melatonin also prevented the loss of DAergic neurons in SNc.[141]
Male Sprague-Dawley ratsUnilateral injection of 6-OHDA (20 g in 5 L) into the right striatum. Melatonin (3 and 10 mg/kg, i.p.) was administrated 1 h before and immediately and 1 and 2 h after 6-OHDA injection. After that, the animals received a daily administration of melatonin in the next 3 days.Melatonin treatment reduced motor deficits and protected against 6-OHDA-induced loss of DAergic neurons in SNc and in dorsolateral striatum.[142]
Male Wistar ratsUnilateral injection of 6-OHDA (12 g) into the right MFB. Melatonin (10 mg/kg/day) was administrated 23 days before and 7 days after (pre- and posttreatment) or only 7 days after (posttreatment) the injection of 6-OHDA.Melatonin treatment protected against the 7-OHDA-induced loss of DAergic neurons, increased antioxidant enzyme activities (SOD, catalase and GPx), and decreased lipid peroxidation. The pretreatment with melatonin was more effective in protecting against the 6-OHDA-induced deficits.[143]

MPP+Female and male Sprague-Dawley ratsPretreatment (30 min) with melatonin (10 mg/kg, i.p.). Unilateral injection of MPP+ (7.4 g) into the right SNc. Animals were subjected to an acute or chronic posttreatment with melatonin.Melatonin treatment reduced lipid peroxidation and protected against DAergic neuronal loss induced by MPP+.[144]
Hepatic mitochondria and striatal synaptosomesPreincubation with melatonin (10−6 to 10−3 M). Incubation with MPP+ (10−6 to 10−3 M).Melatonin prevented the inhibition of complex I induced by MPP+.[145]
Male Wistar ratsUnilateral injection of MPP+ (0.1 mol) into the right striatum. Melatonin (10 mg/kg, i.p.) was administrated 1 h before and 1, 3, and 5 h after MPP+ administration.Melatonin reduced the MPP+-induced DAergic toxicity and recovered the GSH levels.[146]
SH-SY5Y cellsPreincubation (4 h) with melatonin (200 M). Incubation (72 h) with MPP+ (1 mM). Melatonin reduced MPP+-induced mitochondrial DNA oxidative damage, accumulation of oxygen free radicals, generation of mitochondrial membrane potential collapse, and cell death.[147]
Cerebellar granule cellsCoincubation with MPP+ (200 M) and melatonin (1 mM) at the same time.Melatonin protected cell viability and prevented apoptosis. Melatonin also reduced cdk5 expression and the cleavage of cdk5-35 to cdk5-25 induced by MPP+.[148]
SK-N-SH cellsPreincubation (1 h) with melatonin (1 mM). Incubation with MPP+ (0.1 mM).Melatonin prevented the MPP+-induced phosphorylation of c-Jun, activation of caspase-3, DNA fragmentation factor 45 (DFF45), and DNA fragmentation.[149]
Adult Wistar ratsInjection of 1 L of 50 mM MPP+ into the right striatum. Melatonin (10 mg/mL, i.p.) was administrated 0, 1, 2, 3, 4, 24, 48, and 72 h after MPP+ injection.Melatonin protected DAergic neurons from apoptosis induced by MPP+. Melatonin recovered mRNA and protein expression of fibroblast growth factor 9 that was reduced by MPP+ injection.[150]

MPTP C57BL/6 miceSingle injection of MPTP (20 mg/kg, s.c.). Melatonin (10 mg/kg i.p.) was administrated 30 min prior to and every hour (for 3 h) after MPTP injection.Melatonin treatment prevented MPTP-induced lipid peroxidation and TH-positive neurons loss in striatum.[151]
Male C57BL/6 miceSingle injection of MPTP (15 mg/kg, s.c.). Melatonin (5 or 10 mg/kg i.p.), deprenyl (0.37 mg/kg), or deprenyl plus melatonin (0.37 mg/kg and 5 or 10 mg/kg) was administrated 30 min prior to MPTP.Melatonin was able to protect the mitochondrial complex I activity and the oxidative damage in nigrostriatal neurons. Melatonin treatment also potentiates the protective effect of deprenyl on DA levels and TH activity.[152]
Male C57BL/6 miceFour injections of MPTP (15 mg/kg, s.c.) with intervals of 2 h. After 24 h, the animals received three additional injections with the same dose and intervals. Melatonin (20 mg/kg s.c.) was administrated 1 h before the first injection of MPTP.Melatonin treatment prevented the MPTP-induced mitochondrial iNOS in striatum and SNc. Melatonin also protected complex I activity and inhibited lipid peroxidation. [153]
Rat astrocytoma cellPreincubation with melatonin (50, 100, and 200 M). Incubation with MPTP (400 M).Melatonin decreased the MPTP-induced oxidative and nitrosative stress, intracellular calcium, and activation of P-p38 MAPK. Melatonin also normalized the levels of inflammatory proteins, mRNA of proinflammatory cytokines, and NF-B.[154]
Male C57BL/6 miceTen injections of MPTP (15 mg/kg, i.p.) during 5 weeks (2 injections a week). Melatonin (5 mg/kg, i.p.) was administered 1 week before, 5 weeks during, and 12 weeks after MPTP treatment.Melatonin recovered mitochondrial respiration, ATP production, and antioxidant enzyme levels. Melatonin also protected against MPTP-induced DAergic neurons loss and locomotor activity deficits.[155]
Male Swiss miceFour injections of MPTP (20 mg/kg, i.p.) with 2 h between them. Eight days after MPTP injections, animals received L-DOPA/carbidopa (100/10 mg/kg/twice/day, p.o.) and/or melatonin (5 or 10 mg/kg/day, p.o.) for 8 weeks.Melatonin treatment recovered motor performance, striatal DA level, GSH, and antioxidant enzyme activities and reduced lipid peroxidation. Melatonin also improved the motor response to L-DOPA.[156]
Male BALB/c miceMPTP (30 mg/kg, i.p.) was administrated in two injections (16 h apart). Melatonin treatment (10, 20, and 30 mg/kg, i.p.) 30 min before MPTP administration, followed by four doses of melatonin, at every 10 h.Melatonin protected against the MPTP-induced TH-positive neurons loss in SNc and enhanced the effects of L-DOPA treatment.[157]
Embryos of zebrafishIncubation with MPTP (600 M). Incubation with melatonin (0.2 and 1.0 M) at the same time or after the MPTP treatment.Melatonin recovered motor behaviour of the embryos. Melatonin also restored gene expression and normal function of parkin/PINK1/DJ-1/MUL1 loop.[158]

RotenoneDrosophila melanogasterMelatonin (5 mM) and/or rotenone (125 M) were added to the feeding medium for 7 days.Melatonin treatment prevented motor deficits and neuronal loss.[159]
Male Sprague-Dawley ratsRotenone injection (6 g in 1 L) into the right SN. Melatonin (10, 20, and 30 mg/kg, i.p.) was administrated 30 min after rotenone injection and was given every 12 h for 4 days.Melatonin reduced the levels of hydroxyl radicals in the isolated mitochondria and protected GSH levels and antioxidant enzymes activities in SN that were changed by rotenone injection.[160]
Male Wistar ratsRotenone injection (2.5 mg/kg, i.p.) for 10 days. Melatonin (10 mg/kg, i.p.) was administrated for 28 days after the rotenone injection.Melatonin treatment protect TH-positive neurons in SNc and striatal levels of dopamine. Melatonin also inhibit the rotenone-induced depressant-like effect.[161]
Male Sprague-
Dawley rats
Three injections of rotenone (4.0 μg in 2.0 L/site) at three points along its rostrocaudal axis. Animals received melatonin (4.0 g/mL) in drinking water, one week before and nine weeks after rotenone injections.Melatonin treatment protected TH-positive neurons in striatum and SNc. Melatonin also inhibited the rotenone-induced loss in dopamine of SNc and apomorphine-induced rotations.[162]

ManebPC12 cellsIncubation (2 h) with melatonin (1 nM) and/or maneb (1 g/mL). Melatonin prevented the maneb-induced disruption of the mitochondrial transmembrane potential, activation of caspase-3/7, loss in cell viability, and aggregation of α-synuclein.[163]

Maneb plus paraquatMale Swiss miceTreatment with melatonin (30 mg/kg/day, i.p.) for 9 weeks. Treatment with maneb (30 mg/kg, i.p.) plus paraquat (10 mg/kg, i.p.) twice a week, for 9 weeks, 2 hours after melatonin injection.Melatonin treatment protected the maneb/paraquat-induced lipid peroxidation, TH-positive neurons degeneration, increased nitrite content and mRNA levels of cytochrome P-450 2E1, GSTA4-4 activity, and increased levels of glutathione-S-transferase, P-p53, Bax, and caspase-9.[164]

Lentiviral vectorMale Sprague-Dawley ratsInjection with lentiviral vectors encoding A30P mutant human α-synuclein (lenti-A30P) into the right SNc. Melatonin treatment (10 mg/kg/day, i.p.) 2 days before and 8 weeks after the injection of lenti-A30P.Melatonin treatment prevented the loss of TH-positive neurons induced by injection of lenti-A30P.[165]

6-OHDA: 6-hydroxydopamine; COX: cyclooxygenase; DA: dopamine; GDNF: glial cell-derived neurotrophic factor; GPx: glutathione peroxidase; GSH: reduced glutathione; GSTA4-4: glutathione S-transferase alpha 4; i.p.: intraperitoneal; iNOS: inducible nitric oxide synthase; MAPK: mitogen-activated protein kinases; MDA: malondialdehyde; MPP+: 1-methyl-4-phenylpyridinium; MFB: medial forebrain bundle; MPTP: 1-methyl-4-phenyl,1-1,2,3,6-tetrahydropyridine; NF-B: nuclear factor-B; PGE2: prostaglandins E2; s.c.: subcutaneous; SNc: substantia nigra pars compacta; SOD: superoxide dismutase; TH: tyrosine hydroxylase.