Next Article in Journal
Prescriptions of Antipsychotics in Younger and Older Geriatric Patients with Polypharmacy, Their Safety, and the Impact of a Pharmaceutical-Medical Dialogue on Antipsychotic Use
Previous Article in Journal
Local Glucocorticoid Administration Impairs Embryonic Wound Healing
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Lysophospholipids: A Potential Drug Candidates for Neurodegenerative Disorders

1
Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
2
Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Nagano 390-8621, Japan
3
Clinical Pharmacology Educational Center, Nihon Pharmaceutical University, Ina-machi, Saitama 362-0806, Japan
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(12), 3126; https://doi.org/10.3390/biomedicines10123126
Submission received: 22 October 2022 / Revised: 30 November 2022 / Accepted: 1 December 2022 / Published: 3 December 2022
(This article belongs to the Section Neurobiology and Clinical Neuroscience)

Abstract

:
Neurodegenerative diseases (NDs) commonly present misfolded and aggregated proteins. Considerable research has been performed to unearth the molecular processes underpinning this pathological aggregation and develop therapeutic strategies targeting NDs. Fibrillary deposits of α-synuclein (α-Syn), a highly conserved and thermostable protein, are a critical feature in the development of NDs such as Alzheimer’s disease (AD), Lewy body disease (LBD), Parkinson’s disease (PD), and multiple system atrophy (MSA). Inhibition of α-Syn aggregation can thus serve as a potential approach for therapeutic intervention. Recently, the degradation of target proteins by small molecules has emerged as a new therapeutic modality, gaining the hotspot in pharmaceutical research. Additionally, interest is growing in the use of food-derived bioactive compounds as intervention agents against NDs via functional foods and dietary supplements. According to reports, dietary bioactive phospholipids may have cognition-enhancing and neuroprotective effects, owing to their abilities to influence cognition and mental health in vivo and in vitro. However, the mechanisms by which lipids may prevent the pathological aggregation of α-Syn warrant further clarification. Here, we review evidence for the potential mechanisms underlying this effect, with a particular focus on how porcine liver decomposition product (PLDP)-derived lysophospholipids (LPLs) may inhibit α-Syn aggregation.

1. Overview

Neurodegeneration has been identified as the pathophysiological hallmark in most brain-related disorders. Many neurodegenerative diseases (NDs) involve the misfolding and aggregation of specific proteins into abnormal, toxic species [1,2]. Early diagnosis is essential for treatment planning and ensuring that the right support can be provided to patients and their families. However, therapeutic strategies for NDs present unique challenges for drug development. Although no curative treatment is available for NDs, the range of therapeutic and supportive options are expanding. The most common NDs are Alzheimer’s disease (AD), Lewy body disease (LBD), Parkinson’s disease (PD), and multiple system atrophy (MSA) [3]. The observation of amyloid-like protein aggregation in the brains of patients with NDs was first described in the 1960s, and thus far, three-dimensional structures of several of these aggregates have been elucidated [4,5]. These structural data have been utilized for the design of inhibitors of amyloid-like aggregation proteins [6,7,8]. Since decades, the presence of amyloids in NDs has been associated exclusively with pathologies [9,10]. Additionally, strong evidence indicates that the activation of inflammatory processes is a hallmark of NDs. Elevated levels of pro-inflammatory cytokines and chemokines, as well as activated microglia and astrocytes, are found in the brains of patients with AD, even at very early stages of the disease [11]. Ageing affects homeostatic processes that protect against protein misfolding and is associated with an increase in oxidative stress, neuroinflammation, and mitochondrial-lysosomal dysfunction, which have been shown to directly activate microglia cells and astrocytes [12,13,14,15]. It has been argued that extracellular amyloid-β peptide (Aβ) in AD activates microglia and astrocytes, which in turn release tumor necrosis factor alpha (TNF-α) and other cytokines. TNF-α signaling enhances Aβ production and Aβ-induced neuroinflammation [16]. Neuroinflammation, oxidative stress, and mitochondrial dysfunction have all been linked to the progression of NDs [11,17,18]. The neuroprotective effects of resveratrol reportedly stem from its ability to inhibit microglial activation and regulate neuroinflammation [19,20,21]. Our recent study demonstrated that porcine liver decomposition product (PLDP) could improve cognitive function in elderly adults by providing a rich source of phospholipids (PLs) and lysophospholipids (LPLs) [22]. Notably, the main constituent lipids of synaptic vesicles include cholesterol and phospholipids [23]. Although the concentration of PLDP-derived lipids in LPLs is not sufficient to yield cognitive benefits, LPLs may confer anti-neuroinflammatory effects [24,25]. Previous research suggests that LPLs have served not only as structural components of biological membranes but also as biologically active molecules [26]. LPLs influence a plethora of processes, including neurogenesis in the central nervous system (CNS) [27,28,29]. Growing interest in the involvement of extracellular LPLs in the pathology of NDs increasingly indicates that these small molecules may have therapeutic potential for NDs.

2. α-Syn Protein, Aggregates, and Aggregate Inhibitors

2.1. α-Syn in Regulating Brain Physiology

In the brain, α-Syn is found mainly in nerve cells in specialized regions called presynaptic terminals [30]. In neurons, α-Syn exists in an equilibrium between cytosolic and membrane-bound states. α-Syn exists as a natively unfolded monomer in the cytosol, whereas membrane-bound α-Syn adopts an α-helical conformation [31]. α-Syn is highly expressed in the neuronal cell bodies of early-stage PD-affected regions, such as the olfactory bulb, dorsal motor nucleus of the vagus, and substantia nigra [32]. Notably, the expression of α-Syn is developmentally regulated. α-Syn mRNA expression begins in late embryonic stages in rodents and peaks in the first few postnatal weeks, following which it is reduced [33]. However, the mechanism by which α-Syn reaches the synapse and its preference for synaptic vesicle membranes remain unclear. Different therapeutic strategies have been developed for reducing brain dysfunction due to protein aggregates, including direct targeting of misfolded proteins [34,35]. NDs are characterized by the misfolding and aggregation of specific proteins [36]. The α-Syn protein is highly expressed in the mammalian nervous system, and the abnormal buildup of α-Syn forms masses known as Lewy bodies [37,38,39,40]. Interestingly, α-Syn oligomers have been found in exosomes, membrane nanovesicles secreted by cells in the CNS [41]. In the CNS, α-Syn is expressed at high levels in neurons of different brain areas such as the neocortex, hippocampus, substantia nigra, thalamus, and cerebellum [42]. The aggregation of α-Syn plays a central role in PD, as well as in other synucleinopathies [43,44]. Abnormal α-Syn yields neuronal cell inclusions and axonal spheroids, as well as oligodendrocyte aggregates (known as glial cytoplasmic inclusions) that accumulate in MSA, rendering α-Syn fibrils important therapeutic targets in PD and related synucleinopathies [45]. PD is the second most common ND, with a global prevalence of over 6 million [46]. Although the exact cause of PD remains unknown, α-Syn has emerged as the major molecule involved in PD pathogenesis. A pathological hallmark of PD is the presence of Lewy bodies, which are intracellular inclusions of aggregated α-Syn [47]. Lewy body dementia (dementia with detectable Lewy bodies) is the third most common types of degenerative dementia. Lewy body dementia causes progressive declines in mental function, and leads to visual hallucinations, movement disorders, cognitive issues, sleep difficulties, attention fluctuations, and depression [48].

2.2. Structure of α-Syn

The α-Syn protein, which is 140 amino acids long, comprises three domains: an N-terminal, the non-amyloid-β-component (NAC), and C-terminal domains [49]. All known point mutations in the gene encoding α-Syn (SNCA) that are associated with PD are in the N-terminal domain, which is predicted to form an amphipathic α-helix. Additionally, it has a lysine-rich, highly conserved motif similar to that of lipid-binding motifs, and it is responsible for the ability of α-Syn to associate with vesicles and membranes [50,51]. The N-terminal region of α-Syn contains a highly conserved KTKEGV hexamer motif, which yields variations in its surface hydrophobicity [52,53]. Such a periodic motif is characteristic of the amphipathic lipid-binding α-helical domains of apolipoproteins, which have been assigned to subclasses according to their unique structural and functional properties [54,55]. NACore is a peptide fragment spanning residues 68–78 of the α-Syn protein. A previous report suggested that the presence of lipids inhibits NACore (GAVVTGVTAVA) fibril formation. Recently, cryogenic transmission electron microscopy (Cryo-TEM) revealed the presence of non-fibrillar clusters among NACore fibrils formed in the presence of linoleic acid [56]. This effect was additionally observed with PLs and was particularly pronounced with linoleic acid [57]. This inhibitory effect has been attributed to a prolonged lag phase during fibril formation [56]. The α-Syn C-terminal domain contains multiple negatively charged residues, as well as a serine and three tyrosine residues that can be phosphorylated, which may impact α-Syn structure, membrane binding, aggregation, and toxicity [58,59]. The α-Syn protein does not readily aggregate spontaneously [60,61,62], and aggregation is affected by environmental conditions [63,64]. The binding of this protein to lipid membranes offers a possible interface for primary nucleation events to initiate the toxic cascade of α-Syn aggregation. However, previous reports have indicated that α-Syn interacts with certain PLs, which may transform it into a helical conformation [65]. Afitska et al. observed a strong correlation between the induction of α-helix conformation in α-Syn and the inhibition of fibril formation [66]. Thus, helical, membrane-bound α-Syn is unlikely to contribute to aggregation and fibrillation [67]. Small oligomeric forms of α-Syn have been reported to preferentially associate with lipid droplets and cell membranes [68]; α-Syn binds preferentially to small unilamellar vesicles (less than 100 nm in diameter) containing acidic PLs but not to vesicles with a net neutral charge [69]. Nevertheless, according to other reports, α-Syn strongly binds to large unilamellar vesicles with either non-ionic or zwitterionic lipid headgroups [70]. In solution, monomeric α-Syn lacks any secondary structure and specific intrachain interactions and undergoes self-assembly without external additives. However, numerous early epidemiological studies correlated exposure to metals, polyamines, and glycosaminoglycans with α-Syn aggregation [71,72,73,74]. While metal ions can directly cause brain damage, their effect on PD is based on reactive oxygen species (ROS)-dependent effects and a direct influence on α-Syn aggregation [75,76]. The monomeric form of α-Syn is non-toxic, and it does not activate ROS production in neurons [77,78]. In microglia, ROS are generated primarily by NADPH oxidase 2 (NOX2), and activation of NOX2 in disease-associated microglia is associated with damage-associated molecular patterns signaling and inflammation, especially in cerebrovascular diseases [79]. α-Syn-induced activation of microglial NOX2 has been implicated in PD [80]. Activated microglia and accumulation of proinflammatory factors are present in the substantia nigra and striatum of patients with PD [81]. NOX2 is subsequently recognized to be critical for α-Syn-induced microglial activation and neurodegeneration since pharmacological inhibition or gene deletion of NOX2 attenuates α-Syn-induced microglial activation and related neurotoxicity [82,83]. Phagocytosis of α-Syn, with subsequent activation of NOX, plays a central role in the pathogenesis of microglial activation and associated neurotoxicity induced by aggregated α-Syn [14]. α-Syn has been shown to aggregate into various oligomeric and fibrillary forms; soluble, misfolded α-Syn aggregates are neurotoxic and can spread disease via prion-like transfer [84]. α-Syn aggregation is a hallmark of PD, and different forms of the protein can be used for developing quantitative disease models that reproduce the pathological features of this disorder.

2.3. Phosphorylation and Ubiquitination of α-Syn

α-Syn is most commonly phosphorylated in serine and tyrosine residues. In Lewy bodies, it is typically phosphorylated at S129 and S87 [62,85,86]. Phosphorylation at S129 is closely linked to PD, increasing from 5% in healthy brains to approximately 90% in Lewy bodies [87,88]. However, the reason behind extensive phosphorylation in the pathology of LBD, such as PD and DLB, is unclear. In vitro studies have offered conflicting conclusions regarding the effect of S129 on α-Syn aggregation [89]. Increased Ca2+ influx under mitochondrial impairment has been reported to stimulate a change in the solubility of α-Syn proteins from normally soluble to insoluble and induce Ser129 phosphorylation to generate a proteasomal degradation signal [90]. Because Ser129 phosphorylation plays a role in removing excess amounts of α-Syn, α-Syn aggregates may continuously undergo phosphorylation [89]. It additionally interacts with various proteins, including lipid membranes and fatty acid-binding protein 3, as well as metal ions [75,76,91,92]. In addition, in brain homogenates from diseased human brains and transgenic animals, most S87-P α-Syn was detected in the membrane fractions [93]. S87 is one of the few residues and phosphorylation sites located within the NAC region. A previous study suggested that S87 phosphorylation alters the conformation of membrane-bound α-Syn and decreases its affinity to lipid vesicles, probably by destabilizing the helical conformation and decreasing the lipid-binding affinity of the protein around the phosphorylation site [93]. The cryo-TEM structure of α-Syn fibrils, S87, has been reported to face the outside of the fibril; hence, it remains accessible for disease-associated modification in α-Syn fibrils [45]. Ubiquitination of aggregated or protein filaments has been implicated in the pathogenesis of several NDs [94]. Lewy bodies contain ubiquitin-α-Syn; therefore, they have shown immunoreactivity to anti-ubiquitin antibodies [95]. α-Syn contains eight lysine residues that can be ubiquitinated, and ubiquitin contains multiple internal lysine residues, which can form polyubiquitin chains [96,97]. The presence of ubiquitin in intracellular inclusions in synucleinopathies suggests that abnormally aggregated or misfolded proteins are targeted for ubiquitination in these inclusions in a similar fashion to tau in neurofibrillary tangles of AD [98].

2.4. Small Molecule Modulator of α-Syn Aggregation and Drug Development

Approaches for targeting α-Syn monomers have been designed to reduce protein expression, prevent aggregate formation, or promote protein degradation [99]. Studies have shown that α-Syn displays variable kinetics in vitro, and elucidating its fibrillation kinetics would provide valuable information on the molecular events surrounding this process, as well as data required for the screening of small molecule inhibitors against α-Syn aggregation in vitro. Aggregation of α-Syn reportedly affects dopamine metabolism, increases oxidative stress due to mitochondrial dysfunction, disrupts synaptic function, and impairs vesicular trafficking [100]. Major research efforts have been made towards understanding the molecular basis of PD, with the goal of developing therapies for delaying disease progression. Several current therapeutic strategies for PD aim to reduce the neuronal load of aggregated α-Syn. However, the agents used exhibit certain limitations, including degradation by proteases and inefficient crossing of the blood–brain barrier (BBB). Oligomerization inhibitors targeting α-Syn have been widely investigated [101,102,103,104]. For instance, the small molecule UCB0599 (Neuropore Therapies, Inc.) has been shown to cross the BBB with low toxicity in control subjects; it has performed successfully in phase I clinical trials [105]. UCB0599 acts in the first step of the α-Syn aggregation cascade by preventing misfolding and the formation of α-Syn aggregates on lipid membranes [105]. Patients with early-stage PD are currently enrolled in the phase II ORCHESTRA trial for UCB0599, which is expected to finish in October 2023. Small molecule inhibitors are relatively low molecular weight compounds interacting with α-Syn or their aggregation intermediates, altering the amyloidogenic pathway. Treatment approaches using orally available small chemicals appear more suitable for NDs than antibodies and oligonucleotide therapeutics [106], which may potentially have poor pharmacokinetic and pharmacodynamic properties; however, the CNS bioavailability of small molecule therapies requires improvement.

3. Effects of LPLs on Cognitive Decline

3.1. Dietary Sources of LPLs

Because of their superior emulsification properties, LPLs have numerous applications in the food, cosmetic, and pharmaceutical industries [107]. However, their properties depend strongly on the fatty acid component present and the specific polar head bound to the glycerol backbone [108]. LPLs, as promising feed additives, have been widely used to supplement farm animals diets to improve growth performance, feed efficiency, and dietary fat absorption [109]. LPL supplementation can increase the apparent total tract digestibility of animals [110]. PLs are widely available in the intestinal lumen after eating, and their hydrolysis is catalyzed by phospholipase A2 (PLA2). LPLs (particularly LPC), the digestive products of PLs, have direct roles in mediating chylomicron assembly and secretion [111]. LPC is present in the plasma circulation at relatively high levels and includes species containing both saturated and unsaturated fatty acids [112]. Interestingly, the intake of PL-enriched diets during postnatal brain development was found to increase the number of striatal circuits [113]. In PD, dopaminergic neurons are progressively degenerated, leading to striatal dopamine depletion and movement deficits. α-Syn is intimately involved in the pathogenesis of PD, and has been implicated in the regulation of dopamine synthesis, release, and reuptake [114]. It is possible that dietary PLs have several benefits in health, including improvements in cognition across the lifespan.

3.2. Cognitive Function and LPLs

Mild cognitive impairment (MCI) is characterized by impairment of certain cognitive functions, as defined by Petersen et al. [115], and a diagnosis of mild neurocognitive disorder essentially comprises MCI [116]. It can occur in a subtle form, such as MCI, in the early stages of PD in up to 25% of newly diagnosed patients [117,118,119]. MCI is a distinct stage of cognitive loss that falls between the expected cognitive decline of physiological aging and the more serious loss of mental abilities associated with dementia. It is characterized by impairments in memory, language, thinking, or judgment that are severe enough to interfere with daily life [120]. Individuals with MCI have an increased risk of developing dementia caused by AD or other neurological conditions [121]. Growing evidence supports the hypothesis that dietary factors may play a role in healthy aging, including exertion of a protective effect against age-related cognitive decline [122,123]. The concept of functional foods was first introduced in Japan, a country with a long history of using foods for their health benefits [124]. The market for functional food ingredients covers the sale of functional food ingredients containing bioactive compounds and ingredients used in manufacturing functional food products [125]. The ingredients in functional foods provide health benefits, and some of them include supplements or other additives. Functional foods and bioactive compounds can strongly intensify the therapeutic efficacy of certain drugs by influencing different pathways [126]. A new understanding of these interactions is emerging owing to intense research on nutraceuticals in diseases, including NDs [127]. For example, Yuyama et al. showed that glucosylceramides (GlcCer) from konjac extract are linked to the attenuation of amyloid-like protein in the CNS [128]. In the brains of GlcCer-treated mice, decreased levels of several inflammatory cytokines were accompanied by the recovery of impaired synaptic densities [129]. Future studies need to evaluate GlcCer and examine their relationship with α-Syn in the brain to determine the possible use of plasma GlcCer as a prognostic biomarker of disease progression. Several dietary components including PLs, plasmalogens, ω3 fatty acid, carotenoids, vitamins, and phenolic compounds have been identified to affect cognitive functions [130]. Many studies indicate that neuroinflammation plays a fundamental role in the progression of the neuropathological changes that characterize NDs [131,132,133]; microglia are the main cellular effectors of this process [134]. Indeed, neuroinflammation is initiated by microglia, which are resident immune cells of the CNS. Once activated, microglia can synthesize and release several neurotrophic factors and antioxidants to withstand the pathological progression of NDs [135].

3.3. PLDP and LPLs

In previous studies, over 50% of patients with PD without dementia demonstrated cognitive alterations and 20% primarily exhibited memory deficits [136]. We previously reported that PLDP induces a significant increase in the Hasegawa’s dementia scale-revised (HDS-R) score and the Wechsler memory scale (WMS) score in a randomized, double-blind, placebo-controlled study in healthy humans [22]. PLDP was recently approved as a food with functional claims (FFC) in Japan. FFC was introduced in April 2015 for increasing the availability of products that are clearly labeled with certain health functions [137]. Oral administration of PLDP has been shown to enhance visual memory and delayed recall in healthy adults [22]. Notably, the oral route is the most common and preferred method of drug administration for several reasons, such as non-invasiveness, patient compliance, and convenience of drug administration [138]. These results suggest that PLDP may help improve brain function. Interestingly, PLDP is a rich source of PLs, and composition analysis of PLDP revealed that the most abundant PLs and LPLs belonged to the phosphatidylcholine (PC) and phosphatidylethanolamine (PE) classes [25,139]. LPLs influence signaling, proliferation, neural activity, and inflammation to mediate various important processes, including the pathogenesis of cerebral ischemia, vascular dementia, and AD [25,140,141,142]. However, no direct evidence indicates that any PLDP component improves cognitive function, and the effect of PLDP components on cognitive function remains unclear. Our recent study identified novel cooperative actions of LPLs that inhibit IL-6 expression and the accumulation of intracellular ROS in microglia after lipopolysaccharide (LPS)-induced neuroinflammation [25]. This activation was significantly inhibited by lysophosphatidylcholine (LPC) and lysophosphatidylethanolamine (LPE), suggesting that these molecules exert significant protective effects against LPS-induced inflammation (Figure 1). This finding is important considering that microglia-mediated neuroinflammation is regarded as a pathological mechanism in many NDs, such as AD and LBD, and is a pivotal event accelerating cognitive or functional decline. Furthermore, dietary LPC and docosahexaenoic acid (DHA) have been reported to efficiently increase DHA levels in the brain, improving brain function in adult mice [143]. This report describes a novel nutraceutical approach for preventing and treating neurological diseases (such as AD) associated with DHA deficiency.

4. LPLs and NDs

4.1. Neuroinflammation and LPLs

PLs are increasingly recognized for their roles in neural function in the brain [144,145,146]. LPLs are glycerophospholipids that lack one acyl chain and possess only one acylated hydroxyl group on the glycerol backbone, rendering them considerably more hydrophilic than diacylglycerol lipids [26,108]. LPLs are found only in small amounts in biological cell membranes in the form of membrane-derived signaling molecules produced by phospholipase A1 (PLA1), PLA2, and phospholipase D (PLD) enzymes [108]. The cell membrane comprises phospholipids, with PC and PE being the most abundant [24,139]. PLA2 catalyzes the hydrolysis of cell membrane-associated phospholipids to form free fatty acid and LPLs. In cells, these LPLs are intermediate precursors for biosynthesis of other cellular lipids and are thus present at low intracellular concentrations. However, multiple LPLs with varying head groups and hydrocarbon tails are highly abundant in extracellular environments such as the plasma and interstitial fluids [147]. LPLs and their receptors have been detected in a wide range of tissues and cell types, indicating their importance in many physiological processes, including those in the nervous system [148,149]. Our previous study suggested that PLDP, a rich source of LPLs, improves cognitive function in old age [25,139]. LPCs are the major class of LPLs in PLDP, followed by LPE, LPI, and lysophosphatidylserine (LPS). Additionally, traces of lysophosphatidic acid (LPA) have been detected in PLDP [139]. LPLs are amphipathic molecules composed of three sections: a diglycerol group, a hydrophilic head consisting of a charged phosphate moiety, and a small organic molecule covalently bound to the phosphate. The headgroup charge apparently contributes to both strength and specificity in protein interactions. The ability of α-Syn to interact with membrane PLs was recognized after the protein was discovered to be a component of Lewy bodies in the brain [150]. Several studies indicate that membrane interactions promote the folding of α-Syn into amyloid-like structures, which aligns with the direct observation of membranes associated with pathological protein aggregates in many NDs [151]. Furthermore, our recent data showed that the LPS-mediated inflammatory response, including the increase in microglial cytokine production, was suppressed by LPC or LPE exposure. In addition, we observed a synergistic effect between LPC and LPE in LPS-treated microglial cells [25]. LPLs have advantages as CNS-targeting drugs because they are highly lipid-soluble, and they may exhibit good bioavailability. This finding is important considering the role that microglia-mediated neuroinflammation plays in promoting functional decline in many NDs [152].

4.2. LPLs and the Brain

PLs and LPLs can transform into each other through the “Lands cycle” to maintain lipid homeostasis [153]. Some lipids, such as LPLs, exhibit neurotransmitter and/or neuromodulatory function, and could act as neuroprotective agents [154,155]. Mouse brain phospholipids have been reported to be highly enriched with long-chain polyunsaturated fatty acids [156,157]. Recent studies show that LPC is the preferred carrier of polyunsaturated fatty acids across the BBB into the brain [158]. The concentration of LPC in the blood plasma of healthy individuals usually ranges from 200 to 300 μM [112]. LPC is produced by the hydrolysis of PC by PLA2 (involving the removal of a fatty acid group at the sn-2 position) or by the lecithin cholesterol acyltransferase (LCAT) reaction. In a Drosophila model, PLA2G6 dysfunction, which leads to PARK14-related familial Parkinson’s disease, has been reported to damage the remodeling pathway of phospholipids and induce α-Syn aggregation through alteration of the binding affinity between α-Syn and the synaptic membrane [159]. In circulation, LPC has a short half-life due to rapid degradation, which prevents impairment of various vascular functions [148]. Presently, activated platelets are believed to release LPLs such as LPC, LPE, and LPS, which are then converted to LPA by the lyso-PLD present in the serum [160]. LPC reportedly exerts potent anti-aggregatory effects on platelets [161]. Platelet dysfunction is common during NDs, and impaired platelet function is a feature of several NDs, including AD, PD, Huntington’s disease (HD), amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), and prion diseases [162]. A recent study confirmed that the modification of lipoproteins by secretory phospholipases inhibited platelet activation and aggregation. The authors identified that LPC plays an essential role in these effects [163]. LPC is increasingly recognized as a key factor positively associated with NDs [164,165], with plasma levels of LPC reported to be decreased in patients with AD [158]. Adults with AD have presented lower LPC concentrations in the blood plasma, cerebrospinal fluid (CSF), and brain tissue than able-bodied individuals [166]. LPC consists of a phosphocholine headgroup and glycerol backbone linked to a variable fatty acid group, bound at either the sn-1 or sn-2 position. In tissues and plasma, polyunsaturated fatty acids (PUFAs) are predominantly bound at the sn-2 position of LPC, whereas saturated fatty acids (SFAs) are typically bound at the sn-1 position [167]. Plasma fatty acids supplied to the brain are derived from two main pools: plasma non-esterified fatty acids and those esterified in the form of LPC. Previous studies have demonstrated that, compared to free fatty acids (FAs), FAs bound to LPCs are more efficiently transported across the BBB into the brain [168,169]. Major facilitator superfamily domain-containing 2A (MFSD2A; currently known as human sodium carbonate electrogenic LPC symporter 1) is an orphan transporter that has been shown to act as a specific receptor for LPC. LPCs transport long-chain PUFAs (such as DHA) across the BBB [170]. Other studies have shown that the brain can only synthesize a few fatty acids; thus, most fatty acids must enter the brain from the blood, passing through the BBB [171]. However, cholesterol and lipoproteins cannot cross the BBB under normal physiological conditions [170,171]. For a small molecule drug to cross the BBB in pharmacologically significant amounts, the molecule must have dual molecular characteristics, namely, a molecular mass of 400–500 Da and high lipid solubility. Therefore, we hypothesize that LPLs may be used for developing an oral, small molecule inhibitor of α-Syn misfolding for slowing disease progression.

4.3. LPLs and Therapeutic Potential for NDs

Our recent report proved that LPC16:0, LPC18:0, LPC18:1, and LPE16:0 (known components of PLDP extracted lipids) strongly inhibit α-Syn aggregation [172] (Figure 1). When α-Syn was co-incubated with LPLs, thioflavin-T (an amyloid-binding dye) fluorescent emissions declined remarkably, indicating decreased fibril formation [172]. The role of α-Syn amyloid fibrillation has been recognized in several neurological diseases including PD. In early stages, fibrillation occurs by a transition from a helical structure to extended states in monomeric α-Syn, followed by the formation of β-sheets [173]. This α-helix to β-sheet transition promotes the formation of amyloid fibrils by generating unstable and temporary α-Syn configurations. α-Syn shows variable kinetics in vitro and clarifying its fibrillation kinetics would be valuable for to studying molecular events and screening small molecule inhibitors against α-Syn aggregation [174]. A previous study suggested that several LPLs have a chaperone-like function in protein folding [175]. Monomeric α-Syn is the predominant species in the cytoplasm of cells, and are excreted in cells that do not form a synaptic compartment. However, synaptic membrane binding may be crucial for temporal higher-order multimeric conformation, and PLs appear to have chaperone-like characteristics during this process. These effects could contribute to the neuroprotective effects of LPLs. In the future, we aim to identify functional LPLs contained in PLDP-derived lipids, confirm their clinical effects, and develop new drugs for treating NDs. In particular, we plan to evaluate the efficacy and safety of LPLs using an in vivo model for drug discovery against synucleinopathy. In summary, dietary lipids were originally believed to indirectly affect the brain through their effects on cardiovascular physiology, but are gaining recognition for their direct actions on the nervous system [176]. Further studies are warranted to investigate the role of LPLs in regulating cognition, and the potential for these lipids to reverse age-associated cognitive decline in vivo.

5. Conclusions and Future Perspectives

A growing number of publications report that lipids interact with α-Syn during aggregation in NDs. Amyloids are aggregates of associated proteins, and their formation is involved in several diseases often associated with older individuals. The molecular mechanisms influencing the formation of amyloids remain poorly understood. However, amyloid formation in the physiological environment markedly appears in the presence of a large variety of molecules, such as lipids. Unfortunately, no treatment is currently available for curing NDs or altering their progression. However, numerous new treatments are under clinical investigation. Therapeutic drug development requires a deep understanding of the structural properties of α-Syn monomers, oligomers, and fibrils, both in vitro and in vivo. We recently identified functional LPLs within PLDP-derived lipids (PEL) [25]. NDs lack satisfactory treatments primarily because the BBB hinders the passage of drugs from the bloodstream to the brain; this has motivated the development of novel strategies for neuro-drug delivery into the CNS. Thus, drugs need to be modified to improve their delivery. We propose that LPLs are highly attractive research targets in terms of biological activity and possible applications (Figure 2). Additionally, various features of cell membranes (such as lipid composition, charge, curvature, and lipid packing) can modulate the binding of α-Syn to them. We argue that the observed inhibitory effect on fibril formation is due to the association of α-Syn oligomers and LPLs at the early stage of the aggregation process. An important aspect of this mechanism is that it is non-monomeric α-Syn structures that associate with the LPL aggregates. Similar mechanisms of action could be relevant in amyloid formation occurring in vivo, where aggregation occurs in a lipid-rich environment [56]. Under normal conditions, α-Syn adopts an α-helical secondary structure when bound to lipid interfaces, which might be important for explaining differences in how LPLs affect the amyloid formation of different proteins. The ability of LPLs to bind directly to α-Syn and inhibit its aggregation, thereby blocking cell-to-cell propagation of the aggregates, suggests that LPLs may serve as promising therapeutic agents for preventing NDs. Therefore, further studies are necessary for determining the main phospholipids contained within PEL and their effects.

Author Contributions

T.K., H.H., Y.M. and T.T. conceived and designed the study. T.K. and T.T. acquired the data. T.K. and T.T. analyzed and interpreted the data. T.K. and T.T. wrote the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Japan Society for the Promotion of Science KAKENHI grant (grant number 22H03516) awarded to T.T.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

All authors have no conflicts of interests to declare.

Abbreviations

α-Synα-synuclein
BBBblood-brain barrier
CNScentral nervous system
DPLsdiacylphospholipids
LPLs lysophospholipids
PLDPporcine liver decomposition product
LBLewy body
MCImild cognitive impairment
PCphosphatidylcholine
LPAlysophosphatidic acid
LPElysophosphatidylethanolamine
LPClysophosphatidylcholine
LPSlipopolysaccharide
ROSreactive oxygen species

References

  1. Bouvier-Müller, A.; Ducongé, F. Nucleic acid aptamers for neurodegenerative diseases. Biochimie 2018, 145, 73–83. [Google Scholar] [CrossRef] [PubMed]
  2. Calabrese, G.; Molzahn, C.; Mayor, T. Protein interaction networks in neurodegenerative diseases: From physiological function to aggregation. J. Biol. Chem. 2022, 298, 102062. [Google Scholar] [CrossRef] [PubMed]
  3. Moussaud, S.; Jones, D.R.; Moussaud-Lamodière, E.L.; Delenclos, M.; Ross, O.A.; McLean, P.J. Alpha-synuclein and tau: Teammates in neurodegeneration? Mol. Neurodegener. 2014, 9, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Cohen, A.S.; Calkins, E. Electron Microscopic Observations on a Fibrous Component in Amyloid of Diverse Origins. Nature 1959, 183, 1202–1203. [Google Scholar] [CrossRef]
  5. Merker, H.-J.; Shibolet, S.; Sohar, E.; Gafni, J.; Heller, H. Periodic Cross-banding in Amyloid Filaments. Nature 1966, 211, 1401–1402. [Google Scholar] [CrossRef]
  6. Seidler, P.M.; Boyer, D.R.; Rodriguez, J.A.; Sawaya, M.R.; Cascio, D.; Murray, K.; Gonen, T.; Eisenberg, D.S. Structure-based inhibitors of tau aggregation. Nat. Chem. 2018, 10, 170–176. [Google Scholar] [CrossRef]
  7. Griner, S.L.; Seidler, P.; Bowler, J.; A Murray, K.; Yang, T.P.; Sahay, S.; Sawaya, M.R.; Cascio, D.; A Rodriguez, J.; Philipp, S.; et al. Structure-based inhibitors of amyloid beta core suggest a common interface with tau. eLife 2019, 8, e46924. [Google Scholar] [CrossRef]
  8. Balasco, N.; Diaferia, C.; Morelli, G.; Vitagliano, L.; Accardo, A. Amyloid-Like Aggregation in Diseases and Biomaterials: Osmosis of Structural Information. Front. Bioeng. Biotechnol. 2021, 9, 641372. [Google Scholar] [CrossRef]
  9. Soto, C.; Pritzkow, S. Protein misfolding, aggregation, and conformational strains in neurodegenerative diseases. Nat. Neurosci. 2018, 21, 1332–1340. [Google Scholar] [CrossRef]
  10. Wells, C.; Brennan, S.; Keon, M.; Ooi, L. The role of amyloid oligomers in neurodegenerative pathologies. Int. J. Biol. Macromol. 2021, 181, 582–604. [Google Scholar] [CrossRef]
  11. Currais, A.; Fischer, W.; Maher, P.; Schubert, D. Intraneuronal protein aggregation as a trigger for inflammation and neurodegeneration in the aging brain. FASEB J. 2017, 31, 5–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Van Den Berge, N.; Ferreira, N.; Mikkelsen, T.W.; Alstrup, A.K.O.; Tamgüney, G.; Karlsson, P.; Terkelsen, A.J.; Nyengaard, J.R.; Jensen, P.H.; Borghammer, P. Ageing promotes pathological alpha-synuclein propagation and autonomic dysfunction in wild-type rats. Brain 2021, 144, 1853–1868. [Google Scholar] [CrossRef] [PubMed]
  13. Su, X.; Maguire-Zeiss, K.A.; Giuliano, R.; Prifti, L.; Venkatesh, K.; Federoff, H.J. Synuclein activates microglia in a model of Parkinson’s disease. Neurobiol. Aging 2008, 29, 1690–1701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Zhang, W.; Wang, T.; Pei, Z.; Miller, D.S.; Wu, X.; Block, M.L.; Wilson, B.; Zhang, W.; Zhou, Y.; Hong, J.-S.; et al. Aggregated α-synuclein activates microglia: A process leading to disease progression in Parkinson’s disease. FASEB J. 2005, 19, 533–542. [Google Scholar] [CrossRef] [PubMed]
  15. Chou, T.-W.; Chang, N.P.; Krishnagiri, M.; Patel, A.P.; Lindman, M.; Angel, J.P.; Kung, P.-L.; Atkins, C.; Daniels, B.P. Fibrillar α-synuclein induces neurotoxic astrocyte activation via RIP kinase signaling and NF-κB. Cell Death Dis. 2021, 12, 756. [Google Scholar] [CrossRef] [PubMed]
  16. Minter, M.R.; Taylor, J.M.; Crack, P.J. The contribution of neuroinflammation to amyloid toxicity in Alzheimer’s disease. J. Neurochem. 2016, 136, 457–474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Tsukahara, T.; Sahara, Y.; Ribeiro, N.; Tsukahara, R.; Gotoh, M.; Sakamoto, S.; Handa, H.; Murakami-Murofushi, K. Adenine nucleotide translocase 2, a putative target protein for 2-carba cyclic phosphatidic acid in microglial cells. Cell. Signal. 2021, 82, 109951. [Google Scholar] [CrossRef]
  18. Swerdlow, R.H. Mitochondria and Mitochondrial Cascades in Alzheimer’s Disease. J. Alzheimer’s Dis. 2018, 62, 1403–1416. [Google Scholar] [CrossRef] [Green Version]
  19. Capiralla, H.; Vingtdeux, V.; Zhao, H.; Sankowski, R.; Al-Abed, Y.; Davies, P.; Marambaud, P. Resveratrol mitigates lipopolysaccharide- and Aβ-mediated microglial inflammation by inhibiting the TLR4/NF-κB/STAT signaling cascade. J. Neurochem. 2012, 120, 461–472. [Google Scholar] [CrossRef] [Green Version]
  20. Qi, Y.; Shang, L.; Liao, Z.; Su, H.; Jing, H.; Wu, B.; Bi, K.; Jia, Y. Intracerebroventricular injection of resveratrol ameliorated Aβ-induced learning and cognitive decline in mice. Metab. Brain Dis. 2019, 34, 257–266. [Google Scholar] [CrossRef]
  21. Huang, J.; Huang, N.; Xu, S.; Luo, Y.; Li, Y.; Jin, H.; Yu, C.; Shi, J.; Jin, F. Signaling mechanisms underlying inhibition of neuroinflammation by resveratrol in neurodegenerative diseases. J. Nutr. Biochem. 2021, 88, 108552. [Google Scholar] [CrossRef] [PubMed]
  22. Matsuda, Y.; Haniu, H.; Tsukahara, T.; Uemura, T.; Inoue, T.; Sako, K.-I.; Kojima, J.; Mori, T.; Sato, K. Oral administration of porcine liver decomposition product for 4 weeks enhances visual memory and delayed recall in healthy adults over 40 years of age: A randomized, double-blind, placebo-controlled study. Exp. Gerontol. 2020, 141, 111064. [Google Scholar] [CrossRef] [PubMed]
  23. Takamori, S.; Holt, M.; Stenius, K.; Lemke, E.A.; Grønborg, M.; Riedel, D.; Urlaub, H.; Schenck, S.; Brügger, B.; Ringler, P.; et al. Molecular Anatomy of a Trafficking Organelle. Cell 2006, 127, 831–846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Tsukahara, T.; Haniu, H.; Uemura, T.; Matsuda, Y. Therapeutic Potential of Porcine Liver Decomposition Product: New Insights and Perspectives for Microglia-Mediated Neuroinflammation in Neurodegenerative Diseases. Biomedicines 2020, 8, 446. [Google Scholar] [CrossRef]
  25. Tsukahara, T.; Hara, H.; Haniu, H.; Matsuda, Y. The Combined Effects of Lysophospholipids against Lipopolysaccharide-induced Inflammation and Oxidative Stress in Microglial Cells. J. Oleo Sci. 2021, 70, 947–954. [Google Scholar] [CrossRef] [PubMed]
  26. Grzelczyk, A.; Gendaszewska-Darmach, E. Novel bioactive glycerol-based lysophospholipids: New data—New insight into their function. Biochimie 2013, 95, 667–679. [Google Scholar] [CrossRef]
  27. Hao, Y.; Guo, M.; Feng, Y.; Dong, Q.; Cui, M. Lysophospholipids and Their G-Coupled Protein Signaling in Alzheimer’s Disease: From Physiological Performance to Pathological Impairment. Front. Mol. Neurosci. 2020, 13, 58. [Google Scholar] [CrossRef] [Green Version]
  28. Rosell-Valle, C.; Pedraza, C.; Manuel, I.; Moreno-Rodríguez, M.; Rodríguez-Puertas, R.; Castilla-Ortega, E.; Caramés, J.M.; Conde, A.I.G.; Zambrana-Infantes, E.; Ortega-Pinazo, J.; et al. Chronic central modulation of LPA/LPA receptors-signaling pathway in the mouse brain regulates cognition, emotion, and hippocampal neurogenesis. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2021, 108, 110156. [Google Scholar] [CrossRef]
  29. Birgbauer, E. Lysophosphatidic Acid Signalling in Nervous System Development and Function. NeuroMolecular Med. 2021, 23, 68–85. [Google Scholar] [CrossRef]
  30. Bridi, J.; Hirth, F. Mechanisms of α-Synuclein Induced Synaptopathy in Parkinson’s Disease. Front. Neurosci. 2018, 12, 80. [Google Scholar] [CrossRef]
  31. Burré, J.; Sharma, M.; Südhof, T.C. α-Synuclein assembles into higher-order multimers upon membrane binding to promote SNARE complex formation. Proc. Natl. Acad. Sci. USA 2014, 111, E4274–E4283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Taguchi, K.; Watanabe, Y.; Tsujimura, A.; Tanaka, M. Expression of α-synuclein is regulated in a neuronal cell type-dependent manner. Anat. Sci. Int. 2019, 94, 11–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Burré, J. The Synaptic Function of α-Synuclein. J. Park. Dis. 2015, 5, 699–713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Wille, H.; Dorosh, L.; Amidian, S.; Schmitt-Ulms, G.; Stepanova, M. Combining molecular dynamics simulations and experimental analyses in protein misfolding. Adv. Protein Chem. Struct. Biol. 2019, 118, 33–110. [Google Scholar] [CrossRef]
  35. Vassallo, N.; Galvagnion, C.; Chi, E.Y. Editorial: Amyloid-Membrane Interactions in Protein Misfolding Disorders: From Basic Mechanisms to Therapy. Front. Cell Dev. Biol. 2022, 10, 870791. [Google Scholar] [CrossRef]
  36. Giacomelli, C.; Daniele, S.; Martini, C. Potential biomarkers and novel pharmacological targets in protein aggregation-related neurodegenerative diseases. Biochem. Pharmacol. 2017, 131, 1–15. [Google Scholar] [CrossRef]
  37. Kiechle, M.; Grozdanov, V.; Danzer, K.M. The Role of Lipids in the Initiation of α-Synuclein Misfolding. Front. Cell Dev. Biol. 2020, 8, 562241. [Google Scholar] [CrossRef]
  38. Goedert, M.; Jakes, R.; Spillantini, M.G. The Synucleinopathies: Twenty Years On. J. Parkinsons Dis. 2017, 7, S51–S69. [Google Scholar] [CrossRef] [Green Version]
  39. Spillantini, M.G.; Schmidt, M.L.; Lee, V.M.-Y.; Trojanowski, J.Q.; Jakes, R.; Goedert, M. α-Synuclein in Lewy bodies. Nature 1997, 388, 839–840. [Google Scholar] [CrossRef]
  40. Jasutkar, H.G.; Oh, S.E.; Mouradian, M.M. Therapeutics in the Pipeline Targeting α-Synuclein for Parkinson’s Disease. Pharmacol. Rev. 2022, 74, 207–237. [Google Scholar] [CrossRef]
  41. Danzer, K.M.; Kranich, L.R.; Ruf, W.P.; Cagsal-Getkin, O.; Winslow, A.R.; Zhu, L.; Vanderburg, C.R.; McLean, P.J. Exosomal cell-to-cell transmission of alpha synuclein oligomers. Mol. Neurodegener. 2012, 7, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Iwai, A.; Masliah, E.; Yoshimoto, M.; Ge, N.; Flanagan, L.; de Silva, H.R.; Kittel, A.; Saitoh, T. The precursor protein of non-Aβ component of Alzheimer’s disease amyloid is a presynaptic protein of the central nervous system. Neuron 1995, 14, 467–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Spillantini, M.G.; Crowther, R.A.; Jakes, R.; Hasegawa, M.; Goedert, M. α-Synuclein in filamentous inclusions of Lewy bodies from Parkinson’s disease and dementia with Lewy bodies. Proc. Natl. Acad. Sci. USA 1998, 95, 6469–6473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Galvin, J.E.; Lee, V.M.; Trojanowski, J.Q. Synucleinopathies: Clinical and pathological implications. Arch. Neurol. 2001, 58, 186–190. [Google Scholar] [CrossRef]
  45. Guerrero-Ferreira, R.; I Taylor, N.M.; Mona, D.; Ringler, P.; E Lauer, M.; Riek, R.; Britschgi, M.; Stahlberg, H. Cryo-EM structure of alpha-synuclein fibrils. eLife 2018, 7, e36402. [Google Scholar] [CrossRef]
  46. GBD 2016 Dementia Collaborators. Global, regional, and national burden of Alzheimer’s disease and other dementias, 1990–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019, 18, 88–106. [Google Scholar] [CrossRef] [Green Version]
  47. Kim, W.S.; Kågedal, K.; Halliday, G.M. Alpha-synuclein biology in Lewy body diseases. Alzheimer’s Res. Ther. 2014, 6, 73. [Google Scholar] [CrossRef] [Green Version]
  48. Geser, F.; Wenning, G.; Poewe, W.; McKeith, I. How to diagnose dementia with Lewy bodies: State of the art. Mov. Disord. 2005, 20 (Suppl. S12), S11–S20. [Google Scholar] [CrossRef]
  49. Stefanis, L. alpha-Synuclein in Parkinson’s Disease. Cold Spring Harb. Perspect. Med. 2011, 2, a009399. [Google Scholar] [CrossRef] [Green Version]
  50. Emamzadeh, F.N. Alpha-synuclein structure, functions, and interactions. J. Res. Med. Sci. 2016, 21, 29. [Google Scholar] [CrossRef]
  51. Wang, C.; Zhao, C.; Li, D.; Tian, Z.; Lai, Y.; Diao, J.; Liu, C. Versatile Structures of α-Synuclein. Front. Mol. Neurosci. 2016, 9, 48. [Google Scholar] [CrossRef] [PubMed]
  52. Sarchione, A.; Marchand, A.; Taymans, J.-M.; Chartier-Harlin, M.-C. Alpha-Synuclein and Lipids: The Elephant in the Room? Cells 2021, 10, 2452. [Google Scholar] [CrossRef] [PubMed]
  53. Fecchio, C.; Palazzi, L.; de Laureto, P.P. α-Synuclein and Polyunsaturated Fatty Acids: Molecular Basis of the Interaction and Implication in Neurodegeneration. Molecules 2018, 23, 1531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Segrest, J.P.; De Loof, H.; Dohlman, J.G.; Brouillette, C.G.; Ananthara-Maiah, G.M. Amphipathic helix motif: Classes and properties. Proteins 1990, 8, 103–117. [Google Scholar] [CrossRef] [PubMed]
  55. Segrest, J.; Jones, M.; De Loof, H.; Brouillette, C.; Venkatachalapathi, Y.; Anantharamaiah, G. The amphipathic helix in the exchangeable apolipoproteins: A review of secondary structure and function. J. Lipid Res. 1992, 33, 141–166. [Google Scholar] [CrossRef]
  56. Pallbo, J.; Olsson, U.; Sparr, E. Strong inhibition of peptide amyloid formation by a fatty acid. Biophys. J. 2021, 120, 4536–4546. [Google Scholar] [CrossRef]
  57. Pallbo, J.; Imai, M.; Gentile, L.; Takata, S.-I.; Olsson, U.; Sparr, E. NACore Amyloid Formation in the Presence of Phospholipids. Front. Physiol. 2020, 11, 592117. [Google Scholar] [CrossRef]
  58. Ueda, K.; Fukushima, H.; Masliah, E.; Xia, Y.; Iwai, A.; Yoshimoto, M.; Otero, D.A.; Kondo, J.; Ihara, Y.; Saitoh, T. Molecular cloning of cDNA encoding an unrecognized component of amyloid in Alzheimer disease. Proc. Natl. Acad. Sci. USA 1993, 90, 11282–11286. [Google Scholar] [CrossRef] [Green Version]
  59. Burré, J.; Sharma, M.; Südhof, T.C. Cell Biology and Pathophysiology of α-Synuclein. Cold Spring Harb. Perspect. Med. 2017, 8, a024091. [Google Scholar] [CrossRef]
  60. Galvagnion, C.; Buell, A.K.; Meisl, G.; Michaels, T.; Vendruscolo, M.; Knowles, T.; Dobson, C.M. Lipid vesicles trigger α-synuclein aggregation by stimulating primary nucleation. Nat. Chem. Biol. 2015, 11, 229–234. [Google Scholar] [CrossRef]
  61. Campioni, S.; Carret, G.; Jordens, S.; Nicoud, L.; Mezzenga, R.; Riek, R. The Presence of an Air–Water Interface Affects Formation and Elongation of α-Synuclein Fibrils. J. Am. Chem. Soc. 2014, 136, 2866–2875. [Google Scholar] [CrossRef] [PubMed]
  62. Bell, R.; Vendruscolo, M. Modulation of the Interactions Between α-Synuclein and Lipid Membranes by Post-translational Modifications. Front. Neurol. 2021, 12, 661117. [Google Scholar] [CrossRef] [PubMed]
  63. Peng, C.; Gathagan, R.J.; Covell, D.J.; Medellin, C.; Stieber, A.; Robinson, J.L.; Zhang, B.; Pitkin, R.M.; Olufemi, M.F.; Luk, K.C.; et al. Cellular milieu imparts distinct pathological α-synuclein strains in α-synucleinopathies. Nature 2018, 557, 558–563. [Google Scholar] [CrossRef] [PubMed]
  64. Stephens, A.D.; Zacharopoulou, M.; Schierle, G.S.K. The Cellular Environment Affects Monomeric α-Synuclein Structure. Trends Biochem. Sci. 2019, 44, 453–466. [Google Scholar] [CrossRef]
  65. Jo, E.; McLaurin, J.; Yip, C.M.; St George-Hyslop, P.; Fraser, P.E. α-Synuclein Membrane Interactions and Lipid Specificity. J. Biol. Chem. 2000, 275, 34328–34334. [Google Scholar] [CrossRef] [Green Version]
  66. Afitska, K.; Priss, A.; Yushchenko, D.A.; Shvadchak, V.V. Structural Optimization of Inhibitors of α-Synuclein Fibril Growth: Affinity to the Fibril End as a Crucial Factor. J. Mol. Biol. 2020, 432, 967–977. [Google Scholar] [CrossRef]
  67. Zhu, M.; Fink, A.L. Lipid Binding Inhibits α-Synuclein Fibril Formation. J. Biol. Chem. 2003, 278, 16873–16877. [Google Scholar] [CrossRef] [Green Version]
  68. Cole, N.B.; Murphy, D.D.; Grider, T.; Rueter, S.; Brasaemle, D.; Nussbaum, R.L. Lipid Droplet Binding and Oligomerization Properties of the Parkinson’s Disease Protein α-Synuclein. J. Biol. Chem. 2002, 277, 6344–6352. [Google Scholar] [CrossRef] [Green Version]
  69. Davidson, W.S.; Jonas, A.; Clayton, D.F.; George, J.M. Stabilization of α-Synuclein Secondary Structure upon Binding to Synthetic Membranes. J. Biol. Chem. 1998, 273, 9443–9449. [Google Scholar] [CrossRef] [Green Version]
  70. Narayanan, V.; Scarlata, S. Membrane Binding and Self-Association of α-Synucleins. Biochemistry 2001, 40, 9927–9934. [Google Scholar] [CrossRef]
  71. Antony, T.; Hoyer, W.; Cherny, D.; Heim, G.; Jovin, T.M.; Subramaniam, V. Cellular Polyamines Promote the Aggregation of α-Synuclein. J. Biol. Chem. 2003, 278, 3235–3240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Fernandez, C.O.; Hoyer, W.; Zweckstetter, M.; A Jares-Erijman, E.; Subramaniam, V.; Griesinger, C.; Jovin, T.M. NMR of α-synuclein–polyamine complexes elucidates the mechanism and kinetics of induced aggregation. EMBO J. 2004, 23, 2039–2046. [Google Scholar] [CrossRef] [PubMed]
  73. Cohlberg, J.A.; Li, J.; Uversky, V.N.; Fink, A.L. Heparin and other glycosaminoglycans stimulate the formation of amyloid fibrils from alpha-synuclein in vitro. Biochemistry 2002, 41, 1502–1511. [Google Scholar] [CrossRef]
  74. Rasia, R.M.; Bertoncini, C.W.; Marsh, D.; Hoyer, W.; Cherny, D.; Zweckstetter, M.; Griesinger, C.; Jovin, T.M.; Fernández, C.O. Structural characterization of copper(II) binding to α-synuclein: Insights into the bioinorganic chemistry of Parkinson’s disease. Proc. Natl. Acad. Sci. USA 2005, 102, 4294–4299. [Google Scholar] [CrossRef] [Green Version]
  75. Vidović, M.; Rikalovic, M.G. Alpha-Synuclein Aggregation Pathway in Parkinson’s Disease: Current Status and Novel Therapeutic Approaches. Cells 2022, 11, 1732. [Google Scholar] [CrossRef] [PubMed]
  76. Santner, A.; Uversky, V.N. Metalloproteomics and metal toxicology of α-synuclein. Metallomics 2010, 2, 378–392. [Google Scholar] [CrossRef] [PubMed]
  77. Cremades, N.; Cohen, S.I.; Deas, E.; Abramov, A.Y.; Chen, A.Y.; Orte, A.; Sandal, M.; Clarke, R.W.; Dunne, P.; Aprile, F.A.; et al. Direct Observation of the Interconversion of Normal and Toxic Forms of α-Synuclein. Cell 2012, 149, 1048–1059. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Abramov, A.Y.; Potapova, E.V.; Dremin, V.V.; Dunaev, A.V. Interaction of Oxidative Stress and Misfolded Proteins in the Mechanism of Neurodegeneration. Life 2020, 10, 101. [Google Scholar] [CrossRef]
  79. Haslund-Vinding, J.; McBean, G.; Jaquet, V.; Vilhardt, F. NADPH oxidases in oxidant production by microglia: Activating receptors, pharmacology and association with disease. Br. J. Pharmacol. 2017, 174, 1733–1749. [Google Scholar] [CrossRef] [Green Version]
  80. Hou, L.; Bao, X.; Zang, C.; Yang, H.; Sun, F.; Che, Y.; Wu, X.; Li, S.; Zhang, D.; Wang, Q. Integrin CD11b mediates α-synuclein-induced activation of NADPH oxidase through a Rho-dependent pathway. Redox Biol. 2018, 14, 600–608. [Google Scholar] [CrossRef]
  81. Tiwari, P.C.; Pal, R. The potential role of neuroinflammation and transcription factors in Parkinson disease. Dialogues Clin. Neurosci. 2017, 19, 71–80. [Google Scholar] [CrossRef] [PubMed]
  82. Chunming, J.; Wang, W.; Ling, J.; Jiang, C. α-Mangostin Inhibits α-Synuclein-Induced Microglial Neuroinflammation and Neurotoxicity. Cell. Mol. Neurobiol. 2016, 36, 811–820. [Google Scholar] [CrossRef] [Green Version]
  83. Ghosh, A.A.; Verma, D.K.; Cabrera, G.; Ofori, K.; Hernandez-Quijada, K.; Kim, J.-K.; Chung, J.H.; Moore, M.; Moon, S.H.; Seo, J.B.; et al. A Novel NOX Inhibitor Treatment Attenuates Parkinson’s Disease-Related Pathology in Mouse Models. Int. J. Mol. Sci. 2022, 23, 4262. [Google Scholar] [CrossRef] [PubMed]
  84. Leak, R.K.; Frosch, M.P.; Beach, T.G.; Halliday, G.M. Alpha-synuclein: Prion or prion-like? Acta Neuropathol. 2019, 138, 509–514. [Google Scholar] [CrossRef] [PubMed]
  85. Oueslati, A. Implication of Alpha-Synuclein Phosphorylation at S129 in Synucleinopathies: What Have We Learned in the Last Decade? J. Park. Dis. 2016, 6, 39–51. [Google Scholar] [CrossRef] [Green Version]
  86. Okochi, M.; Walter, J.; Koyama, A.; Nakajo, S.; Baba, M.; Iwatsubo, T.; Meijer, L.; Kahle, P.J.; Haass, C. Constitutive Phosphorylation of the Parkinson’s Disease Associated α-Synuclein. J. Biol. Chem. 2000, 275, 390–397. [Google Scholar] [CrossRef] [Green Version]
  87. Fujiwara, H.; Hasegawa, M.; Dohmae, N.; Kawashima, A.; Masliah, E.; Goldberg, M.S.; Shen, J.; Takio, K.; Iwatsubo, T. α-Synuclein is phosphorylated in synucleinopathy lesions. Nature 2002, 4, 160–164. [Google Scholar] [CrossRef]
  88. Anderson, J.P.; Walker, D.E.; Goldstein, J.M.; de Laat, R.; Banducci, K.; Caccavello, R.J.; Barbour, R.; Huang, J.; Kling, K.; Lee, M.; et al. Phosphorylation of Ser-129 Is the Dominant Pathological Modification of α-Synuclein in Familial and Sporadic Lewy Body Disease. J. Biol. Chem. 2006, 281, 29739–29752. [Google Scholar] [CrossRef] [Green Version]
  89. Samuel, F.; Flavin, W.P.; Iqbal, S.; Pacelli, C.; Renganathan, S.D.S.; Trudeau, L.-E.; Campbell, E.M.; Fraser, P.E.; Tandon, A. Effects of Serine 129 Phosphorylation on α-Synuclein Aggregation, Membrane Association, and Internalization. J. Biol. Chem. 2016, 291, 4374–4385. [Google Scholar] [CrossRef] [Green Version]
  90. Arawaka, S.; Sato, H.; Sasaki, A.; Koyama, S.; Kato, T. Mechanisms underlying extensive Ser129-phosphorylation in α-synuclein aggregates. Acta Neuropathol. Commun. 2017, 5, 48. [Google Scholar] [CrossRef]
  91. Rodriguez, L.; Marano, M.M.; Tandon, A. Import and Export of Misfolded α-Synuclein. Front. Neurosci. 2018, 12, 344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Kawahata, I.; Bousset, L.; Melki, R.; Fukunaga, K. Fatty Acid-Binding Protein 3 is Critical for α-Synuclein Uptake and MPP+-Induced Mitochondrial Dysfunction in Cultured Dopaminergic Neurons. Int. J. Mol. Sci. 2019, 20, 5358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Paleologou, K.E.; Oueslati, A.; Shakked, G.; Rospigliosi, C.C.; Kim, H.-Y.; Lamberto, G.R.; Fernandez, C.O.; Schmid, A.; Chegini, F.; Gai, W.P.; et al. Phosphorylation at S87 Is Enhanced in Synucleinopathies, Inhibits alpha-Synuclein Oligomerization, and Influences Synuclein-Membrane Interactions. J. Neurosci. 2010, 30, 3184–3198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Le Guerroué, F.; Youle, R.J. Ubiquitin signaling in neurodegenerative diseases: An autophagy and proteasome perspective. Cell Death Differ. 2021, 28, 439–454. [Google Scholar] [CrossRef] [PubMed]
  95. Gómez-Tortosa, E.; Newell, K.; Irizarry, M.C.; Sanders, J.L.; Hyman, B.T. α-Synuclein immunoreactivity in dementia with Lewy bodies: Morphological staging and comparison with ubiquitin immunostaining. Acta Neuropathol. 2000, 99, 352–357. [Google Scholar] [CrossRef]
  96. Nonaka, T.; Iwatsubo, T.; Hasegawa, M. Ubiquitination of α-Synuclein. Biochemistry 2005, 44, 361–368. [Google Scholar] [CrossRef]
  97. Stefanis, L.; Emmanouilidou, E.; Pantazopoulou, M.; Kirik, D.; Vekrellis, K.; Tofaris, G. How is alpha-synuclein cleared from the cell? J. Neurochem. 2019, 150, 577–590. [Google Scholar] [CrossRef] [Green Version]
  98. Hasegawa, M.; Fujiwara, H.; Nonaka, T.; Wakabayashi, K.; Takahashi, H.; Lee, V.M.-Y.; Trojanowski, J.Q.; Mann, D.; Iwatsubo, T. Phosphorylated α-Synuclein Is Ubiquitinated in α-Synucleinopathy Lesions. J. Biol. Chem. 2002, 277, 49071–49076. [Google Scholar] [CrossRef] [Green Version]
  99. Agerschou, E.D.; Flagmeier, P.; Saridaki, T.; Galvagnion, C.; Komnig, D.; Heid, L.; Prasad, V.; Shaykhalishahi, H.; Willbold, D.; Dobson, C.M.; et al. An engineered monomer binding-protein for α-synuclein efficiently inhibits the proliferation of amyloid fibrils. eLife 2019, 8, e46112. [Google Scholar] [CrossRef]
  100. Rosado-Ramos, R.; Godinho-Pereira, J.; Marques, D.; Figueira, I.; Outeiro, T.F.; Menezes, R.; dos Santos, C.N. Small Molecule Fisetin Modulates Alpha–Synuclein Aggregation. Molecules 2021, 26, 3353. [Google Scholar] [CrossRef]
  101. Zhu, M.; Rajamani, S.; Kaylor, J.; Han, S.; Zhou, F.; Fink, A.L. The Flavonoid Baicalein Inhibits Fibrillation of α-Synuclein and Disaggregates Existing Fibrils. J. Biol. Chem. 2004, 279, 26846–26857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Xu, Y.; Zhang, Y.; Quan, Z.; Wong, W.; Guo, J.; Zhang, R.; Yang, Q.; Dai, R.; McGeer, P.L.; Qing, H. Epigallocatechin Gallate (EGCG) Inhibits Alpha-Synuclein Aggregation: A Potential Agent for Parkinson’s Disease. Neurochem. Res. 2016, 41, 2788–2796. [Google Scholar] [CrossRef]
  103. Chau, E.; Kim, H.; Shin, J.; Martinez, A.; Kim, J.R. Inhibition of alpha-synuclein aggregation by AM17, a synthetic resveratrol derivative. Biochem. Biophys. Res. Commun. 2021, 574, 85–90. [Google Scholar] [CrossRef] [PubMed]
  104. Levin, J.; Sing, N.; Melbourne, S.; Morgan, A.; Mariner, C.; Spillantini, M.G.; Wegrzynowicz, M.; Dalley, J.W.; Langer, S.; Ryazanov, S.; et al. Safety, tolerability and pharmacokinetics of the oligomer modulator anle138b with exposure levels sufficient for therapeutic efficacy in a murine Parkinson model: A randomised, double-blind, placebo-controlled phase 1a trial. eBioMedicine 2022, 80, 104021. [Google Scholar] [CrossRef] [PubMed]
  105. Smit, J.W.; Basile, P.; Prato, M.K.; Detalle, L.; Mathy, F.; Schmidt, A.; Lalla, M.; Germani, M.; Domange, C.; Biere, A.; et al. Phase 1/1b Studies of UCB0599, an Oral Inhibitor of α-Synuclein Misfolding, Including a Randomized Study in Parkinson’s Disease. Mov. Disord. 2022, 37, 2045–2056. [Google Scholar] [CrossRef]
  106. Hyun, S.; Shin, D. Chemical-Mediated Targeted Protein Degradation in Neurodegenerative Diseases. Life 2021, 11, 607. [Google Scholar] [CrossRef]
  107. Shah, A.K.M.A.; Nagao, T.; Kurihara, H.; Takahashi, K. Production of a Health-Beneficial Food Emulsifier by Enzymatic Partial Hydrolysis of Phospholipids Obtained from the Head of Autumn Chum Salmon. J. Oleo Sci. 2017, 66, 147–155. [Google Scholar] [CrossRef] [Green Version]
  108. D’Arrigo, P.; Servi, S. Synthesis of Lysophospholipids. Molecules 2010, 15, 1354–1377. [Google Scholar] [CrossRef] [Green Version]
  109. Zhang, M.; Bai, H.; Zhao, Y.; Wang, R.; Li, G.; Zhang, G.; Zhang, Y. Effects of Dietary Lysophospholipid Inclusion on the Growth Performance, Nutrient Digestibility, Nitrogen Utilization, and Blood Metabolites of Finishing Beef Cattle. Antioxidants 2022, 11, 1486. [Google Scholar] [CrossRef]
  110. Zhao, P.; Li, H.; Hossain, M.; Kim, I. Effect of emulsifier (lysophospholipids) on growth performance, nutrient digestibility and blood profile in weanling pigs. Anim. Feed Sci. Technol. 2015, 207, 190–195. [Google Scholar] [CrossRef]
  111. Hui, D.Y. Intestinal phospholipid and lysophospholipid metabolism in cardiometabolic disease. Curr. Opin. Infect. Dis. 2016, 27, 507–512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Takatera, A.; Takeuchi, A.; Saiki, K.; Morisawa, T.; Yokoyama, N.; Matsuo, M. Quantification of lysophosphatidylcholines and phosphatidylcholines using liquid chromatography–tandem mass spectrometry in neonatal serum. J. Chromatogr. B 2006, 838, 31–36. [Google Scholar] [CrossRef] [PubMed]
  113. Guillermo, R.B.; Yang, P.; Vickers, M.H.; McJarrow, P.; Guan, J. Supplementation with complex milk lipids during brain development promotes neuroplasticity without altering myelination or vascular density. Food Nutr. Res. 2015, 59, 25765. [Google Scholar] [CrossRef] [Green Version]
  114. Senior, S.L.; Ninkina, N.; Deacon, R.; Bannerman, D.; Buchman, V.L.; Cragg, S.J.; Wade-Martins, R. Increased striatal dopamine release and hyperdopaminergic-like behaviour in mice lacking both alpha-synuclein and gamma-synuclein. Eur. J. Neurosci. 2008, 27, 947–957. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Petersen, R.C.; Smith, G.E.; Waring, S.C.; Ivnik, R.J.; Tangalos, E.G.; Kokmen, E. Mild Cognitive Impairment: Clinical characterization and outcome. Arch. Neurol. 1999, 56, 303–308. [Google Scholar] [CrossRef] [PubMed]
  116. Petersen, R.C. Early Diagnosis of Alzheimers Disease: Is MCI Too Late? Curr. Alzheimer Res. 2009, 6, 324–330. [Google Scholar] [CrossRef]
  117. Foltynie, T.; Brayne, C.R.; Robbins, T.W.; Barker, R.A. The cognitive ability of an incident cohort of Parkinson’s patients in the UK. The CamPaIGN study. Brain 2004, 127, 550–560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Muslimović, D.; Post, B.; Speelman, J.D.; Schmand, B. Cognitive profile of patients with newly diagnosed Parkinson disease. Neurology 2005, 65, 1239–1245. [Google Scholar] [CrossRef]
  119. Hassin-Baer, S.; Cohen, O.S.; Israeli-Korn, S.; Yahalom, G.; Benizri, S.; Sand, D.; Issachar, G.; Geva, A.B.; Shani-Hershkovich, R.; Peremen, Z. Identification of an early-stage Parkinson’s disease neuromarker using event-related potentials, brain network analytics and machine-learning. PLoS ONE 2022, 17, e0261947. [Google Scholar] [CrossRef]
  120. Facal, D.; Spuch, C.; Valladares-Rodriguez, S. New Trends in Cognitive Aging and Mild Cognitive Impairment. Geriatrics 2022, 7, 80. [Google Scholar] [CrossRef]
  121. Campbell, N.L.; Unverzagt, F.; LaMantia, M.A.; Khan, B.A.; Boustani, M.A. Risk Factors for the Progression of Mild Cognitive Impairment to Dementia. Clin. Geriatr. Med. 2013, 29, 873–893. [Google Scholar] [CrossRef] [Green Version]
  122. Godos, J.; Currenti, W.; Angelino, D.; Mena, P.; Castellano, S.; Caraci, F.; Galvano, F.; Del Rio, D.; Ferri, R.; Grosso, G. Diet and Mental Health: Review of the Recent Updates on Molecular Mechanisms. Antioxidants 2020, 9, 346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Caruso, G.; Godos, J.; Privitera, A.; Lanza, G.; Castellano, S.; Chillemi, A.; Bruni, O.; Ferri, R.; Caraci, F.; Grosso, G. Phenolic Acids and Prevention of Cognitive Decline: Polyphenols with a Neuroprotective Role in Cognitive Disorders and Alzheimer’s Disease. Nutrients 2022, 14, 819. [Google Scholar] [CrossRef] [PubMed]
  124. Guiné, R.P.F.; Florença, S.G.; Barroca, M.J.; Anjos, O. The Link between the Consumer and the Innovations in Food Product Development. Foods 2020, 9, 1317. [Google Scholar] [CrossRef] [PubMed]
  125. Gómez-Pinilla, F. Brain foods: The effects of nutrients on brain function. Nat. Rev. Neurosci. 2008, 9, 568–578. [Google Scholar] [CrossRef] [Green Version]
  126. Chianese, R.; Coccurello, R.; Viggiano, A.; Scafuro, M.; Fiore, M.; Coppola, G.; Operto, F.F.; Fasano, S.; Laye, S.; Pierantoni, R.; et al. Impact of Dietary Fats on Brain Functions. Curr. Neuropharmacol. 2018, 16, 1059–1085. [Google Scholar] [CrossRef]
  127. Dadhania, V.P.; Trivedi, P.P.; Vikram, A.; Tripathi, D.N. Nutraceuticals against Neurodegeneration: A Mechanistic Insight. Curr. Neuropharmacol. 2016, 14, 627–640. [Google Scholar] [CrossRef]
  128. Yuyama, K.; Igarashi, Y. Linking glycosphingolipids to Alzheimer’s amyloid-ß: Extracellular vesicles and functional plant materials. Glycoconj. J. 2022, 39, 613–618. [Google Scholar] [CrossRef]
  129. Yuyama, K.; Takahashi, K.; Usuki, S.; Mikami, D.; Sun, H.; Hanamatsu, H.; Furukawa, J.; Mukai, K.; Igarashi, Y. Plant sphingolipids promote extracellular vesicle release and alleviate amyloid-β pathologies in a mouse model of Alzheimer’s disease. Sci. Rep. 2019, 9, 16827. [Google Scholar] [CrossRef] [Green Version]
  130. Ozawa, H.; Miyazawa, T.; Miyazawa, T. Effects of Dietary Food Components on Cognitive Functions in Older Adults. Nutrients 2021, 13, 2804. [Google Scholar] [CrossRef]
  131. Kinney, J.W.; BeMiller, S.M.; Murtishaw, A.S.; Leisgang, A.M.; Salazar, A.M.; Lamb, B.T. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimer’s Dement. 2018, 4, 575–590. [Google Scholar] [CrossRef] [PubMed]
  132. Garcia, P.; Jürgens-Wemheuer, W.; Huarte, O.U.; Michelucci, A.; Masuch, A.; Brioschi, S.; Weihofen, A.; Koncina, E.; Coowar, D.; Heurtaux, T.; et al. Neurodegeneration and neuroinflammation are linked, but independent of alpha-synuclein inclusions, in a seeding/spreading mouse model of Parkinson’s disease. Glia 2022, 70, 935–960. [Google Scholar] [CrossRef] [PubMed]
  133. Hammond, T.R.; Marsh, S.E.; Stevens, B. Immune Signaling in Neurodegeneration. Immunity 2019, 50, 955–974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Crotti, A.; Ransohoff, R.M. Microglial Physiology and Pathophysiology: Insights from Genome-wide Transcriptional Profiling. Immunity 2016, 44, 505–515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Guzman-Martinez, L.; Maccioni, R.B.; Andrade, V.; Navarrete, L.P.; Pastor, M.G.; Ramos-Escobar, N. Neuroinflammation as a Common Feature of Neurodegenerative Disorders. Front. Pharmacol. 2019, 10, 1008. [Google Scholar] [CrossRef] [Green Version]
  136. Janvin, C.C.; Larsen, J.P.; Aarsland, D.; Hugdahl, K. Subtypes of mild cognitive impairment in parkinson’s disease: Progression to dementia. Mov. Disord. 2006, 21, 1343–1349. [Google Scholar] [CrossRef] [PubMed]
  137. Kamioka, H.; Tsutani, K.; Origasa, H.; Yoshizaki, T.; Kitayuguchi, J.; Shimada, M.; Wada, Y.; Takano-Ohmuro, H. Quality of Systematic Reviews of the Foods with Function Claims in Japan: Comparative Before- and After-Evaluation of Verification Reports by the Consumer Affairs Agency. Nutrients 2019, 11, 1583. [Google Scholar] [CrossRef] [Green Version]
  138. Alqahtani, M.S.; Kazi, M.; Alsenaidy, M.A.; Ahmad, M.Z. Advances in Oral Drug Delivery. Front. Pharmacol. 2021, 12, 618411. [Google Scholar] [CrossRef]
  139. Tsukahara, T.; Haniu, H.; Uemura, T.; Matsuda, Y. Porcine liver decomposition product-derived lysophospholipids promote microglial activation in vitro. Sci. Rep. 2020, 10, 3748. [Google Scholar] [CrossRef] [Green Version]
  140. Gotoh, M.; Sano-Maeda, K.; Murofushi, H.; Murakami-Murofushi, K. Protection of Neuroblastoma Neuro2A Cells from Hypoxia-Induced Apoptosis by Cyclic Phosphatidic Acid (cPA). PLoS ONE 2012, 7, e51093. [Google Scholar] [CrossRef] [PubMed]
  141. Sheikh, A.M.; Michikawa, M.; Kim, S.; Nagai, A. Lysophosphatidylcholine increases the neurotoxicity of Alzheimer’s amyloid β1-42 peptide: Role of oligomer formation. Neuroscience 2015, 292, 159–169. [Google Scholar] [CrossRef] [PubMed]
  142. Mulder, C.; Wahlund, L.-O.; Teerlink, T.; Blomberg, M.; Veerhuis, R.; Van Kamp, G.J.; Scheltens, P.; Scheffer, P.G. Decreased lysophosphatidylcholine/phosphatidylcholine ratio in cerebrospinal fluid in Alzheimer’s disease. J. Neural Transm. 2003, 110, 949–955. [Google Scholar] [CrossRef] [PubMed]
  143. Sugasini, D.; Thomas, R.; Yalagala, P.C.R.; Tai, L.M.; Subbaiah, P.V. Dietary docosahexaenoic acid (DHA) as lysophosphatidylcholine, but not as free acid, enriches brain DHA and improves memory in adult mice. Sci. Rep. 2017, 7, 11263. [Google Scholar] [CrossRef] [Green Version]
  144. Schverer, M.; O’Mahony, S.M.; O’Riordan, K.J.; Donoso, F.; Roy, B.L.; Stanton, C.; Dinan, T.G.; Schellekens, H.; Cryan, J.F. Dietary phospholipids: Role in cognitive processes across the lifespan. Neurosci. Biobehav. Rev. 2020, 111, 183–193. [Google Scholar] [CrossRef] [PubMed]
  145. Donoso, F.; Schverer, M.; Rea, K.; Pusceddu, M.M.; Roy, B.L.; Dinan, T.G.; Cryan, J.F.; Schellekens, H. Neurobiological effects of phospholipids in vitro: Relevance to stress-related disorders. Neurobiol. Stress 2020, 13, 100252. [Google Scholar] [CrossRef] [PubMed]
  146. Hachem, M.; Nacir, H. Emerging Role of Phospholipids and Lysophospholipids for Improving Brain Docosahexaenoic Acid as Potential Preventive and Therapeutic Strategies for Neurological Diseases. Int. J. Mol. Sci. 2022, 23, 3969. [Google Scholar] [CrossRef]
  147. Mutoh, T.; Rivera, R.; Chun, J. Insights into the pharmacological relevance of lysophospholipid receptors. Br. J. Pharmacol. 2012, 165, 829–844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Tan, S.T.; Ramesh, T.; Toh, X.R.; Nguyen, L.N. Emerging roles of lysophospholipids in health and disease. Prog. Lipid Res. 2020, 80, 101068. [Google Scholar] [CrossRef]
  149. Falomir-Lockhart, L.J.; Cavazzutti, G.F.; Giménez, E.; Toscani, A.M. Fatty Acid Signaling Mechanisms in Neural Cells: Fatty Acid Receptors. Front. Cell. Neurosci. 2019, 13, 162. [Google Scholar] [CrossRef] [Green Version]
  150. Mahul-Mellier, A.-L.; Burtscher, J.; Maharjan, N.; Weerens, L.; Croisier, M.; Kuttler, F.; Leleu, M.; Knott, G.W.; Lashuel, H.A. The process of Lewy body formation, rather than simply α-synuclein fibrillization, is one of the major drivers of neurodegeneration. Proc. Natl. Acad. Sci. USA 2020, 117, 4971–4982. [Google Scholar] [CrossRef]
  151. Kachappilly, N.; Srivastava, J.; Swain, B.P.; Thakur, P. Interaction of alpha-synuclein with lipids. Methods Cell Biol. 2022, 169, 43–66. [Google Scholar] [CrossRef]
  152. Subhramanyam, C.S.; Wang, C.; Hu, Q.; Dheen, S.T. Microglia-mediated neuroinflammation in neurodegenerative diseases. Semin. Cell Dev. Biol. 2019, 94, 112–120. [Google Scholar] [CrossRef] [PubMed]
  153. Lands, W.E. Metabolism of glycerolipids. 2. The enzymatic acylation of lysolecithin. J. Biol. Chem. 1960, 235, 2233–2237. [Google Scholar] [CrossRef] [PubMed]
  154. Tayebati, S.K. Phospholipid and Lipid Derivatives as Potential Neuroprotective Compounds. Molecules 2018, 23, 2257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Blondeau, N.; Lauritzen, I.; Widmann, C.; Lazdunski, M.; Heurteaux, C. A Potent Protective Role of Lysophospholipids against Global Cerebral Ischemia and Glutamate Excitotoxicity in Neuronal Cultures. J. Cereb. Blood Flow Metab. 2002, 22, 821–834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Qi, K.; Hall, M.; Deckelbaum, R.J. Long-chain polyunsaturated fatty acid accretion in brain. Curr. Opin. Clin. Nutr. Metab. Care 2002, 5, 133–138. [Google Scholar] [CrossRef]
  157. Zheng, L.; Xie, C.; Zheng, J.; Dong, Q.; Si, T.; Zhang, J.; Hou, S.-T. An imbalanced ratio between PC(16:0/16:0) and LPC(16:0) revealed by lipidomics supports the role of the Lands cycle in ischemic brain injury. J. Biol. Chem. 2021, 296, 100151. [Google Scholar] [CrossRef]
  158. Semba, R.D. Perspective: The Potential Role of Circulating Lysophosphatidylcholine in Neuroprotection against Alzheimer Disease. Adv. Nutr. 2020, 11, 760–772. [Google Scholar] [CrossRef]
  159. Mori, A.; Hatano, T.; Inoshita, T.; Shiba-Fukushima, K.; Koinuma, T.; Meng, H.; Kubo, S.-I.; Spratt, S.; Cui, C.; Yamashita, C.; et al. Parkinson’s disease-associated iPLA2-VIA/ PLA2G6 regulates neuronal functions and α-synuclein stability through membrane remodeling. Proc. Natl. Acad. Sci. USA 2019, 116, 20689–20699. [Google Scholar] [CrossRef] [Green Version]
  160. Takagi, Y.; Nishikado, S.; Omi, J.; Aoki, J. The Many Roles of Lysophospholipid Mediators and Japanese Contributions to This Field. Biol. Pharm. Bull. 2022, 45, 1008–1021. [Google Scholar] [CrossRef]
  161. Yuan, Y.; Jackson, S.; Newnham, H.; A Mitchell, C.; Salem, H. An essential role for lysophosphatidylcholine in the inhibition of platelet aggregation by secretory phospholipase A2. Blood 1995, 86, 4166–4174. [Google Scholar] [CrossRef] [PubMed]
  162. Leiter, O.; Walker, T.L. Platelets in Neurodegenerative Conditions—Friend or Foe? Front. Immunol. 2020, 11, 747. [Google Scholar] [CrossRef]
  163. Curcic, S.; Holzer, M.; Pasterk, L.; Knuplez, E.; Eichmann, T.O.; Frank, S.; Zimmermann, R.; Schicho, R.; Heinemann, A.; Marsche, G. Secretory phospholipase A2 modified HDL rapidly and potently suppresses platelet activation. Sci. Rep. 2017, 7, 8030. [Google Scholar] [CrossRef]
  164. Mapstone, M.; Cheema, A.K.; Fiandaca, M.S.; Zhong, X.; Mhyre, T.R.; MacArthur, L.H.; Hall, W.J.; Fisher, S.G.; Peterson, D.R.; Haley, J.M.; et al. Plasma phospholipids identify antecedent memory impairment in older adults. Nat. Med. 2014, 20, 415–418. [Google Scholar] [CrossRef] [Green Version]
  165. Lin, W.; Zhang, J.; Liu, Y.; Wu, R.; Yang, H.; Hu, X.; Ling, X. Studies on diagnostic biomarkers and therapeutic mechanism of Alzheimer’s disease through metabolomics and hippocampal proteomics. Eur. J. Pharm. Sci. 2017, 105, 119–126. [Google Scholar] [CrossRef]
  166. Liu, Y.; Li, N.; Zhou, L.; Li, Q.; Li, W. Plasma metabolic profiling of mild cognitive impairment and Alzheimer’s disease using liquid chromatography/mass spectrometry. Cent. Nerv. Syst. Agents Med. Chem. 2014, 14, 113–120. [Google Scholar] [CrossRef]
  167. Okudaira, M.; Inoue, A.; Shuto, A.; Nakanaga, K.; Kano, K.; Makide, K.; Saigusa, D.; Tomioka, Y.; Aoki, J. Separation and quantification of 2-acyl-1-lysophospholipids and 1-acyl-2-lysophospholipids in biological samples by LC-MS/MS. J. Lipid Res. 2014, 55, 2178–2192. [Google Scholar] [CrossRef] [Green Version]
  168. Satouchi, K.; Sakaguchi, M.; Shirakawa, M.; Hirano, K.; Tanaka, T. Lysophosphatidylcholine from white muscle of bonito Euthynnus pelamis (Linnaeus): Involvement of phospholipase A1 activity for its production. Biochim. Biophys. Acta 1994, 1214, 303–308. [Google Scholar] [PubMed]
  169. Richard, C.; Calder, P.C. Docosahexaenoic Acid. Adv. Nutr. 2016, 7, 1139–1141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Nguyen, L.N.; Ma, D.; Shui, G.; Wong, P.; Cazenave-Gassiot, A.; Zhang, X.; Wenk, M.R.; Goh, E.L.K.; Silver, D.L. Mfsd2a is a transporter for the essential omega-3 fatty acid docosahexaenoic acid. Nature 2014, 509, 503–506. [Google Scholar] [CrossRef] [PubMed]
  171. Mitchell, R.W.; Hatch, G.M. Fatty acid transport into the brain: Of fatty acid fables and lipid tails. Prostaglandins Leukot. Essent. Fat. Acids 2011, 85, 293–302. [Google Scholar] [CrossRef]
  172. Karaki, T.; Haniu, H.; Matsuda, Y.; Tsukahara, T. Lysophospholipids–potent candidates for brain food, protects neuronal cells against α-Synuclein aggregation. Biomed. Pharmacother. 2022, 156, 113891. [Google Scholar] [CrossRef] [PubMed]
  173. Khammari, A.; Arab, S.S.; Ejtehadi, M.R. The hot sites of α-synuclein in amyloid fibril formation. Sci. Rep. 2020, 10, 12175. [Google Scholar] [CrossRef] [PubMed]
  174. Ghosh, D.; Singh, P.K.; Sahay, S.; Jha, N.N.; Jacob, R.; Sen, S.; Kumar, A.; Riek, R.; Maji, S.K. Structure based aggregation studies reveal the presence of helix-rich intermediate during α-Synuclein aggregation. Sci. Rep. 2015, 5, 9228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Kern, R.; Joseleau-Petit, D.; Chattopadhyay, M.K.; Richarme, G. Chaperone-like Properties of Lysophospholipids. Biochem. Biophys. Res. Commun. 2001, 289, 1268–1274. [Google Scholar] [CrossRef]
  176. Vallés, A.S.; Barrantes, F.J. The synaptic lipidome in health and disease. Biochim. Biophys. Acta 2022, 1864, 184033. [Google Scholar] [CrossRef] [PubMed]
Figure 1. PLDP is a rich source of phospholipids. PLDP extracted lipids (PEL) was extracted from PLDP using the Bligh and Dyer method. This PEL is a rich source of LPLs, including LPC and LPE. LPC and LPE exerted significant protective effects against LPS-induced inflammation and oxidative stress in microglial cells. Various isoforms of PLA2 enzyme hydrolyze PC and PE at the sn-2 position to form LPLs, including LPC and LPE, respectively. α-Syn is bound to LPLs, which are known to be contained in PEL, strongly inhibit α-Syn aggregation.
Figure 1. PLDP is a rich source of phospholipids. PLDP extracted lipids (PEL) was extracted from PLDP using the Bligh and Dyer method. This PEL is a rich source of LPLs, including LPC and LPE. LPC and LPE exerted significant protective effects against LPS-induced inflammation and oxidative stress in microglial cells. Various isoforms of PLA2 enzyme hydrolyze PC and PE at the sn-2 position to form LPLs, including LPC and LPE, respectively. α-Syn is bound to LPLs, which are known to be contained in PEL, strongly inhibit α-Syn aggregation.
Biomedicines 10 03126 g001
Figure 2. Schematic illustration of the potential mechanism underlying lysophospholipid (LPL) and α-Synuclein (α-Syn) aggregation. (A) α-Syn is composed of three domains: the N-terminal, NAC, and C-terminal domains. The N-terminus contains a lipid-binding motif. LPLs lack one fatty acid in comparison to diacylglycerol lipids, and they are much more hydrophilic molecules. (B) In aqueous environments, LPLs undergo basic effects, such as the adsorption of the unfolded monomer α-Syn (positive surface charge) upon release from the surface of the cell membrane (negative surface charge). From experimental data, we hypothesize that when α-Syn is bound to LPLs, LPC18:1 and LPE 18:1 (which are known to be contained in PEL) strongly inhibit α-Syn aggregation.
Figure 2. Schematic illustration of the potential mechanism underlying lysophospholipid (LPL) and α-Synuclein (α-Syn) aggregation. (A) α-Syn is composed of three domains: the N-terminal, NAC, and C-terminal domains. The N-terminus contains a lipid-binding motif. LPLs lack one fatty acid in comparison to diacylglycerol lipids, and they are much more hydrophilic molecules. (B) In aqueous environments, LPLs undergo basic effects, such as the adsorption of the unfolded monomer α-Syn (positive surface charge) upon release from the surface of the cell membrane (negative surface charge). From experimental data, we hypothesize that when α-Syn is bound to LPLs, LPC18:1 and LPE 18:1 (which are known to be contained in PEL) strongly inhibit α-Syn aggregation.
Biomedicines 10 03126 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Karaki, T.; Haniu, H.; Matsuda, Y.; Tsukahara, T. Lysophospholipids: A Potential Drug Candidates for Neurodegenerative Disorders. Biomedicines 2022, 10, 3126. https://doi.org/10.3390/biomedicines10123126

AMA Style

Karaki T, Haniu H, Matsuda Y, Tsukahara T. Lysophospholipids: A Potential Drug Candidates for Neurodegenerative Disorders. Biomedicines. 2022; 10(12):3126. https://doi.org/10.3390/biomedicines10123126

Chicago/Turabian Style

Karaki, Tatsuya, Hisao Haniu, Yoshikazu Matsuda, and Tamotsu Tsukahara. 2022. "Lysophospholipids: A Potential Drug Candidates for Neurodegenerative Disorders" Biomedicines 10, no. 12: 3126. https://doi.org/10.3390/biomedicines10123126

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop