Next Article in Journal
Molecular Pathophysiology of Autosomal Recessive Polycystic Kidney Disease
Next Article in Special Issue
Neuroinflammation: A Signature or a Cause of Epilepsy?
Previous Article in Journal
The Exon Junction Complex Core Represses Cancer-Specific Mature mRNA Re-splicing: A Potential Key Role in Terminating Splicing
Previous Article in Special Issue
Neuroprotective and Immunomodulatory Action of the Endocannabinoid System under Neuroinflammation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Alpha-Synuclein as a Prominent Actor in the Inflammatory Synaptopathy of Parkinson’s Disease

by
Antonella Cardinale
1,2,†,
Valeria Calabrese
1,3,†,
Antonio de Iure
1 and
Barbara Picconi
1,4,*
1
Laboratory Experimental Neurophysiology, IRCCS San Raffaele Pisana, 00166 Rome, Italy
2
Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
3
Department of Medicine, Università degli Studi di Perugia, 06123 Perugia, Italy
4
Department of di Scienze Umane e Promozione della Qualità della Vita, Università Telematica San Raffaele, 00166 Rome, Italy
*
Author to whom correspondence should be addressed.
A.C. and V.C. contributed equally to this work.
Int. J. Mol. Sci. 2021, 22(12), 6517; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22126517
Submission received: 14 May 2021 / Revised: 4 June 2021 / Accepted: 15 June 2021 / Published: 17 June 2021
(This article belongs to the Special Issue Molecular and Cellular Mechanisms in Inflammatory Synaptopathy)

Abstract

:
Parkinson’s disease (PD) is considered the most common disorder of synucleinopathy, which is characterised by intracellular inclusions of aggregated and misfolded α-synuclein (α-syn) protein in various brain regions, and the loss of dopaminergic neurons. During the early prodromal phase of PD, synaptic alterations happen before cell death, which is linked to the synaptic accumulation of toxic α-syn specifically in the presynaptic terminals, affecting neurotransmitter release. The oligomers and protofibrils of α-syn are the most toxic species, and their overexpression impairs the distribution and activation of synaptic proteins, such as the SNARE complex, preventing neurotransmitter exocytosis and neuronal synaptic communication. In the last few years, the role of the immune system in PD has been increasingly considered. Microglial and astrocyte activation, the gene expression of proinflammatory factors, and the infiltration of immune cells from the periphery to the central nervous system (CNS) represent the main features of the inflammatory response. One of the actors of these processes is α-syn accumulation. In light of this, here, we provide a systematic review of PD-related α-syn and inflammation inter-players.

1. Introduction

The reduction of striatal dopaminergic neurons triggers motor symptoms that include bradykinesia, uncontrollable tremor at rest, postural impairment, and rigidity, which together characterise Parkinson’s disease (PD) as a movement disorder [1,2]. Neurodegeneration in the Substantia Nigra pars compacta (SNpc) leads to a marked decrease of dopamine (DA) levels in the synaptic terminals of the dorsal striatum and the consequent loss of nigrostriatal pathway, which allows PD to be described as a synaptopathy [3,4]. Synaptopathy is linked to α-synuclein (α-syn), a small, soluble protein encoded by the SNCA gene on human chromosome 4 [5,6], which is physiologically mainly localised in the presynaptic nerve terminals [7], the mitochondrial-associated membrane (MAM) [8], in which its overexpression increases the extent of contact sites and downregulates MAM activity [9,10,11,12], and in the nucleus [13,14,15,16].
α-syn accumulation compromises the fusion and clustering activity of the synaptic vesicles [17] and then influences neurotransmitter release, inducing the death of nigrostriatal neurons [18]. The transmission of α-syn pathology crosses different brain regions [19], though the impacts of extracellular α-syn on synaptic activity remains largely unknown [20,21]. Pacheco and colleagues [22] showed that extracellular α-syn oligomers facilitate the perforation of the neuronal plasma membrane, increasing its conductance and the influx of both calcium (Ca2+) and glucose, explaining in part the synaptotoxicity observed in PD. α-syn occurs in a dynamic balance between the monomeric and oligomeric forms, which are not easily prone to form fibrils under physiological conditions. Identifying the most toxic species, between fibrils and oligomers, has been difficult. There is evidence that the formation of fibrils mediates α-syn toxicity [23]. On the contrary, oligomeric forms are considered the most toxic species at the synapses [24], where they impair long term potentiation (LTP) and increase basal synaptic transmission through a mechanism dependent on N-Methyl-D-Aspartate (NMDA) receptor activation [25,26,27,28].
PD has a multifactorial aetiology. Indeed, the possible causes depend both on genetic and environmental factors that engage several biological mechanisms and processes, such as the cited α-syn misfolding, mitochondrial dysfunction, oxidative stress, synaptic plasticity, and neuroinflammation. Neuroinflammation, in recent years, has assumed a central role in the pathophysiology of PD and other neurodegenerative diseases. Evidence from post-mortem brains of PD patients, as well as in in vitro and in in vivo models, has highlighted the inflammatory contribution to the disease’s neuropathology [29,30,31,32,33]. In 1988, McGeer and collaborators disclosed the presence of HLA-DR+ microglia (macrophages) in the SN of idiopathic PD patients through immunohistochemical staining [34]. This finding proved the existence of the reactive microglia state around dead and dying dopaminergic neurons. Moreover, these macrophages exhibited phagocytic activity, as demonstrated by the presence of melanin detritus inside them [34]. This study, like others in the recent literature, suggested the involvement of immune system alteration in PD. Moreover, GWAS (genome-wide associations studies) analyses have unveiled the contribution of both innate and adaptive immune responses [35,36,37,38]. Furthermore, data support that oligomeric and fibrillary α-syn forms can activate microglial cells [39,40], suggesting a clear role for this protein in the central inflammation in PD affecting neuronal homeostasis through the modulation of microglia function, which could be either protective or detrimental in PD.
In this review, we will address the critical role of α-syn in relationship with the microglia and astrocytes, as immune resident brain cells, in the context of PD synaptopathy.

2. Synaptopathy

Synapses work within intricate neural networks to coordinate the neuronal activity involved in learning, memory, and behaviour. Therefore, damage to the synapses will have detrimental consequences [41,42]. Synapses have a presynaptic compartment, which includes the axon terminal and their protein machinery involved in neurotransmitter release [43]. After the exocytosis of presynaptic vesicles, neurotransmitters spill out and diffuse in the extracellular synaptic cleft to reach a specific postsynaptic compartment, which is composed of protein machinery that receives and transmits the signals induced by the neurotransmitters [44].
Therefore, early or late synaptic dysfunction drives a group of neurological disorders named synaptopathies [41,45]. This category of diseases includes neurodegenerative disorders, such as PD, which is characterised by a progressive degeneration of dopaminergic neurons located in the midbrain and a decline in cognitive and behavioural functions [4]. This pathological condition has been attributed to the accumulation of α-syn aggregates in neurons having detrimental consequences for neuronal integrity [23,46].
In the first part of this review, we focus on emphasising the current knowledge on synaptopathy and synaptic dysfunction induced by α-syn, and its role in the early stages of PD progression. Then, we describe the relevant role of the glial inflammatory reaction in the pathophysiology of PD, related to the detrimental effects of α-syn.

2.1. α-Synucleinopathy

The neuropathological hallmarks of PD are the progressive loss of dopaminergic neurons, specifically in the SNpc, and the presence of intraneuronal α-syn inclusions, termed Lewy bodies (LBs) and Lewy neurites (LNs) [47,48,49]. Synaptopathy is closely linked to α-syn, a small soluble protein that it is physiologically localised in the presynaptic terminals, compromising the fusion and clustering activity of the synaptic vesicles, and then influencing neurotransmitter release [50,51,52]. The primary presence of α-synucleinopathy is in the synaptic terminals, which results in an early synaptic impairment that precedes axon degeneration, with a following retrograde progression [53,54,55]. In physiological conditions (Figure 1), α-syn monomers allow neurotransmitter release through the modulation of synaptic vesicle maturation and release [46]. However, under pathological conditions, α-syn monomers undergo post-translational modifications, developing toxic α-syn oligomers and fibrils, which are considered the most toxic species of α-syn [56]. These toxic α-syn fibril aggregates determine the onset of synapse deconstruction, and subsequent neurodegeneration [17,57,58]. The first evidence that axons are affected in PD came from the pioneer study performed by Braak and collaborators. The authors were able to demonstrate that α-syn inclusions were not only present at the level of neuronal soma, but also in axonal processes [59]. In addition, in 2003, they demonstrated that extracellular misfolded α-syn could propagate from neurons to glial cells in a prion-like manner [60]. Moreover, Kordower and collaborators [61] discovered the presence of α-syn pathology in dopaminergic neurons implanted about 14 years earlier in PD patients, confirming Braak’s theory. Investigations in animal models and in vitro experiments corroborated the prion-like propagation hypothesis. Pre-formed fibrils (PFFs) of α-syn, in cell culture [62] or injected in rodent brains, engaged endogenous α-syn, producing pathologic inclusions and/or replaying PD features, such as inflammation and DA degeneration [27,63,64,65,66,67].
Specifically, α-syn is localised to presynaptic boutons and is considered as a causative factor for the onset of PD. During the early stage of the aggregation process, it affects synaptic terminals, preceding the loss of striatal dopaminergic neurons, which is considered the most important feature of neurodegeneration in the SN [3,17]. The high association of α-syn with the synaptic vesicles suggests its physiological involvement in the regulation of synaptic transmission, as well as in almost every step of synaptic vesicle recycling, including trafficking, docking, fusion, and recycling after exocytosis [57,68,69]. In fact, alterations in striatal dopamine (DA) release are not correlated with the total amount of DA on striatal tissue or to cell death. Rather, they seem to be caused by functional impairment of neurotransmitter release at the synapse [70]. Garcia-Reitbock and collaborators [71] have demonstrated that in a transgenic α-syn (1–120) mouse model, the protein aggregates were present at striatal dopaminergic terminals and the release of DA from nigrostriatal synaptic terminals was progressively impaired, with nigral dopaminergic neuron loss.

2.2. α-Synuclein Induces Protein Synapse Dysfunctions

Accumulation of pathologic α-syn leads to reductions in synaptic proteins, and progressive impairments in neuronal network function and connectivity, causing neuron death. Burrè and collaborators [72] have shown that α-syn cooperates with cysteine-string protein α (CSPα) to promote SNARE complex assembly and support its folding through direct binding to vesicle-associated membrane protein 2 (VAMP-2)/synaptobrevin 2 on synaptic vesicles. In physiological conditions (Figure 1), α-syn is also important for the maintenance and redistribution of the SNARE complex, which is directly involved in neurotransmitter release [54,72]. Accumulated α-syn alters the levels and localization of SNARE proteins at the presynaptic nerve terminals. These findings suggest that α-syn accumulation causes toxicity to synaptic functions, impairing their connectivity. The presence of large α-syn oligomers inhibits SNARE complex formation, blocking vesicle docking, which is an important process for physiological exocytosis [73]. In 2011, Volpicelli-Daley and collaborators demonstrated, in an in vitro α-syn toxicity model, that the incubation of α-syn fibrils on primary neuronal cultures caused loss of the synaptic vesicle proteins SNARE complex, VAMP2, and synaptosomal-associated protein-25 (SNAP-25), as well as CSPα and synapsin-2 [62]. The researchers further investigated the impact of this preformed α-syn fibrils accumulation on neural network activity. They found that impaired hippocampal network activity occurred prior to the reduction of synaptic protein levels, showing the key role of pathological α-syn on neuronal coordination and connectivity [62]. Therefore, the interactions between aggregated α-syn and synaptic proteins could be considered the molecular basis for functional deficits of synapses in α-synucleinopathies.

2.3. α-Synuclein Induces Synaptic Dysfunction

In PD, synaptic dysfunction precedes neurodegeneration, and in the last twenty years, research has focused on identifying early synaptic alterations, considering not only the presynaptic aspect but also the postsynaptic. Impairment of DA transmission in the hippocampus is not attributed to the loss of dopaminergic cells, but to functional release changes and to the lack of long-term synaptic plasticity—such as LTP—expression, accompanied by a decrease in synaptic NMDA receptors and the GluN2A/GluN2B receptor subunit ratio, caused by the early accumulation of α-syn in the presynaptic terminals [25].
Several studies have indicated that both GluN2A- and GluN2B-containing receptors can be located in either synaptic or extrasynaptic compartments [74,75,76], and the overexpression of α-syn increases the phosphorylation of these glutamate receptor subunits [77]. Subsequent studies focused on analysing the impact of α-syn inclusion formation on excitatory hippocampal neuronal function at different time points, to verify if its interference precedes neuron death. Froula and collaborators [78] found that the formation of protofibril α-syn inclusions increased the frequency of miniature excitatory presynaptic currents action potential-independent (mEPSCs), with an increased number of docked presynaptic vesicles, despite a reduction in dendritic spine density. On the other hand, the frequency or amplitude of spontaneous excitatory postsynaptic currents (sEPSCs) was unaffected [78]. Therefore, these data suggest there are underlying compensatory mechanisms that preserve normal levels of spontaneous synaptic activity driven by action potentials. In hippocampal slices, α-syn oligomers, but not monomers or fibrils, impair LTP and increase basal synaptic transmission through a mechanism dependent on NMDA receptor activation, triggering an enhanced contribution of GluA1-containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors [26]. Interesting studies have demonstrated that the overexpression of C-terminally cleaved human α-syn in mice induces several neuropathological PD changes that are not related to dopaminergic neuron loss [25,79]. Navarria and colleagues [80] have shown a functional interaction between α-syn and NMDA receptors, notably with the GluN2B receptor subunit, which is particularly involved in PD [74,81]. The data showed that the membrane translocation and function of GluN2B can be negatively modulated by α-syn, contributing to neuronal degeneration.
Moreover, α-syn is highly expressed in cholinergic neurons of the pedunculo pontine nucleus and basal forebrain, in which its alterations in the neural network could lead to cognitive deficits. α-syn is also expressed in GABAergic presynaptic terminals of the globus pallidus and Substantia Nigra pars reticulata (SNpr), arising probably from spiny projection neurons (SPNs) of the striatum [82].
The striatum is a subcortical nucleus particularly involved in PD, and it receives strongly excitatory inputs from the cortex and the thalamus [83]. Several studies have demonstrated that the oligomeric forms of α-syn affect synaptic transmission of different neuronal subtypes or synaptic sites, causing motor and behaviour deficits typical of PD [27,28,84]. Tozzi and collaborators [28] have recently shown that the LTP of excitatory synaptic transmission of striatal cholinergic interneurons is compromised by incubation of a nanomolar dose of exogenous human α-syn oligomers (3 nM) on striatal rat slices. The authors linked the loss of LTP in striatal cholinergic interneurons with the close interaction of α-syn and the reduction of GluN2D-mediated NMDA receptor currents. Several years later, the same research group described how the incubation of a high oligomeric α-syn concentration (30 nM) reduced NMDA receptor-mediated synaptic currents and impaired the corticostriatal LTP of striatal SPNs. Furthermore, the treatment of the striatal slices with antibodies targeting α-syn prevented both the α-syn-induced loss of LTP and the reduced synaptic localisation of the GluN2A NMDA receptor subunit [27]. The result of these studies was the awareness that increasing α-syn oligomer concentrations progressively affect NMDA receptor-mediated synaptic plasticity in distinct neuronal populations, indicating that vulnerability to this protein is cell- and region-specific. Another interesting study by Trudel and colleagues [84] recently affirmed that α-syn oligomers induce Ca2+-dependent release of glutamate from astrocytes and found that mice overexpressing α-syn manifest increased tonic release of glutamate in vivo. This extracellular glutamate release activates specific receptors, including extrasynaptic NMDA receptors (eNMDARs), and oligomerised α-syn can directly activate these receptors, contributing to neuronal damage.

3. α-Synuclein and Its Role in Neuroinflammation in Parkinson’s Disease

As mentioned above, α-syn represents a pathological hallmark of PD, in particular intraneuronal inclusions known as LBs and/or LNs [49]. During brain physiological activity, α-syn is specially found within the presynaptic terminal of neurons belonging to the neocortex, striatum, hippocampus, thalamus, and cerebellum [85,86,87], whose cellular function has not yet been clarified, even if its involvement in synaptic plasticity and in the release of neurotransmitters and synaptic vesicles has been recognised, as extensively discussed in the previous section of this review.
The SNCA gene encodes α-syn, and can undergo missense mutations (A53T, A30T and E46K) and multiplication (duplications and triplications), which demonstrates the key role of this protein in PD [6,88,89,90,91,92,93,94]. As previously mentioned, under such pathological conditions, α-syn can be overexpressed and acquire a misfolded conformation. These misfolding species can accumulate, because of impaired autophagy or reduced phagocytic clearance [85,95,96], and could assume aggregated forms, such as oligomers or protofibrils, that cause acute toxicity in the brains of PD patients [97,98]. α-syn aggregates to resist degradation and to prompt, as shown in in vitro experiments, the impairment of macroautophagy, reducing autophasome clearance and promoting dopaminergic neuron death [99]. Moreover, post-translational modifications (ubiquitination, nitration, and phosphorylation) facilitate the formation of these aggregate species, increasing the disease process [85,96,100,101]. However, α-syn is not only a citoplasmatic protein, but can also be found in the extracellular space [102]. Neuronal cells normally throw out α-syn in the extracellular space, which impairs biological processes, such as oxidative stress or mitochondrial and lysosomal dysfunction, amplifying its release [103,104,105,106,107,108]. Thus, extracellular α-syn, probably secreted through exosomes and exocytotic vesicles [103,104,105,106,107,108], could act as damaged-associated molecular patterns (DAMPs), triggering the immune system and neuroinflammation processes [37,109]. As previously indicated, PFF aggregates could activate endogenous α-syn, triggering inflammatory and neurodegenerative responses and, in the final instance, PD pathology [27,63,64,65,66,67]. Thus, in the context of nigrostriatal degeneration in PD, misfolded α-syn species could be considered as being strictly associated with neuroinflammatory events, acting as an antigen capable of activating immune molecules.

4. Innate Immunity

The innate immune system represents the first line of defence in the CNS against cellular debris, protein aggregates, and foreign invading pathogens. It acts by non-specific mechanisms and triggers the adaptive system to help fight. Interestingly, immune cells (microglia, monocytes, NK cells, B cells, T cells) express many of the genes involved in the genetic risk of PD, including SNCA, LRRK2, DJ-1, and Parkin [110].

4.1. Microglia

Microglia are the largest population of resident immune cells in the brain. These macrophage-like cells have many functions in the CNS [97,111,112]. First, they monitor the surrounding environment with their elongated processes to protect it against non-self-agents. Moreover, microglial cells also have important functions in brain development, controlling neuron survival, apoptosis of neuronal subpopulations, synaptic pruning, maturation, and regulating the number of synapses. Microglia influence synaptic transmission and plasticity through several mechanisms and “eat me” signals expressed by neurons, such as chemokine CX3CL1 and the classical complement proteins C1q and C3 [83]. Moreover, microglia produce soluble immune agents—Brain-derived neurotrophic factor (BDNF) and IL-1b—capable of modifying synaptic plasticity (Figure 1). Lastly, the production of the oxidative stress mediator dihydronicotinamide-adenine dinucleotide phosphate (NADPH) oxidase induces the alteration of neuronal properties [83].
Microglia express phagocytic activity by deleting dead neurons, protein aggregates, synapses, pathogens, and other antigens that could damage the CNS [97]. These agents are defined as pathogen-associated molecular patterns (PAMPs), or DAMPs [92], which activate microglia. Microglia then change their morphology, moving from the “resting” state, characterised by a round cell body and long and thin processes, to the “activated” state, in which they assume a large cell body and amoeboid shape, with thicker extensions [113,114]. Moreover, this latter phenotype, named M1, exhibits an increased number of major histocompatibility complex (MHC-I) and MHC-II molecules, and amplified levels of pro-inflammatory species, such as chemokines and cytokines (IL1, IL6, TNF). M1 microglial cells affect blood brain barrier (BBB) homeostasis, causing infiltration of peripheral immune cells and increased inflammation [97,111,112]. Interestingly, there is an alternative activated state, termed the M2 microglia, that, on the contrary, expresses anti-inflammatory agents, such as cytokines (IL4 and IL10), pro-resolving mediators, and resolvins [111].
Understanding of α-syn involvement in the activation of microglia originated from a study that provided the first evidence of an inflammatory response in the BV2 cell line [112,115]. Different research groups have demonstrated the release of proinflammatory factors from microglia following injections of PFFs of synthetic α-syn [109,112,116]. The activation of microglia by α-syn increases both microglial phagocytic activity and the spread of α-syn in a prion-like manner [97,117,118,119]. As described previously, extracellular α-syn activates microglia through toll-like receptor 2 (TRL2), acting as a DAMP, and the presence of fibrillar α-syn induces activation of NF-kB [37,109]. This mechanism is very important for the microglia inflammatory response, and also the NLRP3 (nucleotide-binding oligomerization domain leucine-rich repeat and pyrin domain-containing protein 3) inflammasome [112]. NLRP3 induces the production of IL-1 and IL-18 pro-inflammatory cytokines [37] through caspase 1 proteolytic activity [112]. Moreover, caspase 1 activation causes truncation of α-syn, which forms more aggregates, increasing inflammation [112,120]. However, the evidence regarding NLRP3 in PD is still limited, and further detailed studies are required.

4.2. Astroglia

Like microglia, astrocytes play a crucial role in neuroinflammation. They represent the most abundant type of cell in the brain and are identified through their recognisable star shape with elongated extensions [111,121,122]. They are essential to maintain homeostasis in the CNS, through the structural support of neuronal cells, preservation of the permeability of the BBB, release of neurotrophic factors, clearance of neurotransmitters, synapse formation and pruning, phagocytic activity, and involvement in synaptic plasticity [111,121]. Indeed, they form the “tripartite synapse”, in which astrocytes participate actively in the exchange of synaptic information between the pre- and post-synapses, regulating plasticity through the release of gliotransmitters [123,124] via a Ca2+-dependent release.
Many studies have highlighted the reaction of astrocytes in in vitro and in vivo models of PD, as well as in post-mortem brains of PD patients [85,125,126,127,128,129,130].
In disease conditions, when astrocytes are activated, they change their morphology and gene expression. There are two types of astrocytes: A1 reactive and A2 neuroprotective types. A1 type astrocytes represent reactive microglia-induced cells in stress or disease conditions and are induced by the release of IL-1, TNF and C1q [111,121]. These reactive cells are detected in injured brain regions, where they lose normal functions, release proinflammatory factors (IL-1, IL-1, and TNF), contribute to inflammatory neurodegeneration, and increase their expression of a cytoskeletal protein called Glial Fibrillary Acid Protein (GFAP) [121,131]. However, there are contradictory results from experiments conducted on animals or the post-mortem brains of PD patients. Studies on human PD tissues have displayed null or mild increases of astrocytes and/or GFAP immunoreactivity [131,132,133,134,135]. On the contrary, Parkinsonian animal models undergoing 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or 6-hydroxydopamine (6-OHDA) treatment have exhibited dramatic astrogliosis [131,136,137].
A2 astrocytes are defined as a neuroprotective type and secrete neurotrophic factors, such as nerve growth factor (NGF) and BDNF [111,138]. As mentioned above, the A1 astrocyte population is a feature of aging and neurodegenerative diseases, such as PD and Alzheimer’s disease (AD). Phagocytic activity is one of the normal functions lost by A1 reactive astrocytes. This astrocytic activity is important to delete neuronal debris and protein over-expression. Through this process, astrocytes can “ingest” a considerable quantity of α-syn fibrils, as demonstrated by in vitro studies [121,139]. Notably, astrocytes participate in the spreading of α-syn, which can be released from neurons, taken up by astrocytes, and transferred to another astrocyte via extracellular vesicles or exosomes [70,121,140]. In addition, astrocytes have neurotransmitter receptors and Na+-dependent excitatory amino acid transporters, such as glutamate transporter 1 (GLT-1), excitatory amino acid transporter 2 (EAAT2), and glutamate aspartate transporter (GLAST). Furthermore, they participate in glutamate re-uptake and release, defending neurons from glutamate spill over and excitotoxicity [131,141]. Moreover, proinflammatory factors, secreted by microglia, affect glutamatergic signals, e.g., reducing the expression of glutamate transporters [131,142,143].

5. Adaptive Immunity

As seen previously, neuroinflammation is a complex cerebral phenomenon that involves not only innate immunity, but also the adaptive immune system, arising from periphery due to the alteration of the BBB [141,144,145]. In the past, the brain was considered a privileged organ which was not susceptible to infiltration by peripheral leukocytes, through BBB activity [110,115,146]. However, in the last few years, many research groups have highlighted the presence of T cells in the post-mortem brains of people with neurodegenerative disorders, such as in PD patients or in experimental animal models [64,110,147].
Positron emission tomography (PET) diagnostic analysis in PD patients has pointed out the close relationship between BBB alteration, infiltration of CD+4 and CD+8 lymphocytes, and neuronal loss [111,148].
Indeed, microglia seem to induce the activation of T cells, acting as antigen presenting cells (APCs). Microglia activate CD4+ expressing antigen through MHCII. MHCI activates CD8+ lymphocytes, and is expressed by all cells of the organism [149]. CD4+ species, recruited from the periphery, infiltrate the CNS and can exhibit both an anti- (Th2 and Tregs) or pro-inflammatory phenotypes (Th1 and Th17). During PD progression with dopaminergic cell loss, microglia and immune cells (CD+4 cells, macrophages) undergo alteration of the neurotransmitter balance (DA levels are reduced in SNpc and cortical glutamatergic transmission is over-expressed), increasing neuroinflammation [150].

6. α-Synuclein and Glia Cells in Parkinson’s Disease Synaptopathy

Neuronal transmission at corticostriatal synapses is altered in PD [151], due to the impaired afflux of DA in striatum that leads to LTP and LTD loss [151]. In this scenario, synaptopathy seems to strictly be linked to α-syn aggregated species [83]. Moreover, the immune system seems to be involved in the α-syn-induced PD synaptopathy. Resident immune cells in the brain may regulate synaptic transmission. As seen above, microglia and astrocytes monitor the surrounding environment, but when activated, for example by α-syn aggregation, they produce cytokines, chemokines, neurotransmitters, and growth factors, through which they can modulate neuronal plasticity. Indeed, many studies have demonstrated an increase of cytokine levels in the striatum and CSF of Parkinsonian patients. Interestingly, these molecules are involved in the synaptic plasticity process [151].
Moreover, most of the immune species, belonged to both innate and adaptive systems, expressed dopaminergic receptors and the necessary machinery to synthesize, metabolize and store up DA [76,111,152]. Under physiological conditions, in the nigrostriatal pathway, in which the DA concentration is normally high, this could stimulate low-affinity receptors (D1R and D2R) and exert anti-inflammatory effects [76]. When DA levels are decreased, as shown in PD, there is a stimulation of high-affinity DA receptors (in particular, DA D3R), which can have pro-inflammatory effects that lead to inflammation and consequent neurodegeneration in the brain [76]. Remarkably, Duffy and collaborators, in 2018, studied the activation of microglia after synthetic α-syn-PFF unilateral injections into the striatum. They found out that microglia reactions occurred months before cell death in the SNpc [153], suggesting early activation of the immune system in the brain during PD progression. Additionally, astrocytes are activated by α-syn misfolding, inducing chemokine and cytokine production via TRL4. Moreover, α-syn leads to neurotoxicity following Ca2+ flux and oxidative stress [96].
All together, these findings represent a complex scenario in which α-syn aggregation triggers the neuroinflammatory process, which in turn modulates synaptic activity through the production of soluble immune molecules.

7. Neuroinflammation as a Therapeutic Target in Parkinson’s Disease

Neuroinflammation is considered an early event in the pathophysiology of PD, or a driving mechanism of its progression. Thus, it could be an early therapeutic target to counteract PD. Many recent studies have described the use of different agents that act on inflammatory species [97,111]. In the past few years, it has been hypothesized that there is a correlation between the chronic use of non-steroidal anti-inflammatory drugs (NSAIDs) and reduced risk of developing PD [29,154]. Different meta-analyses have presented conflicting data. In particular, some studies did not find any correlation between the use of anti-inflammatory drugs and PD risk [155,156]. Other research studies showed a beneficial outcome for the use of non-aspirin NSAIDs [157,158], or aspirin in females [159]. Growing evidence supports the importance of neuroinflammation as a therapeutic target, with the aim of limiting disease progression.
Several clinical trials have been conducted in the last few years in this field (https://clinicaltrials.gov (accessed on 10 May 2021)). Immunotherapies against α-syn have been developed to reduce extracellular α-syn levels, which are responsible for triggering neuroinflammation, and its spread. There are two types of immunotherapies: active and passive [160,161].
Active immunotherapy, or vaccination, involves the production of antibodies against α-syn through the patient’s immune system. AFFITOPE® AFF1 belongs to this category, and is characterised by administration of small fragments of α-syn. This immunotherapy has successfully reduced neurodegeneration and increased anti-inflammatory factors in two animal models [111]. AFFITOPE® AFF1 is currently being considered in a phase I clinical study (NCT02267434) [111,162].
Passive immunisation involves the administration of antibodies against α-syn, and has achieved good results in pre-clinical studies by increasing the clearance of α-syn [162]. A phase 2 clinical trial that uses PRX002 as a passive immunotherapy [111,162] is currently underway.
Immunotherapies could be a valid target for therapeutic approaches, either used in combination with current medications or even as an alternative treatment in the future.

8. Conclusions

Synaptic damage and neuronal loss are major neuropathological features of PD. Misfolded α-syn aggregates are associated with disease progression; this is known as synucleinopathy. In the non-pathological brain, α-syn is not toxic and participates in several functions associated with neurotransmission and synaptic plasticity, including synaptic vesicle recycling and neurotransmitter synthesis and release. Alterations to the conformity of α-syn lead to the beginning of pathological processes in synucleinopathies. To date, many studies have identified the functions of α-syn in the regulation of neurotransmission and synaptic plasticity, but further insights are needed to outline its pathological roles. Synaptopathy, α-syn misfolding/aggregation, and neuroinflammation processes seem to interact and contribute to PD pathogenesis. As demonstrated by many of the studies cited above, these mechanisms affect each other, creating a vicious circle in which it is difficult to establish the first pathology trigger. The involvement of α-syn pathology and an altered immune response point to potential new immunomodulatory targets to slow down the progression of the disease, and/or improve its outcome.

Author Contributions

Conceptualization, A.C., B.P. and V.C.; writing—original draft preparation, A.C. and V.C.; writing—review and editing, B.P.; figures preparation, A.d.I. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Italian Ministry of Health, Ricerca Corrente (B.P.).

Acknowledgments

This work was supported by the Italian Ministry of Health, Ricerca Corrente (B.P.), and by a grant from NYU Grossman School of Medicine and The Marlene and Paolo Fresco Institute for Parkinson’s and Movement Disorders, which was made possible with support from Marlene and Paolo Fresco (A.C.).

Conflicts of Interest

No conflicts of interest, financial or otherwise, are declared by the authors.

Abbreviations

6-OHDA6-hydroxydopamine
ADAlzheimer’s disease
AMPAα-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
APCAntigen presenting cell
BBBBlood brain barrier
BDNFBrain-derived neurotrophic factor
CNSCentral Nervous System
CSPαCysteine-string protein α
DADopamine
DAMPsDamage associated molecular patterns
EAAT2Excitatory amino acid transporter 2
eNMDARsExtrasynaptic NMDA receptors
GFAPGlial Fibrillary Acid Protein
GLASTGLutamateASpartate Transporter 1
GLT-1Glutamate transporter 1
GWASGenome-wide associations studies
IL-nInterleukin n
LBsLewy bodies
LNsLewy neurites
LTPLong term potentiation
MHCMajor histocompatibility complex
MPTP1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
NADPHDihydronicotinamide-adenine dinucleotide phosphate
NF-kBNuclear factor kappa-light-chain-enhancer of activated B cells
NGFNerve growth factor
NLRP3Nucleotide-binding oligomerization domain leucine-rich repeat and pyrin domain-containing protein 3
NMDAN-methyl-d-aspartate
NSAIDsNon-steroidal anti-inflammatory drugs
PAMPsPathogen associated molecular patterns
PDParkinson’s Disease
PFFsPre-formed fibrils
SNAP-25Synaptosomal-associated protein-25
SNpcSubstantia nigra pars compacta
SPNsSpiny projection neurons
TRL2Toll-like receptor 2
TNFTumor necrosis factor
VAMP-2Vesicle-associated membrane protein-2
α-synα-synuclein

References

  1. Obeso, J.A.; Stamelou, M.; Goetz, C.G.; Poewe, W.; Lang, A.E.; Weintraub, D.; Burn, D.; Halliday, G.M.; Bezard, E.; Przedborski, S.; et al. Past, present, and future of Parkinson’s disease: A special essay on the 200th anniversary of the shaking palsy. Mov. Disord. 2017, 32, 1264–1310. [Google Scholar] [CrossRef]
  2. Surmeier, D.J. Determinants of dopaminergic neuron loss in Parkinson’s disease. FEBS J. 2018, 285, 3657–3668. [Google Scholar] [CrossRef] [Green Version]
  3. Cheng, F.; Vivacqua, G.; Yu, S. The role of alpha-synuclein in neurotransmission and synaptic plasticity. J. Chem. Neuroanat. 2011, 42, 242–248. [Google Scholar] [CrossRef]
  4. Picconi, B.; Hernandez, L.F.; Obeso, J.A.; Calabresi, P. Motor complications in Parkinson’s disease: Striatal molecular and electrophysiological mechanisms of dyskinesias. Mov. Disord. 2018, 33, 867–876. [Google Scholar] [CrossRef]
  5. Pasanen, P.; Myllykangas, L.; Siitonen, M.; Raunio, A.; Kaakkola, S.; Lyytinen, J.; Tienari, P.J.; Poyhonen, M.; Paetau, A. Novel alpha-synuclein mutation A53E associated with atypical multiple system atrophy and Parkinson’s disease-type pathology. Neurobiol. Aging 2014, 35, 2180.e1–2180.e5. [Google Scholar] [CrossRef]
  6. Polymeropoulos, M.H.; Lavedan, C.; Leroy, E.; Ide, S.E.; Dehejia, A.; Dutra, A.; Pike, B.; Root, H.; Rubenstein, J.; Boyer, R.; et al. Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science 1997, 276, 2045–2047. [Google Scholar] [CrossRef] [Green Version]
  7. Kramer, M.L.; Schulz-Schaeffer, W.J. Presynaptic alpha-synuclein aggregates, not Lewy bodies, cause neurodegeneration in dementia with Lewy bodies. J. Neurosci. 2007, 27, 1405–1410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Guardia-Laguarta, C.; Area-Gomez, E.; Rub, C.; Liu, Y.; Magrane, J.; Becker, D.; Voos, W.; Schon, E.A.; Przedborski, S. Alpha-synuclein is localized to mitochondria-associated ER membranes. J. Neurosci. 2014, 34, 249–259. [Google Scholar] [CrossRef] [PubMed]
  9. Cali, T.; Ottolini, D.; Negro, A.; Brini, M. Alpha-synuclein controls mitochondrial calcium homeostasis by enhancing endoplasmic reticulum-mitochondria interactions. J. Biol. Chem. 2012, 287, 17914–17929. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Guardia-Laguarta, C.; Area-Gomez, E.; Schon, E.A.; Przedborski, S. Novel subcellular localization for alpha-synuclein: Possible functional consequences. Front. Neuroanat. 2015, 9, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Cali, T.; Ottolini, D.; Vicario, M.; Catoni, C.; Vallese, F.; Cieri, D.; Barazzuol, L.; Brini, M. SplitGFP technology reveals dose-dependent ER-mitochondria interface modulation by alpha-Synuclein A53T and A30P mutants. Cells 2019, 8, 1072. [Google Scholar] [CrossRef] [Green Version]
  12. Hijaz, B.A.; Volpicelli-Daley, L.A. Initiation and propagation of alpha-synuclein aggregation in the nervous system. Mol. Neurodegener. 2020, 15, 19. [Google Scholar] [CrossRef]
  13. Pinho, R.; Paiva, I.; Jercic, K.G.; Fonseca-Ornelas, L.; Gerhardt, E.; Fahlbusch, C.; Garcia-Esparcia, P.; Kerimoglu, C.; Pavlou, M.A.S.; Villar-Pique, A.; et al. Nuclear localization and phosphorylation modulate pathological effects of alpha-synuclein. Hum. Mol. Genet. 2019, 28, 31–50. [Google Scholar] [CrossRef] [PubMed]
  14. Desplats, P.; Spencer, B.; Coffee, E.; Patel, P.; Michael, S.; Patrick, C.; Adame, A.; Rockenstein, E.; Masliah, E. Alpha-synuclein sequesters Dnmt1 from the nucleus: A novel mechanism for epigenetic alterations in Lewy body diseases. J. Biol. Chem. 2011, 286, 9031–9037. [Google Scholar] [CrossRef] [Green Version]
  15. Savyon, M.; Engelender, S. SUMOylation in alpha-synuclein homeostasis and pathology. Front. Aging Neurosci. 2020, 12, 167. [Google Scholar] [CrossRef] [PubMed]
  16. Mavroeidi, P.; Xilouri, M. Neurons and glia Interplay in α-synucleinopathies. Int. J. Mol. Sci. 2021, 22, 4994. [Google Scholar] [CrossRef] [PubMed]
  17. Longhena, F.; Faustini, G.; Missale, C.; Pizzi, M.; Spano, P.; Bellucci, A. The contribution of alpha-synuclein spreading to Parkinson’s disease synaptopathy. Neural Plast. 2017, 2017, 5012129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Imbriani, P.; Schirinzi, T.; Meringolo, M.; Mercuri, N.B.; Pisani, A. Centrality of early synaptopathy in Parkinson’s disease. Front. Neurol. 2018, 9, 103. [Google Scholar] [CrossRef] [Green Version]
  19. Del Tredici, K.; Braak, H. Review: Sporadic Parkinson’s disease: Development and distribution of alpha-synuclein pathology. Neuropathol. Appl. Neurobiol. 2016, 42, 33–50. [Google Scholar] [CrossRef]
  20. Bidinosti, M.; Shimshek, D.R.; Mollenhauer, B.; Marcellin, D.; Schweizer, T.; Lotz, G.P.; Schlossmacher, M.G.; Weiss, A. Novel one-step immunoassays to quantify alpha-synuclein: Applications for biomarker development and high-throughput screening. J. Biol. Chem. 2012, 287, 33691–33705. [Google Scholar] [CrossRef] [Green Version]
  21. El-Agnaf, O.M.; Salem, S.A.; Paleologou, K.E.; Curran, M.D.; Gibson, M.J.; Court, J.A.; Schlossmacher, M.G.; Allsop, D. Detection of oligomeric forms of alpha-synuclein protein in human plasma as a potential biomarker for Parkinson’s disease. FASEB J. 2006, 20, 419–425. [Google Scholar] [CrossRef] [PubMed]
  22. Pacheco, C.R.; Morales, C.N.; Ramirez, A.E.; Munoz, F.J.; Gallegos, S.S.; Caviedes, P.A.; Aguayo, L.G.; Opazo, C.M. Extracellular alpha-synuclein alters synaptic transmission in brain neurons by perforating the neuronal plasma membrane. J. Neurochem. 2015, 132, 731–741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Lashuel, H.A.; Overk, C.R.; Oueslati, A.; Masliah, E. The many faces of alpha-synuclein: From structure and toxicity to therapeutic target. Nat. Rev. Neurosci. 2013, 14, 38–48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Ingelsson, M. Alpha-synuclein oligomers-neurotoxic molecules in Parkinson’s disease and other Lewy body disorders. Front. Neurosci. 2016, 10, 408. [Google Scholar] [CrossRef] [Green Version]
  25. Costa, C.; Sgobio, C.; Siliquini, S.; Tozzi, A.; Tantucci, M.; Ghiglieri, V.; Di Filippo, M.; Pendolino, V.; de Iure, A.; Marti, M.; et al. Mechanisms underlying the impairment of hippocampal long-term potentiation and memory in experimental Parkinson’s disease. Brain 2012, 135, 1884–1899. [Google Scholar] [CrossRef] [Green Version]
  26. Diogenes, M.J.; Dias, R.B.; Rombo, D.M.; Vicente Miranda, H.; Maiolino, F.; Guerreiro, P.; Nasstrom, T.; Franquelim, H.G.; Oliveira, L.M.; Castanho, M.A.; et al. Extracellular alpha-synuclein oligomers modulate synaptic transmission and impair LTP via NMDA-receptor activation. J. Neurosci. 2012, 32, 11750–11762. [Google Scholar] [CrossRef] [Green Version]
  27. Durante, V.; de Iure, A.; Loffredo, V.; Vaikath, N.; De Risi, M.; Paciotti, S.; Quiroga-Varela, A.; Chiasserini, D.; Mellone, M.; Mazzocchetti, P.; et al. Alpha-synuclein targets GluN2A NMDA receptor subunit causing striatal synaptic dysfunction and visuospatial memory alteration. Brain 2019, 142, 1365–1385. [Google Scholar] [CrossRef] [Green Version]
  28. Tozzi, A.; de Iure, A.; Bagetta, V.; Tantucci, M.; Durante, V.; Quiroga-Varela, A.; Costa, C.; Di Filippo, M.; Ghiglieri, V.; Latagliata, E.C.; et al. Alpha-synuclein produces early behavioral alterations via striatal cholinergic synaptic dysfunction by interacting with GluN2D N-methyl-D-aspartate receptor subunit. Biol. Psychiatry 2016, 79, 402–414. [Google Scholar] [CrossRef]
  29. Gelders, G.; Baekelandt, V.; Van der Perren, A. Linking neuroinflammation and neurodegeneration in Parkinson’s disease. J. Immunol. Res. 2018, 2018, 4784268. [Google Scholar] [CrossRef] [Green Version]
  30. Krashia, P.; Cordella, A.; Nobili, A.; La Barbera, L.; Federici, M.; Leuti, A.; Campanelli, F.; Natale, G.; Marino, G.; Calabrese, V.; et al. Blunting neuroinflammation with resolvin D1 prevents early pathology in a rat model of Parkinson’s disease. Nat. Commun. 2019, 10, 3945. [Google Scholar] [CrossRef] [Green Version]
  31. Sanchez-Guajardo, V.; Febbraro, F.; Kirik, D.; Romero-Ramos, M. Microglia acquire distinct activation profiles depending on the degree of alpha-synuclein neuropathology in a rAAV based model of Parkinson’s disease. PLoS ONE 2010, 5, e8784. [Google Scholar] [CrossRef]
  32. Theodore, S.; Cao, S.; McLean, P.J.; Standaert, D.G. Targeted overexpression of human alpha-synuclein triggers microglial activation and an adaptive immune response in a mouse model of Parkinson disease. J. Neuropathol. Exp. Neurol. 2008, 67, 1149–1158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Watson, M.B.; Richter, F.; Lee, S.K.; Gabby, L.; Wu, J.; Masliah, E.; Effros, R.B.; Chesselet, M.F. Regionally-specific microglial activation in young mice over-expressing human wildtype alpha-synuclein. Exp. Neurol. 2012, 237, 318–334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. McGeer, P.L.; Itagaki, S.; Boyes, B.E.; McGeer, E.G. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer’s disease brains. Neurology 1988, 38, 1285–1291. [Google Scholar] [CrossRef] [PubMed]
  35. Gagliano, S.A.; Pouget, J.G.; Hardy, J.; Knight, J.; Barnes, M.R.; Ryten, M.; Weale, M.E. Genomics implicates adaptive and innate immunity in Alzheimer’s and Parkinson’s diseases. Ann. Clin. Transl. Neurol. 2016, 3, 924–933. [Google Scholar] [CrossRef]
  36. Hamza, T.H.; Zabetian, C.P.; Tenesa, A.; Laederach, A.; Montimurro, J.; Yearout, D.; Kay, D.M.; Doheny, K.F.; Paschall, J.; Pugh, E.; et al. Common genetic variation in the HLA region is associated with late-onset sporadic Parkinson’s disease. Nat. Genet. 2010, 42, 781–785. [Google Scholar] [CrossRef]
  37. Kam, T.I.; Hinkle, J.T.; Dawson, T.M.; Dawson, V.L. Microglia and astrocyte dysfunction in Parkinson’s disease. Neurobiol. Dis. 2020, 144, 105028. [Google Scholar] [CrossRef]
  38. Pierce, S.; Coetzee, G.A. Parkinson’s disease-associated genetic variation is linked to quantitative expression of inflammatory genes. PLoS ONE 2017, 12, e0175882. [Google Scholar] [CrossRef]
  39. Hoffmann, A.; Ettle, B.; Bruno, A.; Kulinich, A.; Hoffmann, A.C.; von Wittgenstein, J.; Winkler, J.; Xiang, W.; Schlachetzki, J.C.M. Alpha-synuclein activates BV2 microglia dependent on its aggregation state. Biochem. Biophys. Res. Commun. 2016, 479, 881–886. [Google Scholar] [CrossRef] [PubMed]
  40. Park, J.Y.; Paik, S.R.; Jou, I.; Park, S.M. Microglial phagocytosis is enhanced by monomeric alpha-synuclein, not aggregated alpha-synuclein: Implications for Parkinson’s disease. Glia 2008, 56, 1215–1223. [Google Scholar] [CrossRef]
  41. Ardiles, A.O.; Grabrucker, A.M.; Scholl, F.G.; Rudenko, G.; Borsello, T. Molecular and cellular mechanisms of synaptopathies. Neural. Plast. 2017, 2017, 2643943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Taoufik, E.; Kouroupi, G.; Zygogianni, O.; Matsas, R. Synaptic dysfunction in neurodegenerative and neurodevelopmental diseases: An overview of induced pluripotent stem-cell-based disease models. Open Biol. 2018, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Holler, S.; Kostinger, G.; Martin, K.A.C.; Schuhknecht, G.F.P.; Stratford, K.J. Structure and function of a neocortical synapse. Nature 2021, 591, 111–116. [Google Scholar] [CrossRef]
  44. Kennedy, M.B. Signal-processing machines at the postsynaptic density. Science 2000, 290, 750–754. [Google Scholar] [CrossRef] [PubMed]
  45. Lepeta, K.; Lourenco, M.V.; Schweitzer, B.C.; Martino Adami, P.V.; Banerjee, P.; Catuara-Solarz, S.; de La Fuente Revenga, M.; Guillem, A.M.; Haidar, M.; Ijomone, O.M.; et al. Synaptopathies: Synaptic dysfunction in neurological disorders—A review from students to students. J. Neurochem. 2016, 138, 785–805. [Google Scholar] [CrossRef] [PubMed]
  46. Ghiglieri, V.; Calabrese, V.; Calabresi, P. Alpha-synuclein: From early synaptic dysfunction to neurodegeneration. Front. Neurol. 2018, 9, 295. [Google Scholar] [CrossRef] [Green Version]
  47. Bellucci, A.; Mercuri, N.B.; Venneri, A.; Faustini, G.; Longhena, F.; Pizzi, M.; Missale, C.; Spano, P. Review: Parkinson’s disease: From synaptic loss to connectome dysfunction. Neuropathol. Appl. Neurobiol. 2016, 42, 77–94. [Google Scholar] [CrossRef] [Green Version]
  48. Croisier, E.; Moran, L.B.; Dexter, D.T.; Pearce, R.K.; Graeber, M.B. Microglial inflammation in the parkinsonian substantia nigra: Relationship to alpha-synuclein deposition. J. Neuroinflamm. 2005, 2, 14. [Google Scholar] [CrossRef] [Green Version]
  49. Spillantini, M.G.; Schmidt, M.L.; Lee, V.M.; Trojanowski, J.Q.; Jakes, R.; Goedert, M. Alpha-synuclein in Lewy bodies. Nature 1997, 388, 839–840. [Google Scholar] [CrossRef]
  50. Burre, J. The Synaptic function of alpha-synuclein. J. Parkinson Dis. 2015, 5, 699–713. [Google Scholar] [CrossRef] [Green Version]
  51. Eisbach, S.E.; Outeiro, T.F. Alpha-synuclein and intracellular trafficking: Impact on the spreading of Parkinson’s disease pathology. J. Mol. Med. 2013, 91, 693–703. [Google Scholar] [CrossRef]
  52. Picconi, B.; Piccoli, G.; Calabresi, P. Synaptic dysfunction in Parkinson’s disease. Adv. Exp. Med. Biol. 2012, 970, 553–572. [Google Scholar] [PubMed]
  53. Calo, L.; Wegrzynowicz, M.; Santivanez-Perez, J.; Grazia Spillantini, M. Synaptic failure and alpha-synuclein. Mov. Disord. 2016, 31, 169–177. [Google Scholar] [CrossRef]
  54. Chandra, S.; Gallardo, G.; Fernandez-Chacon, R.; Schluter, O.M.; Sudhof, T.C. Alpha-synuclein cooperates with CSPalpha in preventing neurodegeneration. Cell 2005, 123, 383–396. [Google Scholar] [CrossRef]
  55. Wu, Q.; Takano, H.; Riddle, D.M.; Trojanowski, J.Q.; Coulter, D.A.; Lee, V.M. Alpha-synuclein (alphaSyn) preformed fibrils induce endogenous alphaSyn aggregation, compromise synaptic activity and enhance synapse loss in cultured excitatory hippocampal neurons. J. Neurosci. 2019, 39, 5080–5094. [Google Scholar] [CrossRef] [Green Version]
  56. Bras, I.C.; Xylaki, M.; Outeiro, T.F. Mechanisms of alpha-synuclein toxicity: An update and outlook. Prog. Brain Res. 2020, 252, 91–129. [Google Scholar] [PubMed]
  57. Bridi, J.C.; Hirth, F. Mechanisms of alpha-synuclein induced synaptopathy in Parkinson’s disease. Front. Neurosci. 2018, 12, 80. [Google Scholar] [CrossRef] [Green Version]
  58. Peelaerts, W.; Bousset, L.; Van der Perren, A.; Moskalyuk, A.; Pulizzi, R.; Giugliano, M.; Van den Haute, C.; Melki, R.; Baekelandt, V. Alpha-synuclein strains cause distinct synucleinopathies after local and systemic administration. Nature 2015, 522, 340–344. [Google Scholar] [CrossRef] [PubMed]
  59. Braak, H.; Sandmann-Keil, D.; Gai, W.; Braak, E. Extensive axonal Lewy neurites in Parkinson’s disease: A novel pathological feature revealed by alpha-synuclein immunocytochemistry. Neurosci. Lett. 1999, 265, 67–69. [Google Scholar] [CrossRef]
  60. Braak, H.; Del Tredici, K.; Rub, U.; de Vos, R.A.; Jansen Steur, E.N.; Braak, E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol. Aging 2003, 24, 197–211. [Google Scholar] [CrossRef]
  61. Kordower, J.H.; Chu, Y.; Hauser, R.A.; Freeman, T.B.; Olanow, C.W. Lewy body-like pathology in long-term embryonic nigral transplants in Parkinson’s disease. Nat. Med. 2008, 14, 504–506. [Google Scholar] [CrossRef]
  62. Volpicelli-Daley, L.A.; Luk, K.C.; Patel, T.P.; Tanik, S.A.; Riddle, D.M.; Stieber, A.; Meaney, D.F.; Trojanowski, J.Q.; Lee, V.M. Exogenous alpha-synuclein fibrils induce Lewy body pathology leading to synaptic dysfunction and neuron death. Neuron 2011, 72, 57–71. [Google Scholar] [CrossRef] [Green Version]
  63. Couch, Y.; Alvarez-Erviti, L.; Sibson, N.R.; Wood, M.J.; Anthony, D.C. The acute inflammatory response to intranigral alpha-synuclein differs significantly from intranigral lipopolysaccharide and is exacerbated by peripheral inflammation. J. Neuroinflamm. 2011, 8, 166. [Google Scholar] [CrossRef] [Green Version]
  64. Harms, A.S.; Delic, V.; Thome, A.D.; Bryant, N.; Liu, Z.; Chandra, S.; Jurkuvenaite, A.; West, A.B. Alpha-synuclein fibrils recruit peripheral immune cells in the rat brain prior to neurodegeneration. Acta Neuropathol. Commun. 2017, 5, 85. [Google Scholar] [CrossRef] [Green Version]
  65. Masuda-Suzukake, M.; Nonaka, T.; Hosokawa, M.; Kubo, M.; Shimozawa, A.; Akiyama, H.; Hasegawa, M. Pathological alpha-synuclein propagates through neural networks. Acta Neuropathol. Commun. 2014, 2, 88. [Google Scholar] [CrossRef] [Green Version]
  66. Sznejder-Pacholek, A.; Joniec-Maciejak, I.; Wawer, A.; Ciesielska, A.; Mirowska-Guzel, D. The effect of alpha-synuclein on gliosis and IL-1alpha, TNFalpha, IFNgamma, TGFbeta expression in murine brain. Pharm. Rep. 2017, 69, 242–251. [Google Scholar] [CrossRef] [PubMed]
  67. Wilms, H.; Rosenstiel, P.; Romero-Ramos, M.; Arlt, A.; Schafer, H.; Seegert, D.; Kahle, P.J.; Odoy, S.; Claasen, J.H.; Holzknecht, C.; et al. Suppression of MAP kinases inhibits microglial activation and attenuates neuronal cell death induced by alpha-synuclein protofibrils. Int. J. Immunopathol. Pharm. 2009, 22, 897–909. [Google Scholar] [CrossRef]
  68. Larsen, K.E.; Schmitz, Y.; Troyer, M.D.; Mosharov, E.; Dietrich, P.; Quazi, A.Z.; Savalle, M.; Nemani, V.; Chaudhry, F.A.; Edwards, R.H.; et al. Alpha-synuclein overexpression in PC12 and chromaffin cells impairs catecholamine release by interfering with a late step in exocytosis. J. Neurosci. 2006, 26, 11915–11922. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Nemani, V.M.; Lu, W.; Berge, V.; Nakamura, K.; Onoa, B.; Lee, M.K.; Chaudhry, F.A.; Nicoll, R.A.; Edwards, R.H. Increased expression of alpha-synuclein reduces neurotransmitter release by inhibiting synaptic vesicle reclustering after endocytosis. Neuron 2010, 65, 66–79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Vargas, J.Y.; Grudina, C.; Zurzolo, C. The prion-like spreading of alpha-synuclein: From in vitro to in vivo models of Parkinson’s disease. Ageing Res. Rev. 2019, 50, 89–101. [Google Scholar] [CrossRef]
  71. Garcia-Reitbock, P.; Anichtchik, O.; Bellucci, A.; Iovino, M.; Ballini, C.; Fineberg, E.; Ghetti, B.; Della Corte, L.; Spano, P.; Tofaris, G.K.; et al. SNARE protein redistribution and synaptic failure in a transgenic mouse model of Parkinson’s disease. Brain 2010, 133, 2032–2044. [Google Scholar] [CrossRef] [PubMed]
  72. Burre, J.; Sharma, M.; Tsetsenis, T.; Buchman, V.; Etherton, M.R.; Sudhof, T.C. Alpha-synuclein promotes SNARE-complex assembly in vivo and in vitro. Science 2010, 329, 1663–1667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Choi, B.K.; Choi, M.G.; Kim, J.Y.; Yang, Y.; Lai, Y.; Kweon, D.H.; Lee, N.K.; Shin, Y.K. Large alpha-synuclein oligomers inhibit neuronal SNARE-mediated vesicle docking. Proc. Natl. Acad. Sci. USA 2013, 110, 4087–4092. [Google Scholar] [CrossRef] [Green Version]
  74. Gardoni, F.; Picconi, B.; Ghiglieri, V.; Polli, F.; Bagetta, V.; Bernardi, G.; Cattabeni, F.; Di Luca, M.; Calabresi, P. A critical interaction between NR2B and MAGUK in L-DOPA induced dyskinesia. J. Neurosci. 2006, 26, 2914–2922. [Google Scholar] [CrossRef] [Green Version]
  75. McGuinness, L.; Taylor, C.; Taylor, R.D.; Yau, C.; Langenhan, T.; Hart, M.L.; Christian, H.; Tynan, P.W.; Donnelly, P.; Emptage, N.J. Presynaptic NMDARs in the hippocampus facilitate transmitter release at theta frequency. Neuron 2010, 68, 1109–1127. [Google Scholar] [CrossRef] [Green Version]
  76. Thomas Broome, S.; Louangaphay, K.; Keay, K.A.; Leggio, G.M.; Musumeci, G.; Castorina, A. Dopamine: An immune transmitter. Neural Regen. Res. 2020, 15, 2173–2185. [Google Scholar]
  77. Yang, J.; Hertz, E.; Zhang, X.; Leinartaite, L.; Lundius, E.G.; Li, J.; Svenningsson, P. Overexpression of alpha-synuclein simultaneously increases glutamate NMDA receptor phosphorylation and reduces glucocerebrosidase activity. Neurosci. Lett. 2016, 611, 51–58. [Google Scholar] [CrossRef] [PubMed]
  78. Froula, J.M.; Henderson, B.W.; Gonzalez, J.C.; Vaden, J.H.; McLean, J.W.; Wu, Y.; Banumurthy, G.; Overstreet-Wadiche, L.; Herskowitz, J.H.; Volpicelli-Daley, L.A. Alpha-synuclein fibril-induced paradoxical structural and functional defects in hippocampal neurons. Acta Neuropathol. Commun. 2018, 6, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Bellucci, A.; Navarria, L.; Zaltieri, M.; Falarti, E.; Bodei, S.; Sigala, S.; Battistin, L.; Spillantini, M.; Missale, C.; Spano, P. Induction of the unfolded protein response by alpha-synuclein in experimental models of Parkinson’s disease. J. Neurochem. 2011, 116, 588–605. [Google Scholar] [CrossRef]
  80. Navarria, L.; Zaltieri, M.; Longhena, F.; Spillantini, M.G.; Missale, C.; Spano, P.; Bellucci, A. Alpha-synuclein modulates NR2B-containing NMDA receptors and decreases their levels after rotenone exposure. Neurochem. Int. 2015, 85–86, 14–23. [Google Scholar] [CrossRef]
  81. Mellone, M.; Stanic, J.; Hernandez, L.F.; Iglesias, E.; Zianni, E.; Longhi, A.; Prigent, A.; Picconi, B.; Calabresi, P.; Hirsch, E.C.; et al. NMDA receptor GluN2A/GluN2B subunit ratio as synaptic trait of levodopa-induced dyskinesias: From experimental models to patients. Front. Cell Neurosci. 2015, 9, 245. [Google Scholar] [CrossRef] [Green Version]
  82. Henrich, M.T.; Geibl, F.F.; Lakshminarasimhan, H.; Stegmann, A.; Giasson, B.I.; Mao, X.; Dawson, V.L.; Dawson, T.M.; Oertel, W.H.; Surmeier, D.J. Determinants of seeding and spreading of alpha-synuclein pathology in the brain. Sci. Adv. 2020, 6, eabc2487. [Google Scholar] [CrossRef]
  83. Mancini, A.; Ghiglieri, V.; Parnetti, L.; Calabresi, P.; Di Filippo, M. Neuro-immune cross-talk in the striatum: From basal ganglia physiology to circuit dysfunction. Front. Immunol. 2021, 12, 644294. [Google Scholar] [CrossRef]
  84. Trudler, D.; Sanz-Blasco, S.; Eisele, Y.S.; Ghatak, S.; Bodhinathan, K.; Akhtar, M.W.; Lynch, W.P.; Pina-Crespo, J.C.; Talantova, M.; Kelly, J.W.; et al. Alpha-synuclein oligomers induce glutamate release from astrocytes and excessive extrasynaptic NMDAR activity in neurons, thus contributing to synapse loss. J. Neurosci. 2021, 41, 2264–2273. [Google Scholar] [CrossRef] [PubMed]
  85. Bruck, D.; Wenning, G.K.; Stefanova, N.; Fellner, L. Glia and alpha-synuclein in neurodegeneration: A complex interaction. Neurobiol. Dis. 2016, 85, 262–274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Iwai, A.; Masliah, E.; Yoshimoto, M.; Ge, N.; Flanagan, L.; de Silva, H.A.; Kittel, A.; Saitoh, T. The precursor protein of non-A beta component of Alzheimer’s disease amyloid is a presynaptic protein of the central nervous system. Neuron 1995, 14, 467–475. [Google Scholar] [CrossRef] [Green Version]
  87. Norris, E.H.; Giasson, B.I.; Lee, V.M. Alpha-synuclein: Normal function and role in neurodegenerative diseases. Curr Top. Dev. Biol. 2004, 60, 17–54. [Google Scholar] [PubMed]
  88. Chartier-Harlin, M.C.; Kachergus, J.; Roumier, C.; Mouroux, V.; Douay, X.; Lincoln, S.; Levecque, C.; Larvor, L.; Andrieux, J.; Hulihan, M.; et al. Alpha-synuclein locus duplication as a cause of familial Parkinson’s disease. Lancet 2004, 364, 1167–1169. [Google Scholar] [CrossRef]
  89. Edwards, T.L.; Scott, W.K.; Almonte, C.; Burt, A.; Powell, E.H.; Beecham, G.W.; Wang, L.; Zuchner, S.; Konidari, I.; Wang, G.; et al. Genome-wide association study confirms SNPs in SNCA and the MAPT region as common risk factors for Parkinson disease. Ann. Hum. Genet. 2010, 74, 97–109. [Google Scholar] [CrossRef] [Green Version]
  90. Kruger, R.; Kuhn, W.; Muller, T.; Woitalla, D.; Graeber, M.; Kosel, S.; Przuntek, H.; Epplen, J.T.; Schols, L.; Riess, O. Ala30Pro mutation in the gene encoding alpha-synuclein in Parkinson’s disease. Nat. Genet. 1998, 18, 106–108. [Google Scholar] [CrossRef]
  91. Ross, O.A.; Braithwaite, A.T.; Skipper, L.M.; Kachergus, J.; Hulihan, M.M.; Middleton, F.A.; Nishioka, K.; Fuchs, J.; Gasser, T.; Maraganore, D.M.; et al. Genomic investigation of alpha-synuclein multiplication and parkinsonism. Ann. Neurol. 2008, 63, 743–750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Schonhoff, A.M.; Williams, G.P.; Wallen, Z.D.; Standaert, D.G.; Harms, A.S. Innate and adaptive immune responses in Parkinson’s disease. Prog. Brain Res. 2020, 252, 169–216. [Google Scholar] [PubMed]
  93. Singleton, A.B.; Farrer, M.; Johnson, J.; Singleton, A.; Hague, S.; Kachergus, J.; Hulihan, M.; Peuralinna, T.; Dutra, A.; Nussbaum, R.; et al. Alpha-synuclein locus triplication causes Parkinson’s disease. Science 2003, 302, 841. [Google Scholar] [CrossRef] [Green Version]
  94. Zarranz, J.J.; Alegre, J.; Gomez-Esteban, J.C.; Lezcano, E.; Ros, R.; Ampuero, I.; Vidal, L.; Hoenicka, J.; Rodriguez, O.; Atares, B.; et al. The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia. Ann. Neurol. 2004, 55, 164–173. [Google Scholar] [CrossRef] [PubMed]
  95. Winslow, A.R.; Chen, C.W.; Corrochano, S.; Acevedo-Arozena, A.; Gordon, D.E.; Peden, A.A.; Lichtenberg, M.; Menzies, F.M.; Ravikumar, B.; Imarisio, S.; et al. Alpha-synuclein impairs macroautophagy: Implications for Parkinson’s disease. J. Cell Biol. 2010, 190, 1023–1037. [Google Scholar] [CrossRef] [Green Version]
  96. Xilouri, M.; Stefanis, L. Autophagic pathways in Parkinson disease and related disorders. Expert Rev. Mol. Med. 2011, 13, e8. [Google Scholar] [CrossRef] [PubMed]
  97. Liu, C.Y.; Wang, X.; Liu, C.; Zhang, H.L. Pharmacological targeting of microglial activation: New therapeutic approach. Front. Cell Neurosci. 2019, 13, 514. [Google Scholar] [CrossRef] [Green Version]
  98. Theillet, F.X.; Binolfi, A.; Bekei, B.; Martorana, A.; Rose, H.M.; Stuiver, M.; Verzini, S.; Lorenz, D.; van Rossum, M.; Goldfarb, D.; et al. Structural disorder of monomeric alpha-synuclein persists in mammalian cells. Nature 2016, 530, 45–50. [Google Scholar] [CrossRef] [Green Version]
  99. Tanik, S.A.; Schultheiss, C.E.; Volpicelli-Daley, L.A.; Brunden, K.R.; Lee, V.M. Lewy body-like alpha-synuclein aggregates resist degradation and impair macroautophagy. J. Biol. Chem. 2013, 288, 15194–15210. [Google Scholar] [CrossRef] [Green Version]
  100. Giasson, B.I.; Duda, J.E.; Murray, I.V.; Chen, Q.; Souza, J.M.; Hurtig, H.I.; Ischiropoulos, H.; Trojanowski, J.Q.; Lee, V.M. Oxidative damage linked to neurodegeneration by selective alpha-synuclein nitration in synucleinopathy lesions. Science 2000, 290, 985–989. [Google Scholar] [CrossRef]
  101. Tofaris, G.K.; Razzaq, A.; Ghetti, B.; Lilley, K.S.; Spillantini, M.G. Ubiquitination of alpha-synuclein in Lewy bodies is a pathological event not associated with impairment of proteasome function. J. Biol. Chem. 2003, 278, 44405–44411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. El-Agnaf, O.M.; Salem, S.A.; Paleologou, K.E.; Cooper, L.J.; Fullwood, N.J.; Gibson, M.J.; Curran, M.D.; Court, J.A.; Mann, D.M.; Ikeda, S.; et al. Alpha-synuclein implicated in Parkinson’s disease is present in extracellular biological fluids, including human plasma. FASEB J. 2003, 17, 1945–1947. [Google Scholar] [CrossRef] [PubMed]
  103. Emmanouilidou, E.; Melachroinou, K.; Roumeliotis, T.; Garbis, S.D.; Ntzouni, M.; Margaritis, L.H.; Stefanis, L.; Vekrellis, K. Cell-produced alpha-synuclein is secreted in a calcium-dependent manner by exosomes and impacts neuronal survival. J. Neurosci. 2010, 30, 6838–6851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Jang, A.; Lee, H.J.; Suk, J.E.; Jung, J.W.; Kim, K.P.; Lee, S.J. Non-classical exocytosis of alpha-synuclein is sensitive to folding states and promoted under stress conditions. J. Neurochem. 2010, 113, 1263–1274. [Google Scholar] [PubMed]
  105. Lee, H.J.; Bae, E.J.; Lee, S.J. Extracellular alpha-synuclein—A novel and crucial factor in Lewy body diseases. Nat. Rev. Neurol. 2014, 10, 92–98. [Google Scholar] [CrossRef] [PubMed]
  106. Lee, S.J.; Lim, H.S.; Masliah, E.; Lee, H.J. Protein aggregate spreading in neurodegenerative diseases: Problems and perspectives. Neurosci. Res. 2011, 70, 339–348. [Google Scholar] [CrossRef] [Green Version]
  107. Pacheco, C.; Aguayo, L.G.; Opazo, C. An extracellular mechanism that can explain the neurotoxic effects of alpha-synuclein aggregates in the brain. Front. Physiol. 2012, 3, 297. [Google Scholar] [CrossRef] [Green Version]
  108. Schirinzi, T.; Madeo, G.; Martella, G.; Maltese, M.; Picconi, B.; Calabresi, P.; Pisani, A. Early synaptic dysfunction in Parkinson’s disease: Insights from animal models. Mov. Disord. 2016, 31, 802–813. [Google Scholar] [CrossRef]
  109. Kim, C.; Ho, D.H.; Suk, J.E.; You, S.; Michael, S.; Kang, J.; Joong Lee, S.; Masliah, E.; Hwang, D.; Lee, H.J.; et al. Neuron-released oligomeric alpha-synuclein is an endogenous agonist of TLR2 for paracrine activation of microglia. Nat. Commun. 2013, 4, 1562. [Google Scholar] [CrossRef] [Green Version]
  110. Chao, Y.; Wong, S.C.; Tan, E.K. Evidence of inflammatory system involvement in Parkinson’s disease. BioMed Res. Int. 2014, 2014, 308654. [Google Scholar] [CrossRef]
  111. Pajares, M.; Rojo, A.I.; Manda, G.; Bosca, L.; Cuadrado, A. Inflammation in Parkinson’s disease: Mechanisms and therapeutic implications. Cells 2020, 9, 1687. [Google Scholar] [CrossRef]
  112. Varanita, T.; Bubacco, L. Alpha-synuclein and neuroinflammation in Parkinson’s disease. In Genetics, Neurology, Behavior, and Diet in Parkinson’s Disease: The Neuroscience of Parkinson’s Disease; Martin, C.R., Preedy, V.R., Eds.; Academic Press: Cambridge, MA, USA, 2020; Volume 2. [Google Scholar]
  113. Davalos, D.; Grutzendler, J.; Yang, G.; Kim, J.V.; Zuo, Y.; Jung, S.; Littman, D.R.; Dustin, M.L.; Gan, W.B. ATP mediates rapid microglial response to local brain injury in vivo. Nat. Neurosci. 2005, 8, 752–758. [Google Scholar] [CrossRef] [PubMed]
  114. Nimmerjahn, A.; Kirchhoff, F.; Helmchen, F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science 2005, 308, 1314–1318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Alvarez-Erviti, L.; Couch, Y.; Richardson, J.; Cooper, J.M.; Wood, M.J. Alpha-synuclein release by neurons activates the inflammatory response in a microglial cell line. Neurosci. Res. 2011, 69, 337–342. [Google Scholar] [CrossRef] [PubMed]
  116. Chesselet, M.F.; Richter, F.; Zhu, C.; Magen, I.; Watson, M.B.; Subramaniam, S.R. A progressive mouse model of Parkinson’s disease: The Thy1-aSyn (“Line 61”) mice. Neurotherapeutics 2012, 9, 297–314. [Google Scholar] [CrossRef] [Green Version]
  117. George, S.; Rey, N.L.; Tyson, T.; Esquibel, C.; Meyerdirk, L.; Schulz, E.; Pierce, S.; Burmeister, A.R.; Madaj, Z.; Steiner, J.A.; et al. Microglia affect alpha-synuclein cell-to-cell transfer in a mouse model of Parkinson’s disease. Mol. Neurodegener. 2019, 14, 34. [Google Scholar] [CrossRef]
  118. Hansen, C.; Angot, E.; Bergstrom, A.L.; Steiner, J.A.; Pieri, L.; Paul, G.; Outeiro, T.F.; Melki, R.; Kallunki, P.; Fog, K.; et al. Alpha-synuclein propagates from mouse brain to grafted dopaminergic neurons and seeds aggregation in cultured human cells. J. Clin. Investig. 2011, 121, 715–725. [Google Scholar] [CrossRef] [Green Version]
  119. Olanow, C.W.; Savolainen, M.; Chu, Y.; Halliday, G.M.; Kordower, J.H. Temporal evolution of microglia and alpha-synuclein accumulation following foetal grafting in Parkinson’s disease. Brain 2019, 142, 1690–1700. [Google Scholar] [CrossRef]
  120. Wang, W.; Nguyen, L.T.; Burlak, C.; Chegini, F.; Guo, F.; Chataway, T.; Ju, S.; Fisher, O.S.; Miller, D.W.; Datta, D.; et al. Caspase-1 causes truncation and aggregation of the Parkinson’s disease-associated protein alpha-synuclein. Proc. Natl. Acad. Sci. USA 2016, 113, 9587–9592. [Google Scholar] [CrossRef] [Green Version]
  121. Tremblay, M.E.; Cookson, M.R.; Civiero, L. Glial phagocytic clearance in Parkinson’s disease. Mol. Neurodegener. 2019, 14, 16. [Google Scholar] [CrossRef] [Green Version]
  122. Wahis, J.; Hennes, M.; Arckens, L.; Holt, M.G. Star power: The emerging role of astrocytes as neuronal partners during cortical plasticity. Curr. Opin. Neurobiol. 2020, 67, 174–182. [Google Scholar] [CrossRef]
  123. Andoh, M.; Koyama, R. Microglia regulate synaptic development and plasticity. Dev. Neurobiol. 2021. [Google Scholar] [CrossRef]
  124. Perea, G.; Navarrete, M.; Araque, A. Tripartite synapses: Astrocytes process and control synaptic information. Trends Neurosci. 2009, 32, 421–431. [Google Scholar] [CrossRef]
  125. Braak, H.; Sastre, M.; Del Tredici, K. Development of alpha-synuclein immunoreactive astrocytes in the forebrain parallels stages of intraneuronal pathology in sporadic Parkinson’s disease. Acta Neuropathol. 2007, 114, 231–241. [Google Scholar] [CrossRef]
  126. Chavarria, C.; Rodriguez-Bottero, S.; Quijano, C.; Cassina, P.; Souza, J.M. Impact of monomeric, oligomeric and fibrillar alpha-synuclein on astrocyte reactivity and toxicity to neurons. Biochem. J. 2018, 475, 3153–3169. [Google Scholar] [CrossRef] [PubMed]
  127. Damier, P.; Hirsch, E.C.; Zhang, P.; Agid, Y.; Javoy-Agid, F. Glutathione peroxidase, glial cells and Parkinson’s disease. Neuroscience 1993, 52, 1–6. [Google Scholar] [CrossRef]
  128. Garcia-Esparcia, P.; Llorens, F.; Carmona, M.; Ferrer, I. Complex deregulation and expression of cytokines and mediators of the immune response in Parkinson’s disease brain is region dependent. Brain Pathol. 2014, 24, 584–598. [Google Scholar] [CrossRef] [PubMed]
  129. Gu, X.L.; Long, C.X.; Sun, L.; Xie, C.; Lin, X.; Cai, H. Astrocytic expression of Parkinson’s disease-related A53T alpha-synuclein causes neurodegeneration in mice. Mol. Brain 2010, 3, 12. [Google Scholar] [CrossRef] [Green Version]
  130. Mythri, R.B.; Venkateshappa, C.; Harish, G.; Mahadevan, A.; Muthane, U.B.; Yasha, T.C.; Srinivas Bharath, M.M.; Shankar, S.K. Evaluation of markers of oxidative stress, antioxidant function and astrocytic proliferation in the striatum and frontal cortex of Parkinson’s disease brains. Neurochem. Res. 2011, 36, 1452–1463. [Google Scholar] [CrossRef]
  131. Miyazaki, I.; Asanuma, M. Neuron-astrocyte interactions in Parkinson’s disease. Cells 2020, 9, 2623. [Google Scholar] [CrossRef]
  132. Knott, C.; Wilkin, G.P.; Stern, G. Astrocytes and microglia in the substantia nigra and caudate-putamen in Parkinson’s disease. Parkinsonism Relat. Disord. 1999, 5, 115–122. [Google Scholar] [CrossRef]
  133. McGeer, P.L.; McGeer, E.G. Glial reactions in Parkinson’s disease. Mov. Disord. 2008, 23, 474–483. [Google Scholar] [CrossRef] [PubMed]
  134. Mirza, B.; Hadberg, H.; Thomsen, P.; Moos, T. The absence of reactive astrocytosis is indicative of a unique inflammatory process in Parkinson’s disease. Neuroscience 2000, 95, 425–432. [Google Scholar] [CrossRef]
  135. Tong, J.; Ang, L.C.; Williams, B.; Furukawa, Y.; Fitzmaurice, P.; Guttman, M.; Boileau, I.; Hornykiewicz, O.; Kish, S.J. Low levels of astroglial markers in Parkinson’s disease: Relationship to alpha-synuclein accumulation. Neurobiol. Dis. 2015, 82, 243–253. [Google Scholar] [CrossRef] [Green Version]
  136. Morales, I.; Sanchez, A.; Rodriguez-Sabate, C.; Rodriguez, M. The astrocytic response to the dopaminergic denervation of the striatum. J. Neurochem. 2016, 139, 81–95. [Google Scholar] [CrossRef]
  137. Zhu, Y.F.; Wang, W.P.; Zheng, X.F.; Chen, Z.; Chen, T.; Huang, Z.Y.; Jia, L.J.; Lei, W.L. Characteristic response of striatal astrocytes to dopamine depletion. Neural Regen. Res. 2020, 15, 724–730. [Google Scholar] [PubMed]
  138. Liddelow, S.A.; Barres, B.A. Reactive astrocytes: Production, function, and therapeutic potential. Immunity 2017, 46, 957–967. [Google Scholar] [CrossRef] [Green Version]
  139. Lindstrom, V.; Gustafsson, G.; Sanders, L.H.; Howlett, E.H.; Sigvardson, J.; Kasrayan, A.; Ingelsson, M.; Bergstrom, J.; Erlandsson, A. Extensive uptake of alpha-synuclein oligomers in astrocytes results in sustained intracellular deposits and mitochondrial damage. Mol. Cell Neurosci. 2017, 82, 143–156. [Google Scholar] [CrossRef]
  140. Rostami, J.; Holmqvist, S.; Lindstrom, V.; Sigvardson, J.; Westermark, G.T.; Ingelsson, M.; Bergstrom, J.; Roybon, L.; Erlandsson, A. Human astrocytes transfer aggregated alpha-synuclein via tunneling nanotubes. J. Neurosci. 2017, 37, 11835–11853. [Google Scholar] [CrossRef] [Green Version]
  141. Iovino, M.; Messana, T.; De Pergola, G.; Iovino, E.; Guastamacchia, E.; Licchelli, B.; Vanacore, A.; Giagulli, V.A.; Triggiani, V. Brain angiotensinergic regulation of the immune system: Implications for cardiovascular and neuroendocrine responses. Endocr. Metab. Immune Disord. Drug Targets 2020, 20, 15–24. [Google Scholar] [CrossRef]
  142. Takaki, J.; Fujimori, K.; Miura, M.; Suzuki, T.; Sekino, Y.; Sato, K. L-glutamate released from activated microglia downregulates astrocytic L-glutamate transporter expression in neuroinflammation: The ‘collusion’ hypothesis for increased extracellular L-glutamate concentration in neuroinflammation. J. Neuroinflamm. 2012, 9, 275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Zou, J.Y.; Crews, F.T. TNF alpha potentiates glutamate neurotoxicity by inhibiting glutamate uptake in organotypic brain slice cultures: Neuroprotection by NF kappa B inhibition. Brain Res. 2005, 1034, 11–24. [Google Scholar] [CrossRef] [PubMed]
  144. De Vries, H.E.; Kooij, G.; Frenkel, D.; Georgopoulos, S.; Monsonego, A.; Janigro, D. Inflammatory events at blood-brain barrier in neuroinflammatory and neurodegenerative disorders: Implications for clinical disease. Epilepsia 2012, 53, 45–52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Patel, J.P.; Frey, B.N. Disruption in the blood-brain barrier: The missing link between brain and body inflammation in bipolar disorder? Neural Plast. 2015, 2015, 708306. [Google Scholar] [CrossRef]
  146. McGeer, P.L.; Kawamata, T.; Walker, D.G.; Akiyama, H.; Tooyama, I.; McGeer, E.G. Microglia in degenerative neurological disease. Glia 1993, 7, 84–92. [Google Scholar] [CrossRef]
  147. Harms, A.S.; Thome, A.D.; Yan, Z.; Schonhoff, A.M.; Williams, G.P.; Li, X.; Liu, Y.; Qin, H.; Benveniste, E.N.; Standaert, D.G. Peripheral monocyte entry is required for alpha-Synuclein induced inflammation and Neurodegeneration in a model of Parkinson disease. Exp. Neurol. 2018, 300, 179–187. [Google Scholar] [CrossRef] [PubMed]
  148. Kortekaas, R.; Leenders, K.L.; van Oostrom, J.C.; Vaalburg, W.; Bart, J.; Willemsen, A.T.; Hendrikse, N.H. Blood-brain barrier dysfunction in parkinsonian midbrain in vivo. Ann. Neurol. 2005, 57, 176–179. [Google Scholar] [CrossRef]
  149. Sanchez-Guajardo, V.; Barnum, C.J.; Tansey, M.G.; Romero-Ramos, M. Neuroimmunological processes in Parkinson’s disease and their relation to alpha-synuclein: Microglia as the referee between neuronal processes and peripheral immunity. ASN Neuro 2013, 5, 113–139. [Google Scholar] [CrossRef] [Green Version]
  150. Gonzalez, H.; Contreras, F.; Pacheco, R. Regulation of the neurodegenerative process associated to Parkinson’s disease by CD4 + T-cells. J. Neuroimmune Pharm. 2015, 10, 561–575. [Google Scholar] [CrossRef]
  151. Di Filippo, M.; Sarchielli, P.; Picconi, B.; Calabresi, P. Neuroinflammation and synaptic plasticity: Theoretical basis for a novel, immune-centred, therapeutic approach to neurological disorders. Trends Pharm. Sci. 2008, 29, 402–412. [Google Scholar] [CrossRef]
  152. Cosentino, M.; Kustrimovic, N.; Ferrari, M.; Rasini, E.; Marino, F. cAMP levels in lymphocytes and CD4(+) regulatory T-cell functions are affected by dopamine receptor gene polymorphisms. Immunology 2018, 153, 337–341. [Google Scholar] [CrossRef]
  153. Duffy, M.F.; Collier, T.J.; Patterson, J.R.; Kemp, C.J.; Luk, K.C.; Tansey, M.G.; Paumier, K.L.; Kanaan, N.M.; Fischer, D.L.; Polinski, N.K.; et al. Lewy body-like alpha-synuclein inclusions trigger reactive microgliosis prior to nigral degeneration. J. Neuroinflamm. 2018, 15, 129. [Google Scholar] [CrossRef]
  154. Chen, H.; Jacobs, E.; Schwarzschild, M.A.; McCullough, M.L.; Calle, E.E.; Thun, M.J.; Ascherio, A. Nonsteroidal antiinflammatory drug use and the risk for Parkinson’s disease. Ann. Neurol. 2005, 58, 963–967. [Google Scholar] [CrossRef]
  155. Bornebroek, M.; de Lau, L.M.; Haag, M.D.; Koudstaal, P.J.; Hofman, A.; Stricker, B.H.; Breteler, M.M. Nonsteroidal anti-inflammatory drugs and the risk of Parkinson disease. Neuroepidemiology 2007, 28, 193–196. [Google Scholar] [CrossRef]
  156. Manthripragada, A.D.; Schernhammer, E.S.; Qiu, J.; Friis, S.; Wermuth, L.; Olsen, J.H.; Ritz, B. Non-steroidal anti-inflammatory drug use and the risk of Parkinson’s disease. Neuroepidemiology 2011, 36, 155–161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Gagne, J.J.; Power, M.C. Anti-inflammatory drugs and risk of Parkinson disease: A meta-analysis. Neurology 2010, 74, 995–1002. [Google Scholar] [CrossRef] [PubMed]
  158. Hernan, M.A.; Logroscino, G.; Garcia Rodriguez, L.A. Nonsteroidal anti-inflammatory drugs and the incidence of Parkinson disease. Neurology 2006, 66, 1097–1099. [Google Scholar] [CrossRef] [PubMed]
  159. Wahner, A.D.; Bronstein, J.M.; Bordelon, Y.M.; Ritz, B. Nonsteroidal anti-inflammatory drugs may protect against Parkinson disease. Neurology 2007, 69, 1836–1842. [Google Scholar] [CrossRef]
  160. Sardi, S.P.; Cedarbaum, J.M.; Brundin, P. Targeted therapies for Parkinson’s disease: From genetics to the clinic. Mov. Disord. 2018, 33, 684–696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Zella, M.A.S.; Metzdorf, J.; Ostendorf, F.; Maass, F.; Muhlack, S.; Gold, R.; Haghikia, A.; Tonges, L. Novel immunotherapeutic approaches to target alpha-synuclein and related neuroinflammation in Parkinson’s disease. Cells 2019, 8, 105. [Google Scholar] [CrossRef] [Green Version]
  162. Antonini, A.; Bravi, D.; Sandre, M.; Bubacco, L. Immunization therapies for Parkinson’s disease: State of the art and considerations for future clinical trials. Expert Opin. Investig. Drugs 2020, 29, 685–695. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The presynaptic exo-endocytotic cycle is specifically affected in α-syn-related synaptopaties. α-syn accumulation in the presynaptic terminals causes synaptopathy, ultimately leading to neurodegeneration. Under physiological conditions, α-syn, as a monomer, acts at the presynaptic terminals and activates a molecular machinery relevant to synaptic transmission, including the SNARE complex proteins, which recruit synaptic vesicles. After docking and priming, the vesicles undergo SNARE-mediated membrane fusion at the active zone, leading to neurotransmitter release into the synaptic cleft. Formation of toxic α-syn species in Parkinson’s disease (PD), such as oligomers and fibrils, has been shown to play a pivotal role in PD pathogenesis. These toxic species, which accumulate at the presynaptic terminal, lead to altered levels of proteins involved in synaptic transmission, determining synaptic dysfunction. Toxic α-syn accumulation affects the synapses, causing a lack of long-term synaptic plasticity expression, followed by an increase in the phosphorylation of glutamate receptors and a decrease in the GluN2A/GluN2B receptor subunit ratio, contributing to neuronal degeneration. Toxic aggregated α-syn forms participate in inflammatory processes mediated by microglia and astrocytes, which lose their normal physiological functions. Microglia become activated (M1) and trigger an immune response through increased receptor expression (MHCI), secretion of pro-inflammatory cytokines and chemokines, and inducing an astrocyte reaction (A1 type). Reactive astrocytes decrease glutamate uptake and release pro-inflammatory mediators. Finally, peripheral immune cells (T cells and monocytes) raid brain tissue and amplify neuroinflammation, contributing to neurodegeneration.
Figure 1. The presynaptic exo-endocytotic cycle is specifically affected in α-syn-related synaptopaties. α-syn accumulation in the presynaptic terminals causes synaptopathy, ultimately leading to neurodegeneration. Under physiological conditions, α-syn, as a monomer, acts at the presynaptic terminals and activates a molecular machinery relevant to synaptic transmission, including the SNARE complex proteins, which recruit synaptic vesicles. After docking and priming, the vesicles undergo SNARE-mediated membrane fusion at the active zone, leading to neurotransmitter release into the synaptic cleft. Formation of toxic α-syn species in Parkinson’s disease (PD), such as oligomers and fibrils, has been shown to play a pivotal role in PD pathogenesis. These toxic species, which accumulate at the presynaptic terminal, lead to altered levels of proteins involved in synaptic transmission, determining synaptic dysfunction. Toxic α-syn accumulation affects the synapses, causing a lack of long-term synaptic plasticity expression, followed by an increase in the phosphorylation of glutamate receptors and a decrease in the GluN2A/GluN2B receptor subunit ratio, contributing to neuronal degeneration. Toxic aggregated α-syn forms participate in inflammatory processes mediated by microglia and astrocytes, which lose their normal physiological functions. Microglia become activated (M1) and trigger an immune response through increased receptor expression (MHCI), secretion of pro-inflammatory cytokines and chemokines, and inducing an astrocyte reaction (A1 type). Reactive astrocytes decrease glutamate uptake and release pro-inflammatory mediators. Finally, peripheral immune cells (T cells and monocytes) raid brain tissue and amplify neuroinflammation, contributing to neurodegeneration.
Ijms 22 06517 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cardinale, A.; Calabrese, V.; de Iure, A.; Picconi, B. Alpha-Synuclein as a Prominent Actor in the Inflammatory Synaptopathy of Parkinson’s Disease. Int. J. Mol. Sci. 2021, 22, 6517. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22126517

AMA Style

Cardinale A, Calabrese V, de Iure A, Picconi B. Alpha-Synuclein as a Prominent Actor in the Inflammatory Synaptopathy of Parkinson’s Disease. International Journal of Molecular Sciences. 2021; 22(12):6517. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22126517

Chicago/Turabian Style

Cardinale, Antonella, Valeria Calabrese, Antonio de Iure, and Barbara Picconi. 2021. "Alpha-Synuclein as a Prominent Actor in the Inflammatory Synaptopathy of Parkinson’s Disease" International Journal of Molecular Sciences 22, no. 12: 6517. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22126517

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