4.1. Ceftriaxone
Ceftriaxone, an old, third-generation widely used cephalosporin antibiotic, has been demonstrated to exert beneficial neuroprotective effects in preclinical models of several neurological diseases [
37]. Ceftriaxone can supress glutamatergic neuronal excitotoxicity, promote the expression of glutamate transporter-1, and enhance the reuptake of glutamate. In addition, it can bind to α-synuclein and supress its polymerization, regulate the expression of amyloid beta-related genes, and improve neurogenesis [
37]. Given the fact that glutamatergic excitotoxicity plays a pivotal role in the pathophysiology of PDD, it has been hypothesized that it could mitigate cognitive and behavioural deficits in animal models of PD
in vivo. Indeed, ceftriaxone could reverse behavioural deficits and enhance neurogenesis in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced rat models of PDD, since it was able to improve motor function, prevent working memory and object recognition deficits, and promote neurogenesis in the hippocampal dentate gyrus and substantial nigra of the animals [
38]. Interestingly, another study has indicated that ceftriaxone exerted synergistic effects with erythropoietin on the behavioural impairment and neuronal alterations of MPTP-induced rat models of PD, in terms of memory deficits, as well as the degeneration of nigrostriatal dopaminergic projections and the CA1 area of the hippocampus [
39]. Given these promising preclinical results, a double-blinded, randomized, placebo-controlled Phase II clinical trial is investigating the safety and efficacy of the use of ceftriaxone in patients with mild and moderate PDD (NCT03413384).
4.12. Agents for PDD under Investigation: An Update on Preclinical Evidence
Emerging preclinical evidence has revealed the therapeutic potential of several novel candidates for the treatment of cognitive impairment in PD. Gut microbiota and bile acid metabolism has been indicated to play an important role in PD and cognitive impairment in PD in particular [
59]. In this context, a recent study has shown that INT-777, a 6α-ethyl-23(S)-methyl derivative of cholic acid (S-EMCA), which acts as a Takeda G protein-coupled receptor-5 (TGR5) agonist could exert neuroprotective properties in MPTP-induced mouse models of PD in terms of cognitive and motor deficits, at least partially via the regulation of neuroinflammation and mitochondrial function in microglia [
60].
Neuropeptide S (NPS) and NPS receptor (NPSR) has been indicated to play a crucial role in PD pathophysiology. More specifically, NPS can enhance the release of dopamine release, inhibit oxidative damage, and suppress the dopaminergic neuronal loss in preclinical animal models of PD [
61]. NPS was able to improve memory function in MPTP-induced mouse models of PD [
62], suggesting its promising potential for PD-related cognitive impairment.
BDNF is neurotrophic factor critically involved in the molecular pathogenesis of neurodegenerative diseases, including AD and PD. A recent study indicated that BDNF overexpression via adeno-associated viruses (AAV) with BDNF gene injection was associated with improved cognitive performance in MPTP-induced mouse models of PD, which was accompanied by restoration of mitochondrial function and inhibition of dopaminergic neuronal loss [
63].
Furthermore, another study has shown that silibinin, a flavonoid that is derived from milk thistle (Silybum marianum) with hepatoprotective, antioxidative and neuroprotective properties, was able to attenuate cognitive deficits in MPTP-induced mouse models of PD. These effects were associated with reduced cellular apoptosis and a-synuclein aggregation in the hippocampus, as well as decreased oxidative stress and improved mitochondrial function [
64]. Hence, this agent represents another potential therapeutic candidate against PDD, which deserves further investigation.
Oral consumption of probiotics has been associated with improved both motor and non-motor symptoms in PD, such as constipation, depression and anxiety, via their implication in gut-brain axis regulation [
65]. Interestingly, the administration of the probiotic
Bifidobacterium breve could restore the abnormal synaptic plasticity in the hippocampus and facilitatate fear extinction in MPTP-induced mouse models of PD [
66]. In addition, the administration of the probiotic formulation SLAB51 could improve behavioral deficits and prevent dopaminergic neuronal loss in the substantia nigra pars compacta and striatum in 6-hydroxydopamine (6-OHDA)-induced mouse models of PD [
67], further supporting the role of probiotics in attenuating cognitive deficits in PD.
Another agent, osmotin, a adiponectin homolog that modulates the phosphorylation of 5' adenosine monophosphate-activated protein kinase (AMPK) through the adiponectin receptor 1 (AdipoR1), has been found to exert neuroprotective properties in preclinical models of PD. In particular, osmotin treatment was associated with better cognitive performance in a-synuclein transgenic and MPTP-induced mouse models of PD. The underlying mechanisms might involve the inhibition of α-synuclein accumulation via the upregulation of the AMPK/mammalian target of rapamycin (mTOR) signaling pathway and modulation of autophagy, as well as the regulation of neuroinflammation through its implication in mitogen-activated protein kinase (MAPK) pathway [
68].
Cordycepin, a small molecule derived from cordyceps sinensis, is also able to regulate neuroinflammation in MPTP-induced models of PD, by downregulating Toll-like receptor (TLR)/nuclear factor kappa light chain enhancer of activated B cells (NF-κB) pathway [
69]. It has also been demonstrated that cordycepin might exert beneficial effects on cognitive function in MPTP-induced models of PD, by modulating the adenosine A2A receptors andreversing the suppression of synaptic neurotransmission in the hippocampus [
70].
Moreover, administration of huperzine A, a plant-derived lycopodium alkaloid acting as a natural acetylcholinesterase inhibitor, was able to improve memory and learning ability of MPTP-induced murine models of PD, which was accompanied by prevention of dopaminergic degeneration, and modulation of inflammatory and apoptotic mechanisms [
71].
Fibroblast growth factor 21 (FGF21) has been shown to display several biological properties, such as anti-oxidant, anti-inflammatory and anti-apoptotic. FGF21 could prevent dopaminergic neuronal loss in the substantia nigra pars compacta, improve mitochondrial function and inhibit microglia activation in MPTP-induced mouse models of PD. Potential underlying mechanisms involved the upregulation of the AMPK/Peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1α) pathway [
72]. A recent study revealed that FGF21 treatment was associated with better motor and cognitive performance of MPTP-induced mouse models of PD, possibly by re-structuring the profile of gut microbiota, thus preventing the PD-related metabolic alterations in the gut [
73].
It has been indicated that Poloxamer 188, an amphipathic synthetic polymer, protects against MPTP-induced dopaminergic neuronal loss. Recently, this agent was also shown to exert beneficial effects against cognitive deficits in maneb- and paraquat-induced mouse models of PD, potentially by supressing inflammatory responses and microglia activation, and restoring hippocampal synaptic density [
74].
Ginsenoside Rb1, the active ingredient of
Panax ginseng, has been previously shown to attenuate motor impairment and prevent dopaminergic degeneration in MPTP-induced mouse models of PD, by upregulating the glutamate transporter GLT-1 and inhibiting glutamate excitotoxicity [
75]. In addition, Ginsenoside Rb1 could improve memory and spatial learning ability and enhance long-term potentiation (LTP), by upregulating the expression of postsynaptic density-95 (PSD-95) [
76].
Thioredoxin-1 (Trx-1), a redox protein, could also ameliorate memory and learning impairment in MPTP-induced mouse models of PD, by regulating dopamine D1 receptor expression and modulating the NMDAR/extracellular signal-regulated kinase (ERK1/2)/cAMP-response element binding protein (CREB) signaling pathway in the hippocampus [
77].
Tangeretin, a citrus flavonoid, has also been demonstrated to suppress neurodegeneration and neuroinflammation responses in MPTP-induced cognitive impairment in rat models. In particular, in this study, tangeretin could reduce neuronal cell death in the hippocampus, as well as the pro-inflammatory IL-1β, IL-6 and IL-2, and these effects were accompanied by improved memory function [
78].
Istradefylline, an antagonist of adenosine A2A receptor, shows promising effects on patients with advanced PD experiencing motor complications, in terms of reducing OFF episodes [
79]. Although the exact underlying mechanism of action is unclear, it is considered that A2A receptor antagonism may exert its effects via the modulation of gamma-aminobutyric acid (GABA) neurotransmission in the basal ganglia [
79]. Notably, apart from its impact on levodopa induced motor complications, istradefylline administration has been associated with both better motor and cognitive performance of MPTP-treated macaque models of PD, regarding attentional deficits and working memory [
80].
Eugenia uniflora, an extract of Brazilian purple cherry, has been associated with improved memory concerning short and long-term object recognition, social recognition and working memory of MPTP-induced rat models of PD [
81]. These behavioral effects were accompanied by the modulation of the BDNF/tropomyosin receptor kinase B (TrkB)/p75
NTR axis in the hippocampus, which is implicated in synaptic plasticity and neurotransmission. Hence, its clinical efficacy in PDD should be further explored in the future.