Alpha Synuclein Toxicity and Non-Motor Parkinson’s. (2024)

Link/Page Citation

Author(s): Gabriella M. Mazzotta (corresponding author) [1,*]; Carmela Conte (corresponding author) [2,*]

1. Introduction

Parkinson’s disease (PD) is one of the most common neurodegenerative disorders, second only to Alzheimer’s disease, and refers to a-synucleinopathies characterized by loss of the dopaminergic and monoaminergic neurons in substantia nigra pars compacta (SNpc) and ventral tegmental area, as well as by the accumulation, aggregation and spread of a-Synuclein (aSyn) within neurons and non-neuronal cells including microglia, pericytes, astrocytes, and oligodendrocytes [1]. PD is the most common form of movement disorder clinically manifested by bradykinesia, resting tremor, rigidity, slowness of movement, freezing, dizziness, and postural instability as a result of the loss of 70–80% of the dopaminergic neurons in the SNpc [2]. Aside from motor manifestations, PD impacts a plethora of non-motor symptoms (NMS) which may predate the onset of movement disorders by several years [3,4]. They include alterations in the sense of smell [5,6], dysfunction of circadian rhythms [7], and autonomic disorders such as orthostatic hypotension constipation, urinary difficulties, gastrointestinal disorders, anxiety, depression, and impairment or loss of sensory perception [4,8,9]. Non-motor manifestations have been recognized as significant contributors to the overall impact of the disease on the individual’s quality of life [10,11,12].

According to disease stagings by Braak, in stage 1, early NMS such as smell disorders experienced by PD patients are caused by neuronal degeneration in the olfactory bulb and the anterior olfactory nucleus, while degeneration in Braak stage 2 involves the lower brainstem i.e., raphe nucleus, locus coeruleus (LC), pedunculopontine nucleus, and the thalamocortical system, which are associated with sleep disturbances, autonomic dysfunctions, visual hallucinations, and rapid eye movements [13].

Dysfunction of the medullary nuclei located in the brainstem, namely nucleus tractus solitarius, dorsal motor nucleus of the vagus, and nucleus ambigus, regulate various involuntary functions including arterial blood pressure, heart rate, respiratory activity, and renal function. These conditions are associated with orthostatic hypotension, cardiovascular abnormalities, and other autonomic disorders in PD [14]. The emergence of the classic motor symptoms of PD typically occurs at Braak stages 3 and 4. These clinical signs coincide with the involvement of key regions such as the SNpc and other deep nuclei of the midbrain and the forebrain [15]. In the final stages Braak 5 and 6, Lewy bodies are found in the limbic structures and mature neocortex. At these stages, patients with PD experience a variety of neuropsychiatric symptoms, including depression, cognitive impairment, and visual hallucinations [15]. The subthalamic nucleus plays a crucial role in PD. It receives inputs from sensorimotor, associative, and limbic brain regions which serve distinct functions within the basal ganglia circuitry. Specifically, pallidosubthalamic projections are thought to underlie cognitive, emotional, and motivational NMS. This intricate organization underscores the diverse roles of the subthalamic nucleus in both motor and non-motor aspects of PD.

Several studies have reported the positive effect of deep brain stimulation (STN-DBS) on motor and NMS, although PD patients with longer disease duration had limited motor benefits from STN-DBS [16,17,18,19]. Case reports and data from a meta-analysis conducted on dozens of studies indicate that STN-DBS improves NMS and PD patients’ quality of life [20,21,22,23]. Due to the complexity of PD, a more comprehensive understanding of the underlying mechanisms beyond the scope of the Braak staging model is required. Certainly, the recognition of non-motor signs that include other issues such as cognitive and memory impairment known as pre-motor symptoms, can contribute to early diagnosis of PD and help adopt strategies aimed at improving the management of the disease [24,25]. In this review, we highlight recent advancements in the field of aSyn neurotoxicity that have contributed to our understanding of NMS onset that could aid in the development of future treatment strategies and/or early diagnosis.

2. aSyn Toxicity

From a pathological point of view, similar to motor symptoms, NMS can occur as a consequence of the formation of intracytoplasmic Lewy bodies and neurites rich in aSyn aggregates in nigral and extranigral areas.

aSyn is a small, 140 amino acid presynaptic acidic protein encoded by the SNCA gene whose main function appears to be the control of neurotransmitter release [26]. It is a vesicle-bound multimer consisting of three distinct regions: (1) an N-terminus (residues 1–60) region that is crucial for its interaction with lipid membranes [27,28]; (2) a central hydrophobic region (residues 61–95) that is prone to aggregation and named the non-amyloid-ß component (NAC region) [29]; (3) an unstructured C-terminus (residues 96–140) that is highly negatively charged with Ca[sup.2+] binding and chaperone-like activity [30].

In the CNS, aSyn exists in a soluble cytosolic fraction (for as much as 1% of the total protein) and two membrane- and vesicle-binding forms: via the C-terminal domain it interacts with the vesicle-associated membrane protein 2 (VAMP2) leading to the formation of vesicle clusters [31] and affecting vesicle docking as well as the inhibition of vesicle fusion, while via the N-terminal region aSyn was found to bind membrane lipids. This membrane-binding pool appears to prevent pathological aggregation of aSyn [32,33,34]. Mutations in this domain are associated with PD pathology [35,36]. On the contrary, the cytosolic fraction is intrinsically disordered and behaves like a natively unfolded protein, contributing to the formation of aggregate species [37]. aSyn aggregates have been detected in various peripheral biopsies such as the gastrointestinal tract, skin, and salivary glands, even in the early stages of PD [38] supporting the hypothesis that PD might start in the peripheral nervous system before affecting the brain. For instance, aSyn pathology has been found in the enteric nervous system (ENS), leading to the theory that the disease may originate in the gut and then spread to the brain via the vagus nerve [39].

Different factors, such as genetic mutations [40], elevated levels of aSyn, mitochondrial dysfunction [41], oxidative stress (OS) [42], endoplasmic reticulum (ER) stress [43] dysregulation of synaptic vesicle recycling [44], and the autophagy-lysosomal system [45], are well known to contribute to aSyn misfolding, forming ß-sheet rich oligomers and fibrils in Lewy bodies or Lewy neurites accompanying NMS [46,47,48,49]. Studies also support the contribution of neuroinflammation in NMS [50,51,52], pointing to the essential components of the innate immune response such as the toll-like receptors (TLRs) [53,54,55,56]. In particular, pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) can prime and activate the TLRs creating a neuroinflammatory status that may culminate with neuronal death in specific brain areas [57]. Human studies suggest a role for TLR2 and TLR4 in the recognition of toxic species of aSyn as a DAMP that precedes aSyn aggregation [58,59]. Furthermore, polymorphisms in the SNCA gene are also well-recognized factors contributing to the development of non-motor signs [60]. NMS also result from the diffusion of the pathological species of aSyn. In fact, aSyn aggregates spread between interconnected brain areas in a cell-to-cell and prion-like manner which involves neurons and non-neuronal cells [61,62]. Transsynaptic transmission of aSyn may be triggered by oligomeric aSyn-mediated microglial activation in the early phases of the disease [63] or by interactions between endogenous aSyn and mitochondria [64,65]. However, cells differentially mediate the uptake of aSyn fibrils involving processes such as receptor-mediated endocytosis, extracellular vesicles, and tunneling nanotubes. Fibrils can be sequestered and degraded into lysosomes or be trafficked into the cytosol via endocytic pathways, where they can interact with and recruit monomeric aSyns into mature pathological inclusions [66]. In addition, fibrils can be released by damaged or dead cells following mitochondrial dysfunction [41,54,67], disruption of redox balance [42], nitrosative stress [68], impaired autophagic flux [45], and prolonged ER stress [43]. Oligomeric aSyn may alter the voltage-gated receptors resulting in impairment of calcium efflux [69,70].

A schematic representation of the relation between toxic aSyn species and cellular dysfunctions is illustrated in Figure 1.

2.1. Oxidative/Nitrosative Stress

A bidirectional relationship between oxidative/nitrosative stress and aSyn toxicity exists [71]. High levels of reactive oxidative species (ROS) within the neurons have been suggested to induce aSyn toxicity [72,73,74]. Excessive OS causes peroxidation of the membrane lipids followed by the production of the highly toxic 4-hydroxy-2-nonenal (4-HNE). This compound induces the formation of ß-sheets and toxic soluble oligomers of aSyn which are believed to promote neuronal damage [75,76]. Hydroxyl radical species can react with tyrosine residues on aSyn contributing to the formation of intramolecular oxidative covalent cross-linkage between two tyrosine residues known as the dityrosine bond [77]. Two opposing effects have been described for dityrosine: (i) formation and stabilization of aSyn insoluble fibrils or aggregates [78], and (ii) inhibition of aSyn fibrillation by dityrosine-modified monomers and dimers [77]. As aggregation proceeds, dityrosine formation shifts from an aggregation-inhibiting to an aggregation-promoting element [79]. The higher-order assembly as well as the increased stability of growing fibrils appears to significantly reduce their capacity to seed further due to the reduced availability of nucleation sites and the conversion to less dynamic structures. Again, changes in hydrophobicity or charge distribution aside from changing the equilibrium between monomers, oligomers, and fibrils could influence the incorporation of additional monomers and aggregation kinetics [80].

Hypochlorite-oxidized cysteinyl-dopamine is considered a potent redox cycler capable of accelerating OS and contributes to increased autophagy and ultimately to cell death [81]. Additionally, post-translationally aberrant S-nitrosylation caused by reactive nitrogen species and nitric oxide in neurons is known to induce aSyn misfolding and toxicity, and axo-dendritic and dopamine dysfunction [82]. Moreover, Kumar et al. [83] showed that S-nitrosylation of the ubiquitin C-terminal hydrolase-1 induces nucleation with the native aSyn, accelerating the protein aggregation.

2.2. Mitochondrial Dysfunction

Mitochondrial dysfunction plays a central role in the pathogenesis of PD [84]. Several neurotoxins have been found to provoke high levels of ROS, complex I inhibition, and neuronal damage [73]. Additionally, abnormal levels or misfolded forms of aSyn can disrupt the balance of mitochondrial fission, fusion and transport, leading to fragmentated or aggregated mitochondria, and impairment of mitochondrial trafficking, electron transport chain, and calcium signaling [84,85]. aSyn can interact with complex I, resulting in decreased ATP production and increased ROS [86]. The selective autophagic degradation of damaged mitochondria can overwhelm cellular proteostasis, leading to the accumulation of misfolded aSyn and mitochondrial dysfunction [87,88,89]. aSyn can trigger the opening of mitochondrial permeability transition pores, causing mitochondrial swelling and loss of membrane potential [90], and can interact with other proteins involved in mitochondrial quality control, such as Parkin and PINK [91]. Dysregulation of these interactions can impair mitochondrial function and dynamics, contributing to the pathogenesis of PD.

2.3. ER Stress

The accumulation of newly synthesized or improperly folded proteins in the ER can saturate the folding machinery, leading to ER stress which promptly activates the unfolded protein response (UPR) to restore normal ER function. In response to prolonged stress, UPR loses the capacity to assist in protein folding and its dysregulation culminates with neuronal death as a protective measure to prevent further damage [43]. In this condition, the activation of C/EBP hom*ologous Protein by Protein Kinase R-like ER Kinase, Activating Transcription Factor 4 (ATF4), ATF6, or X-box Binding Protein 1 appears to be central for the induction of ER stress-driven apoptotic signaling, ensuring that cells with irreparable damage are eliminated to maintain tissue homeostasis. ER-phagy, a selective form of autophagy targeting the ER for degradation, plays a crucial role in the clearance of misfolded aSyn. The interaction with ER-resident autophagy receptor FAM134B and the involvement of calnexin are key components for the recruitment of unfolded aSyn, encapsulation of ER fragments into autophagosomes, transport to and fusion with lysosomes, and degradation and clearance of misfolded synuclein [92].

2.4. Neuroinflammation

A PubMed search for “inflammation and non-motor symptoms of PD” yielded 302 results from the past 10 years, indicating that the issue is of great interest and importance. Although some debate still ensues, several studies support the crucial role of the immune/inflammatory response in the pathogenesis of NMS in PD. Once triggered, inflammatory load commences a series of toxic events that correlate with a number of NMS including depression, cognitive impairment, psychosis, sleep disturbance, GI dysfunction, and others. The major result is a massive release by microglia, the CNS-resident immune cell of several pro-inflammatory cytokines including IL-1, IL6, TNFa, and IL-17. This may be the result of the activation of a variety of signaling pathways or represent the trigger for NMS, or both.

The switching of microglia from the M1 to M2 phenotype can be facilitated by endogenous or extracellular aSyn. For example, iPSC-derived macrophages from patients with SNCA triplication display accumulation of aSyn, impairment of phagocytosis, and reduced ability of dopaminergic and GABAergic differentiation [93,94]. On the other hand, extracellular aSyn released by damaged neurons can propagate to neighboring microglial cells and activate microglia via the TLRs-NLRP3 or NF-kB-NLRP3 axes to produce further inflammatory mediators, including caspase-1-mediated IL-1ß. aSyn can act as an NLRP3 inflammasome inductor leading to the processing and secretion of mature IL-1ß contributing to Lewy body formation in brain tissue of PD patients and in other peripheral tissues. aSyn can stimulate endogenous protein aggregation leading to the spread of aSyn pathology and to the development of NMS and motor symptoms [95].

Another important factor associated with the NLRP3 inflammasome is the aSyn-mediated upregulation of the Kv1.3 channel in microglia and the consequent increase in K[sup.+] efflux. This process could affect dopamine release by neurons, which in turn could inhibit NLRP3 inflammasome formation, constricting the microglia to utilize DA from neighboring neurons to prevent its own activation and thereby avoid the establishment of an inflammatory state that would lead to further aSyn aggregation [96,97].

It has been found that the activity of indoleamine-2,3-dioxygenase (IDO), the rate-limiting enzymes of tryptophan metabolism, is induced by high levels of pro-inflammatory cytokines which may lead to decreased serotonin production, depression, and Schizophrenia [98,99]. The treatment of the 6-hydroxydopamine mouse model of PD with IDO inhibitors rescued oxidative stress, neuroinflammation, apoptosis, and dopamine depletion [100].

Persistent striatal neuroinflammation is associated with sleep disturbances, cognitive deficits and behavioral changes [101] and may correlate with uncorrected nutritional status [102] or stress accompanying hypothalamic–pituitary–adrenal axis disruption [103].

The central role of neuroinflammation in PD includes its ability to impair mitochondrial and lysosome activity via dysregulating aSyn folding and clearance. Mitochondrial dysfunction is also connected to MAO-B activity in astrocytes [104].

Mounting evidence also shows that PD patients exhibit gut inflammation. The molecular mechanisms linking inflammation to aSyn pathology have been described in the “Gastrointestinal symptoms” paragraph of the present review.

Both neurons and glia can release adenosine which when acting on purinergic neurons leads to the secretion of pro-inflammatory cytokines, and the activation of microglia.

These findings raise intriguing questions about the role of neuroinflammation in NMS in PD, which although still questioned, requires further investigations before the development of treatments against this multifaceted and omnipresent phenomenon in PD patients.

2.5. Lipids

There is increasing evidence that high levels of oligomeric polyunsaturated fatty acids (PUFA’s) play an important role in neuronal toxicity. Notably, lipid peroxidation of PUFAs represents a feature of PD [105]. Specifically, there is a substantial increase in reactive aldehyde species such as malondialdehyde, 4-hydroxy-2-nonenal, cholesterol lipid hydroperoxide, and F2-isoprostanes in the SNpc of patients with Parkinson’s as well as in the anterior cingulate cortex of PD subjects [105,106].

Many studies also showed physical interaction between aSyn and PUFAs which results in a higher propensity to pathological aggregation [107,108,109,110]. The mutual electrostatic interaction between aSyn and membranes containing negatively charged lipids is found to affect aSyn’s characteristics and the membrane’s composition. In particular, changes in the levels of fatty acids, sphingolipids, and cholesterol have been reported [111].

2.6. Autophagic/Lysosomal Disruption

Autophagic/lysosomal disruption is closely linked to aSyn toxicity, particularly in the context of PD. The autophagy-lysosome pathway is a crucial mechanism responsible for the degradation and recycling of damaged or misfolded proteins, which include autophagosome formation, fusion with lysosomes, and degradation by lysosomal hydrolases. Overexpression or aggregation of aSyn can disrupt the autophagy-lysosome pathway leading to the accumulation of misfolded aSyn in the brain, while inefficient clearance of aSyn aggregates facilitates their spread to neighboring cells, propagating proteotoxicity. The translocation of cytosolic aSyn to the lysosomal lumen is mediated by the binding of heat shock cognate 70 chaperone to the KFERQ sequence which, with other co-chaperones, directs aSyn to the chaperone-mediated autophagy (CMA) adaptor Lysosomal-Associated Membrane Protein 2 (LAMP2A) located on the lysosomal membrane. Under stress conditions, exposure of the KFERQ motif can trigger CMA to remove harmful aSyn. Dysregulation of CMA is related to impaired degradation of aSyn and may contribute to the pathogenesis of PD [112].

Autophagy of mitochondria, or mitophagy, is intricately regulated by specific posttranslational modifications that “tag” the cargo for degradation, and by the Tank-binding kinase 1-mediated phosphorylation of optineurin that enhances its binding to ubiquitin and light chain 3 (LC3). Once bound to the ubiquitinated mitochondria, adaptors recruit LC3 so that the mitochondria are encapsulated by a double membrane structure called the autophagosome. Then, the autophagosome fuses with a lysosome forming an autolysosome, where the mitochondria are degraded and recycled [113]. Elevated aSyn levels inhibit macrophagic flux. Moreover, mutant forms of aSyn, such as A53T and A30P, exhibit a stronger binding affinity for LAMP2A that impairs the efficiency of aSyn clearance [114]. A53T and E46K aSyn variants also engage functional LC3B monomers into insoluble microaggregates on the surface of late endosomes favoring aSyn exosome excretion and seeding [82].

2.7. Metals

Although the precise role of metals in the pathogenesis of PD is still debated, evidence suggests their involvement in conformational effects is related to the binding of metal ions to aSyn and subsequent aggregation and accumulation. Elevated levels of metals such as iron, zinc, aluminum, lead, and copper have been found in the brain and in the cerebrospinal fluid (CSF) of PD patients [115]. Albeit with different affinities and stoichiometries, metals, especially copper, can establish electrostatic interactions with the C-terminus of aSyn, and more strongly phosphorylate Tyr-125 and Ser-129 of aSyn (p-aSyn), increasing their propensity to fibrillation. The aggregation speed of the acetylated A53T variant is higher with respect to the wild-type protein, suggesting an intrinsic self-assembly of the aSyn mutants into aggregates [116]. The toxicity of iron has also been investigated. Abeyawardhane et al. [117] suggest that the impact of Fe(II) on aSyn structure is higher than Fe(III) because of its elevated reactivity with O[sub.2], resulting in the production of H[sub.2]O[sub.2] and triggering ß-sheet generation; it can act both as an initiator and as a potential allosteric cofactor of protein misfolding [118]. The interaction of aSyn with both oxidation states of copper ions (Cu(I) and (Cu(II)) generates ROS, and contributes to OS potentially leading to its aggregation into pathological fibrils [119]. Aluminum is a metal that can cross the blood-brain barrier (BBB) and accumulate in the brain [120]. It has been found to co-localize with Biondi ring tangles in PD brains [121]. There is evidence suggesting that aluminum can induce OS, directly interact with aSyn, and disrupt the balance of essential ions in the brain, thus accelerating the fibrillation process of aSyn aggregation and contributing to the development and progression of PD.

3. Non-Motor Symptoms Associated with aSyn Pathology

Non-motor symptoms in PD include both sympathetic and parasympathetic dysfunctions and may concern, among other issues, neurobehavioral changes, pain, olfaction impairment, sleep and circadian dysfunctions, gastrointestinal symptoms, urogenital disturbances, and cardiovascular problems. Figure 2 illustrates the cardinal non-motor symptoms in the prodromal stage of PD.

3.1. Depression

The wide spectrum of NMS implicates the molecular interaction between the dopaminergic, glutamatergic, noradrenergic, and serotoninergic systems in which a significant number of neurons are lost [122]. Depression is a psychiatric condition estimated to affect approximately half of all PD patients [123]. Approximately 17% of individuals recapitulate symptoms consistent with major depression [124,125]. Research indicates that aSyn expression is upregulated in major depressive disorder (MDD) patients and that the pathology begins with the early accumulation in the olfactory system, particularly in the anterior olfactory nucleus, and then spreads to the limbic system. Moreover, neuroimaging studies revealed alterations in the limbic system including the cingulate gyrus, hippocampus, amygdala, hypothalamus, nucleus accumbens, ventral striatum, and orbitofrontal cortex [126]. Unfortunately, L-DOPA, the gold standard for the treatment of motor symptoms, does not alleviate depressive symptoms, indicating that depression is associated with deficits in serotoninergic neurotransmission in the brainstem, raphe nuclei, and limbic system circuitry [127,128]. In fact, serotonin and its metabolite, 5-HIAA, is decreased in the plasma of PD patients with depression [129]. Although the precise mechanisms for depression in PD are not fully elucidated, growing data reports the presence of aggregates of aSyn in the monoaminergic brainstem structure, specifically in dendritic and axonal processes that, associate with the loss of neurons in the aforementioned brain areas, giving rise to disturbances of neuroplasticity and depressive signs that occur in PD [130,131,132]. A recent study showed that adeno-associated virus (AAV5)-induced overexpression of wild-type human aSyn in r 5-HT neurons in the raphe nucleus causes progressive accumulation, phosphorylation, and aggregation of aSyn, an increases in brain-derived neurotrophic factor deficiency (BDNF), and alteration of 5-HT and norepinephrine (NE) systems, all affecting mood control [133]. Moreover, dysregulation of NE transmission may contribute to chronic neuroinflammation. Transgenic mice expressing human aSyn developed toxic species of aSyn in LC neurons, upregulation of GFAP with astrogliosis, microglial abundance, neuroinflammation, LC fiber degeneration, disruption of DA metabolism, and dysregulated NE neurotransmission, which results in changes of emotion and appearance of depressive symptoms [132,133]. Overexpression of aSyn in the hippocampus potently reduced the levels of the SNARE proteins Syntaxin, synapsin 1/2, and Vamp2 and activated C1qa, C1qb, C1qc, C4a, C4b, and C3 complement components, inducing gliosis and increasing hippocampal levels of IL-1b, IL6, and TNFa cytokines. Furthermore, the microglial phagocytosis marker CD68, lysosome marker Lamp1, and cell death markers Caspase-3 and Bax, were significantly increased in aSyn injected mice. These studies demonstrated that accumulation of aSyn in the hippocampus leads to the impairment of neurogenesis, synapse loss, and neuron death [130].

3.2. Anxiety

A high proportion of patients with PD (22.2–66.7%) display signs of anxiety, with females being at higher risk than male patients [134,135,136]. Several forms of anxiety are described in PD, including generalized anxiety, panic attacks, social phobia, agoraphobia, and obsessive-compulsive disorder. Genetic and epigenetic factors may be implicated in anxiety in PD patients. One study has suggested the involvement of gene variants of aSyn as well as LRRK2, DJ-1, PINK1, GBA, and BDNF [137]. Anxiety has been associated with genomic DNA levels of mutant aSyn in a transgenic mouse model overexpressing the human A53T aSyn [138]. Also, serotoninergic dysfunction was observed in an A53T aSyn mouse model [139].

Stoyka et al. [140] showed that intrastriatal injections of fibrils of aSyn caused abnormal accumulation of aSyn aggregates in the cortex, amygdala and pSer129-aSyn inclusions that are resistant to proteinase K in the cortex. Likewise, injection of aSyn preformed fibrils into the bilateral olfactory bulb of A53T transgenic mice spreads into connected regions provoking severe pathology in the hippocampus, bed nucleus of the stria terminalis, and central nucleus of the amygdala with a significant increase in total aSyn and p-aSyn. Animals exhibited hyposmia, anxiety-like behavior, and memory impairment, which are consequences of atrophy, neuronal loss, microglial activation and reactive astrogliosis [141]. These changes contribute to psychiatric disturbances. Miquel-Rio et al. [133] showed that overexpression of human aSyn in raphe 5-HT neurons leads to aSyn accumulation/aggregation in raphe nuclei and anxiety-like phenotypes.

Increase in hippocampal aSyn expression has also been detected in rats with high levels of innate anxiety, suggesting that anxiety can cause aSyn accumulation, in addition to being a contributor of synuclein progression [136]. Importantly, the vulnerability to forming inclusions is higher in glutamatergic excitatory neurons compared with inhibitory neurons. Alteration of calcium homeostasis and lower expression of proteins involved in the aSyn degradation machinery could accelerate the formation of aggregates [142,143].

The symptoms of anxiety, as well as depression and apathy in PD can also be caused by a marked loss of serotonergic neurons compared to dopaminergic neurons in the caudate nucleus, hypothalamus, and frontal cortex [144]. In addition, elevated levels of NE in plasma and CSF have been found in PD patients [145].

3.3. Psychosis

Psychosis is a premotor neuropsychiatric condition affecting PD patients [146], characterized by a spectrum of mental symptoms including illusions, delusions, visual hallucinations, schizophrenia (SCZ), and rapid eye movement sleep behavior disorder that are the result of dopamine hyperactivity in the mesolimbic pathway [147,148] and dysfunction of neurotransmitter systems [149]. In the past, psychotic individuals such as schizophrenics treated with antidopaminergic and neuroleptic drugs manifested parkinsonism symptoms. The use of newer atypical antipsychotic drugs has significantly reduced the incidence of psychiatric symptoms [150]. On the other hand, PD psychosis (PDP) is a common side effect derived from dopamine used to treat motor symptoms. However, some data indicates that psychosis may predate motor signs even in the absence of therapy as well as in PD patients, and when manifested is associated with high mortality and morbidity and heavily impacts the quality of life of many patients. There is controversial evidence regarding the role of aSyn in the pathophysiology of PDP. For example, studies have reported downregulation of aSyn expression [151] and lower levels of serum aSyn in SCZ patients compared to healthy controls [152] while another study failed to identify significant differences between SCZ patients and healthy controls [153]. A case study reported that duplication of the SNCA gene was associated with the progression of PD [154]. Additionally, aSyn aggregates could contribute to neuroinflammation, OS, synaptic dysfunction, and downstream neurotransmitter imbalance, the main features of schizophrenia [155]. Furthermore, a pilot study revealed the association between the rs356219 polymorphism in the SNCA gene and psychiatric disorders compared to healthy controls [156]. Data from a human study described aSyn deposition in specific regions involved in directing attention toward visual targets eliciting hallucination in dementia with Lewy bodies [157]. Furthermore, transgenic rats overexpressing human aSyn (aSyn-BAC, harboring the full-length human SNCA locus) showed elevated levels of dopamine as a compensatory response, that was accompanied by aberrant aSyn formation such as hyperphosphorylation, and monomeric C-terminal truncation followed by aggregation pathology. These alterations produced a psychosis-like phenotype [148]. A recent study supports the hypothesis that aberrant lysosome enzyme activities and sphingolipid metabolism may be responsible for aSyn accumulation and SCZ. In particular, decreases in acid sphingomyelinase and increases in alpha-galactosidase activities and aSyn levels were observed in late-onset SCZ patients in comparison to controls [158]. Moreover, an inverse correlation between apolipoprotein E and aSyn was described in PD SCZ [159]. An impaired ubiquitin/proteasome system and mitochondrial dysfunction could contribute to the reduction of the striatal cytochrome oxidase, energy metabolism, and glucose utilization in SCZ [160]. Alteration of the STAT3/mTORC2 signaling pathways in the hypothalamus of Thy1-aSyn transgenic mice seems to associate with the disruption of feeding behavior and energy metabolism and interfere with aSyn-linked pathology and SCZ phenotypes [161].

3.4. Cognitive Impairment

Cognitive decline is a prominent feature of PD and dementia with Lewy bodies (DLB) which comprises deficits ranging from mild cognitive impairment to severe PD dementia (PDD).

It is not fully understood whether aSyn directly impacts cognition and cognitive progression in PD. However, studies have mainly focused on changes in aSyn levels, properties, and function. High levels as well as low levels of total aSyn in the CSF are found to worsen cognitive performance [162,163]. A prospective study in PD patients and controls demonstrated that low levels of Aß42 in the CSF were identified as an aSyn-independent predictor of cognitive decline in PD. Aß[sub.42] is a peptide that is associated with Alzheimer’s disease, the most prevalent form of dementia. Nevertheless, high plasma aSyn levels have recently been associated with a higher prevalence of cognitive decline and faster progression of clinical dementia [164]. Likewise, LB pathology, evaluated by a seed amplification assay [165,166,167,168] is associated with increased progression of DLB and rapid early alterations in cognitive performance [169]. These findings suggest a transport of CNS aSyn to the periphery that could help clinicians detect, by non-invasive practices, the aSyn levels as early as possible serving as a potential surrogate biomarker of risk of cognitive impairment. A recent transcriptomic study in an aSyn-based model of PD showed region-specific gene expression changes associated with cognitive deficit [170].

Mutations in the aSyn gene, including A53T, E46K, H50Q, G51D, A53E/A53V, and aSyn triplication are correlated with cognitive decline [171]. The interplay between LRRK2 and aSyn phosphorylation was demonstrated in postmortem PD patients [156]. Additionally, Rab GTPases are substrates of LRRK2, and its phosphorylation positively regulates aSyn aggregation [156].

Many microRNA including miR-7, miR-129, miR-135a, miR-153 have been recognized to post-transcriptionally regulate aSyn and thereby associated with cognitive decline [172].

Importantly, aSyn in the plasma, urine, CSF and other biofluids, can be derived from lipid bilayer-encapsulated particles secreted by cells, known as extracellular vesicles, that reflect the internal status of neurons. High levels of EV aSyn have been found in Alzheimer’s and PD patients and their presence has been associated with the prion-like propagation of aSyn aggregates.

Lysosome dysfunction stimulates the release of aSyn-containing vesicles in recipient cells, and although the precise role remains to be elucidated, ubiquitination and sumoylation have been proposed as possible mechanisms for aSyn exosome secretion, especially in condition of impaired autophagy [173].

Furthermore, it has been shown that the ß-glucocerebrosidase substrate, glucosylceramide, may affect the stabilization of soluble oligomeric intermediates of aSyn accelerating the self-propagation of disease and inducing dementia. In turn, aSyn inclusions may inhibit ß-glucocerebrosidase activity by interfering with ER-Golgi trafficking and thereby propagating further accumulation of aSyn [174]. It is important to note that tau filaments and aSyn synergistically promote fibrillation contributing to the development of cognitive impairment in PD patients [175]. The levels of Aß plaques and tau tangles are also correlated with cognitive performance in patients with PDD suggesting the coexistence of amyloid ß and tau pathology [176].

Aside from aSyn accumulation, oxidative stress, astrocyte-associated inflammation and astrogliosis occurring in brain regions including the frontal cortex and hippocampus can promote cognitive dysfunction in PD. Pyramidal cells in hippocampal fields CA3 and granule cells of the dentate gyrus as well as neurons within the amygdala are vulnerable to oxidative stress with negative functional consequences on dendritic growth and neuronal connectivity.

Several mutations of astrocytic TAR DNA binding protein (TDP-43) in the hippocampus have been shown to increase oxidative stress, mitochondrial dysfunction and lipid peroxidation [177] further contributing to the aSyn neurotoxicity and progressive memory deficit in PD [178].

The improvement of the diagnostic and prognostic roles of aSyn in cognitive deficit in PD could derive from its validation in large independent cohort studies.

3.5. Pain

Neuropathic and inflammatory pain are common NMS in PD that can develop many years before the onset of classic motor signs and significantly impact the quality of life of patients. These NMS have been reported in approximately 60–70% of patients and can vary in nature and prevalence. According to Ford’s classification, five types of pain are proposed: musculoskeletal pain, radicular-neuropathic pain, dystonic pain, central neuropathic pain, and akathisia [179]. Central pathways involved in PD pain comprise the lateral pain pathways with projections to the thalamus, primary somatosensory cortex, and the medial spinoreticulothalamic pathways intimately associated with the autonomic nervous system, containing fibers that project to the medullary core and mesencephalon. Neuronal loss and Lewy body formation occur in the medial pain pathways, specifically in the parabrachial nucleus coeruleus and the periaqueductal grey [180]. aSyn misfolding and aggregation may originate in peripheral nerves fibers and spread along neuroanatomical sensory connections causing a multitude of painful perceptions [181]. Inflammatory pain in PD is caused by an abundance of immune responses in the afferent nociceptive nerve fibers that result in sympathetic nerve fiber damage and leads to the development of characteristic signs of neuropathic pain. PD patients experiencing pain exhibit changes in plasma levels of inflammatory cytokines such as IL1, IL-6, IL-10, and TNFa that actively participate in the perturbations of the proteostasis network, aSyn misfolding, and aSyn proteotoxicity [182].

In a spared nerve injury (SNI) model of neuropathic pain, the expression of aSyn has been detected in peptidergic and non-peptidergic nociceptive neurons in the dorsal horn of the spinal cord, signifying an involvement in pain transmission in the CNS. In the same model, the inhibition of aSyn was associated with the suppression of the pronociceptive MAP kinase signaling and an increase in inflammatory mediators in the spinal cord, probably consequent to the activation of immune cells [183]. In addition, Chen et al. [184] showed that d-Opioid receptor activation attenuated MPP(+) and hypoxia induced aSyn overexpression/aggregation by enhancing CREB phosphorylation and TORC1/SIK1/CREB pathways. Yi et al. [185] have found that spinal cord ligation is able to induce increases in PINK expression and aberrant mitophagic flux selectively in GABAergic interneurons that may be related to neuropathic pain in neurodegenerative diseases. Interestingly, deficiency of the TMEM175 neuronal proteins impairs lysosomal and mitochondrial function and increases aSyn aggregation [186]. Overall, these findings highlight that aSyn homeostasis represents a key pathogenic factor in the different types of pain.

3.6. Olfactory Dysfunction

Olfactory dysfunction (OD) is an early NMS symptom of PD affecting more than 90% of patients whose underlying mechanisms are partly defined. Anosmia, the loss of the sense of smell, and ageusia, the loss of taste, have been described as pre-motor symptoms of PD widely recognized as a very early biomarker of the disease and strongly associated with cognitive function [187].

Post-mortem studies of PD brain indicate that aSyn pathology starts in the OB and lower brainstem and developments in limbic system and connected brain regions leading to hyposmia [188].

Animal models created by injecting recombinant preformed aSyn fibrils in the bilateral olfactory bulb (OB) exhibit Lewy Body-like pathology and OD [189,190]. Additionally, the human A30P mutant aSyn-expressing mouse model displays OD along with a reduction in OB neurogenesis and alterations in synaptic vesicular transport [191]. The induction of the aSyn aggregates via overexpression of double mutant human aSyn (A53T and A30P) in the OB, negatively impacts neural activity and odor-evoked response in the OB [192]. A recent study using A53T mutant mice revealed hyperactivity of mitral/tufted cells and disruption in excitation/inhibition balance in that OB that impaired GABAergic transmission and abnormal expression of GABA transporters [193]. A study conducted using [sup.A53T]-a-synuclein transgenic mice revealed a deficit of odor discrimination and odor detection accompanied by a large loss of cholinergic neurons on the mitral cell layer, a decrease in acetylcholinesterase activity, and an alteration in dopamine neurotransmission [194]. The vicious cycle by which the abnormal aggregation of aSyn eliminated neurons, astrocytes, and microglia causes neuroinflammation that in turn promotes additional aSyn aggregation and deposition, has been considered a possible cause of aSyn accumulation in the OB and cell degeneration.

Intranasal lipopolysaccharide challenge in mice was able to trigger the activation of microglia and IL-1ß release followed by the recruitment of IL-1 receptor type I-dependent signaling, indicating that the neuroinflammatory response may be responsible for aSyn pathology [195]. Finally, contracting viral infection during childhood as well as diseases such as asthma, and allergic rhinitis which predispose to the entry of pathogens via the olfactory route, potentially increases the likelihood of developing PD [196,197]. Many viruses, including influenza A, Coxsackie, herpesvirus, and Epstein Barr, have been implicated as either a direct or indirect cause of PD and considered as a prodromal disease [197]. Respiratory viruses enter the brain via the olfactory bulb or Meissner’s plexus, disrupt the BBB, and activate the innate CNS immune system initiating an inflammatory response that persists many years after following the insult generating severe secondary sequelae, similar to parkinsonism. The induction of chemokine and cytokine genes following infections are mediated by TLR signaling and downstream signaling cascades that culminate with the increase in the amount of monomeric and oligomeric aSyn [198]. The inflammatory response induced by the neuro-protective effects of a-synuclein may contribute to its own aggregation. This is an important issue that deserves further investigation.

3.7. Visual Impairment

A variety of visual alterations, including ocular, visuoperceptive, and visuospatial impairments have been associated with PD and are mainly caused by DA depletion and loss of amacrine and inner plexiform cells of the retina. Therefore, eye movement, visual acuity, tritan axis of color perception, recognition of visual stimuli, and spatial relationships between objects are substantially compromised in PD patients [199].

aSyn toxicity was found as Lewy neurites in the retina and optic nerve and oligodendroglial cytoplasmic inclusions in the optic nerve. aSyn aggregates and phospho-Syn positive Lewy body-like neurites have also been described in the retina of PD patients, especially in the ganglion cell layer, the inner nuclear layer, and the inner plexiform layer. These aggregates correlate with retinal neuronal death, reduced retinal DA levels, and motor scores at various stages of the disease [200]. Overexpression of human p-aSyn has been observed in the outer nuclear layer (ONL) of the retina of A53T mice and manifests as structural thinning in the ONL and loss of electroretinography that measures a-wave photoreceptor response [201]. A phosphorylated form of aSyn has also been detected in wholemount human retinal nerve fiber and ganglion cell layers from PD and PDD subjects.

A research study has demonstrated that overexpression of aSyn in the retina led to the formation of 129-phosphorylated aSyn, changes in visual acuity, significant loss of TH-positive amacrine cells, and a decrease in dopaminergic amacrine neurotransmission [202]. Moreover, the intravitreal injection of PFF in mice induces the formation of pS129 aSyn inclusions in the visual cortex, perirhinal and entorhinal cortices, alongside an increase in Iba-1, a known marker of microglia activation in the optic nerve. Therefore, it is reasonable that synuclein pathology may be mediated by optic nerve inflammation or by systemic inflammation that spreads to the optical system [203]. It remains to be clarified whether aSyn impairs the function of different intracellular organelles such as the RE, Golgi apparatus, and the mitochondria or whether organelle dysfunction leads to aSyn toxicity. Although further studies need to be performed, crosstalk between aSyn and mitochondrial disfunction has also been previously described. Amacrine cells are interneurons in the retina that form synapses at their presynaptic endings, called lobular appendages. In PD, aSyn accumulation induces mitochondrial loss in the lobular appendages that are consistent with an energetic failure, loss of connexin 36, and impairment in visual signal transmission [204]. Another study using retinal-pigment-epithelial cells revealed that the accumulation of aSyn in these cells worsens lysosome activity by disrupting the trafficking of lysosomal hydrolases. Reduced ferrinophagy can also occur provoking retinal iron dyshomeostasis and consequent cytotoxicity [205].

Overall, these findings suggest the possibility of detecting aSyn in the retina as a promising non-invasive approach for a prodromic biomarker for PD.

3.8. Sleep and Circadian Dysfunctions

Sleep and circadian dysfunctions are recurrent non-motor symptoms of synucleinopathies, and significantly correlate with poorer quality of life [206].

Among the most common sleep-wake disturbances are rapid eye movement (REM) behavior disorder (RBD), a non-familial sleep disorder characterized by dream enactment behaviors the loss of atonia during REM sleep (REM sleep without atonia or RSWA) [207]. RBD is strictly associated with synucleinopaties [208] to the point that idiopathic/isolated RBD (iRBD), that may precede the onset of motor features by decades, is considered a prodromal form of synucleinopathy and is therefore a highly specific marker for future development of the pathology. Furthermore, aSyn aggregates are considered a biomarker for the development of diagnostic assays. For example, aSyn aggregates in stool samples of iRBD patients can be detected and measured, supporting the diagnosis of prodromal synucleinopathies [209].

Transgenic mice overexpressing human A53T a-syn (SNCA[sup.A53T/-]) in its natively expressed regions exhibited REM sleep without atonia, similar to RBD patients, at 5 months of age [210]. Compared to wild-type mice, in whom REM sleep is characterized by sinusoidal theta-frequency electroencephalographic activity and absent or minimal electromyogram activity, SNCA[sup.A53T/-] mice displayed excessive muscle twitches in body and limb during REM sleep as well as transient increases in muscle activity [208,211,212].

Restless leg syndrome (RLS), a very common movement disorder manifested by disturbing symptoms in lower limbs, more severe in the evening or night, has a high prevalence in PD patients [213] and some studies report the onset of PD at a younger age in subjects with RLS [214].

Insomnia and excessive daytime sleepiness (EDS) are also common in PD patients. Insomnia has been associated with depressed mood, autonomic symptoms fatigue, and age [215]. Subjective EDS has been reported in up to 50% of patients with PD [212]; it has been associated with the neurodegenerative process itself, that is extended to the dopaminergic and non-dopaminergic neurons in the lower brainstem and midbrain [216] involved in sleep-wake regulation [217]. These observations of poorly consolidated rest/activity patterns in humans are paralleled in animal models. Rodent models for PD display impairment in the sleep-wake parameters, such as deficit in REM sleep, overwhelming episodes of sleep, similar to “sleep attacks”, and increased sleepiness [218,219]. In Drosophila, the pan-neuronal overexpression of pre-fibrillar human aSyn oligomers impacts the sleep-like rest behavior (defined as the absence of movement for 5 consecutive minutes), in terms of number and length of sleep episodes and total sleep [220,221,222,223]. Flies exhibit decreased sleep (especially at night), increased number of sleep bouts (indicating sleep fragmentation), and decreased wake activity [220,222,223]. These alterations precede the onset of motor symptoms and can be related to the presence of pre-fibrillar aSyn oligomers that prevent protein aggregation [220,221,223].

Sleep is coordinated by the interaction of homeostatic and circadian mechanisms, that regulate sleep debt and the periodicity of sleep/wake propensity, respectively [224]. Circadian rhythms are controlled by an endogenous mechanism that comprises: a pacemaker, orchestrated by interconnected transcriptional/translational feedback loops, input pathways for light and other stimuli that synchronize the pacemaker to the environment and output pathways that convert the molecular oscillation of clock components in overt rhythms [225]. In the primary loop, the molecular oscillation involves the transcription factors CLOCK and BMAL1, which, as a heterodimer, regulates the rhythmic expression of output genes. The CLOCK/BMAL1 complex also activates the expression of the negative elements of the loop, Period, and Cryptochrome genes, whose products inhibit their own expression by inactivating the activity of CLOCK/BMAL1 [225]. The circadian physiology is based on a hierarchical network of central and peripheral oscillators; the central pacemaker (master clock) is located in the suprachiasmatic nuclei (SCN) of the hypothalamus: it receives signals from the environment and transmits temporal information to downstream peripheral clocks, located in organs such as the heart, lungs, liver and adrenal glands, through neurotransmitters and neuromodulators [225]. Melatonin secretion is a rhythmic output generated by the SCN: melatonin and circadian clock gene expression can be easily measured in serum and peripheral blood mononuclear cells, respectively, serving as reliable markers of circadian rhythmicity [225].

Several studies have reported lower levels and dampened melatonin oscillation and a lack of time-dependent variation in BMAL1 expression in PD patients, compared to healthy controls, indicating that the peripheral molecular clock is affected in PD [109,211,226].

Additionally, in Drosophila, the sleep defects reported in flies overexpressing pre-fibrillar human aSyn oligomers are associated with a severe impairment of two key circadian features, the anticipation of the dark/light transition and the circadian periodicity [220].

Daily metabolic demand of an appropriate distribution of metabolites within tissues is regulated by circadian rhythm. These metabolites in turn regulate many proteins of the clock machinery [227]. Cutting-edge metabolomic studies could help map the distribution of the metabolome in brain tissues. An interesting study by Peek et al. showed that NAD[sup.+] metabolite is crucial in regulating the circadian rhythm and also activating the NAD[sup.+]-dependent sirtuin 3 (SIRT3), a mitochondrial protein playing a crucial role in mediating mitochondrial oxidative function and autophagy [228]. It has been found that deposition of aSyn oligomers in mitochondria causes downregulation of SIRT3 activity and a decrease in mitochondrial biogenesis, indicating a crosstalk between aSyn, SIRT3 and circadian rhythms [229].

3.9. Gastrointestinal Symptoms

The involvement of aSyn as an important player in the complex process leading to GI symptoms in PD is well documented. Several large cohort and case-control studies show correlations between gastrointestinal (GI) dysfunctions and PD, DLB, and PDD during the disease course and overlap with autonomic symptoms [230,231].

GI disorders have been suggested to both cause PD as well as increase the risk of developing PD. Two distinct subtypes of PD have been initially proposed based on the origin and progression of aSyn pathology: a “brain-first” subtype, in which aSyn pathology originates in the OB or other brainstem regions and then spreads to the peripheral autonomic nervous system, and a “body-first” subtype in which the pathological process originates in the enteric nervous system and then ascends via the vagus nerve or other autonomic pathways to reach the brain [232]. However, recent post-mortem studies showed that Lewy pathology is triggered in the GI tract or autonomic nervous system without concomitant involvement of the OB, therefore supporting a single-hit brain-first hypothesis [233,234].

Loss of enteric dopamine cells and degeneration of vagal nuclei represent the main cause of GI symptoms such as dysphagia, sialorrhea, bloating, nausea, vomiting, gastroparesis, and constipation.

p-aSyn deposits in vagus nerve Schwann cells resulted in inflammatory response induced by interaction and activation of a family of pattern recognition receptors, Toll-like receptor 2 (TLR2), implicated in regulation of aSyn release [235]. On the other hand, the interaction between p-aSyn and TLR2/4 has been previously described in murine models of PD as well as in post-mortem human PD brain and is associated with pro-inflammatory responses and PD pathology [236,237]. p-129Syn was detected in submandibular gland samples in PD patients as well as the distal esophagus, while a progressive decrease was observed from the stomach to the small and large intestine and rectum [238].

GI symptoms affect nearly 80% of subjects before or after the onset of motor manifestations and some of them are life-threatening symptoms, such as dysphagia which increases the risk of pneumonia. Furthermore, PD patients experience a more widespread onset of lifelong GI symptoms, and constipation is widely recognized as one of the most prevalent NMS of PD. The intestinal microbiome also play a role in the bidirectional communication between the brain and the gut has emerged as a significant aspect of PD pathology [239,240,241].

Aberrant aggregation of aSyn is responsible for vagus nerve and ENS degeneration, decreased intestinal peristalsis, and constipation [242,243]. A study revealed that human A53T aSyn transgenic mice exhibit severe signs of GI dysfunction together with aSyn aggregation in the ENS [244]. Interestingly, it has also been shown that pathological species of aSyn can be carried through extracellular vesicles secreted by red blood cells into the GI tract, contributing to the onset and/or progression of this PD pathology.

Gut microbiota of PD patients have been found to be rich in Enterobacteriaceae [245]. This microorganism family, natively expresses aSyn, and is able to perform anaerobic nitrate respiration with the production of antioxidant nitrite which initiates a cascade of oxidative events that lead to aSyn aggregation [246].

In recent years, many have speculated about the role of short-chain fatty acids (SCFAs) produced by the gut microbiome, especially butyrate and propionate, on the development of synculeinopathies. The effects of short-chain fatty acids (SCFAs) appear paradoxical and contradictory, likely influenced by factors such as injection routes, mixture composition, and concentration. [247,248,249,250]. For example, lower SCFA levels and higher calprotectin detected in the stool of PD patients correlated with GI symptoms [251]. An interesting preclinical study conducted in an aSyn pre-formed fibrils (aSyn PFFs)-induced rat model of PD documented protective effects of sodium butyrate that reduced inflammatory markers such as TNFa, IL-1, IL-6, and increased DA content [252]. Moreover, fecal transplants from PD patients to aSyn overexpressing mice have been shown to promote aSyn-dependent activation of microglia that was associated with worsening motor deficit, suggesting an important role for gut microbiota in regulating the abnormal aggregation of aSyn, and the involvement of microbiota-gut-brain-axis pathways [253]. For this reason, transplanting probiotics into the gastrointestinal tract of PD patients could be a promising strategy to restore gut dysbiosis, reduce inflammation, and elude aSyn alteration and proteotoxicity.

3.10. Sexual and Urinary Dysfunctions

Sexual and urinary dysfunctions are common NMS in PD patients that involve neuronal cytoplasmic aSyn inclusions in the peripheral autonomic nerve fibers. In approximately 70% of patients, nocturia is accompanied by urge urinary incontinence and detrusor hyperactivity, often followed by the appearance of motor symptoms [254,255].

aSyn aggregates and p-aSyn have been found in the frontal cortex, basal ganglia, pontine nuclei, sacral spinal cord, pelvic plexus, and genitourinary tract of PD patients with urinary dysfunction [256]. Significantly lower levels of dopamine transporter uptake, possibly a consequence of degeneration of the nigrostriatal dopaminergic neurons and caudate nucleus, have been found in PD, PDD and DLB patients with lower urinary tract symptoms [257,258,259,260]. An animal study has also revealed the disruption of D1-GABAergic striatal output neurons as a possible cause of bladder hyperactivity in PD, suggesting the complex orchestra behind urinary impairment in PD [261].

Data analysis by employing multidimensional self-administered questionnaires to evaluate sexual performance and relationship dissatisfaction suggests that there is a high prevalence of sexual dysfunction among young-onset male and female PD compared to healthy subjects [262], which is related to severity of depressive symptoms especially in female, motor, psychological, cognitive disturbances [263]. Generally, sexual dysfunction appears after the development of motor symptoms. Tremor, bradykinesia, dyskinesia as well as medication such as dopamine agonists, can interfere with sexual function. However, an elevated number of patients experience sexual dysfunction many years before PD onset. Gender difference in sexual dysfunctions arise from hormones. In fact, a higher protection from DA depletion is provided by the higher exposure to estrogens in females. A number of studies have shown a range of symptoms ranging from hyposexual to hypersexual behaviors. For example, erectile dysfunction has been reported in diagnosed male PD and DLB cases, and loss of libido and org*smic dysfunction were found in both women and men [264] while hypersexual behaviors are often associated with PD treatment [250]. A study has demonstrated impairment in courtship rituals and in copulation in a Drosophila A30P-mediated PD model [265]. A study in PD patients revealed that sexual dysfunction is more frequent in early-onset PD patients compared to late onset PD patients [266]. The control of sexual functions depends on the projections from multiple areas of the brain to the spinal cord. The parasympathetic system is involved in erectile function and in the release of neurotransmitters such as nitric oxide and acetylcholine. The hypothalamus also plays an important role in controlling sexual performance, contributing to the interaction between nervous and endocrine systems and to the exchange of messages between the brain and the spinal cord.

It is possible that various toxic insults could interrupt or impair this intricate cross-talk inducing, the development of insoluble inclusions, such as aSyn oligomers, and triggering toxic events that lead to neuronal death. Although many other studies need to be performed to identify the molecular mechanisms underlying sexual dysfunctions, genetic factors, such as polymorphisms in the dopamine D4 receptor gene, appear to contribute to individual differences in human sexual behavior [267]. All these studies underline the importance of sexual dysfunction in PD as they negatively impact the quality of life of patients and partnership. A such, it is equally important to address sexual and urinary dysfunction in PD, since symptoms can be treated.

3.11. Cardiovascular Symptoms

Epidemiological studies have suggested a possible relationship between the incidence of cardio-cerebrovascular disease and PD, either as risk factors or as manifestations of PD itself [268,269]. Studies using Mendelian randomization revealed that PD is correlated with a high risk of coronary artery disease, stroke, ischemic stroke, and cardioembolic stroke compared with age- and gender-matched healthy individuals [270,271]. On the other hand, the deposition of aSyn in the brain of PD patients may be a potential pathogenic contributor to cardiovascular disease and stroke. The involvement of the cardiovascular system in PD reflects aSyn deposition in sympathetic noradrenergic nerves, defects of the autonomic cardiac innervation and abnormal function of residual noradrenergic endings [272].

The loss of myocardial noradrenergic innervation together with the loss of myocardial norepinephrine caused by Lewy body deposition are responsible for orthostatic hypotension with substantial drop in systolic and diastolic blood pressure of at least 20 mmHg and 10 mmHg, respectively [273]. In PD patients, a significant alteration in the electrophysiological activity and remodeling of the myocardium has also been described. The multi-organ deposition of aSyn in PD extends to myocardial tissue, signifying that aSyn fibrils dispersed in the myocardium may be pathogenic. Myocardial tissues from PD autopsies display p-aSyn deposits and Lewy body pathology that correlate with denervation of sympathetic and noradrenergic endings in cardiac tissue [274,275,276]. Accumulation of aSyn aggregates in paravertebral sympathetic ganglia appear to be chronologically preceded by those in the distal axons of the cardiac sympathetic nervous system, suggesting centripetal degeneration [277]. Of note, cardiac dysfunction can be present even when CNS aSyn deposition is limited to the brainstem, with an absence of clinical symptoms [278,279]. Nigrostriatal dopaminergic neurons are vulnerable to high glucose levels and to alterations in insulin signaling and undergo severe injury producing high levels of ROS, via mitochondrial destruction, and low levels of the antioxidant glutathione. Hyperglycemia associated with high uric acid levels cause inflammation and increase in the risk of cardiovascular disease (CVD) in PD [280].

Additionally, high levels of oxidated low-density lipoproteins (LDL), and LDL-cholesterol, as well as changes in sphingolipid and ceramide metabolism, have been found in PD patients, supporting a higher risk of CVD consequent to a greater propensity to atherosclerotic plaque formation. On the other hand, the interaction between cholesterol and aSyn appears to facilitate aSyn aggregation [281]. It remains unclear whether cardiac a-syn accumulation plays a causative role for orthostatic hypotension in PD people [282].

4. Conclusions

Given the growing incidence and the long list of NMS in the prodromic stage of PD, as well as the negative impact on health-related quality of life, it is imperative for clinicians to achieve an early diagnosis aimed at prompt intervention to alleviate the symptoms and hopefully slow the progression of the disease. The correlation between aSyn deposition and the appearance of NMS in the 2- to 20-year pre-motor deficit period is well-documented, although the exact mechanisms have not been fully elucidated. Different proteoforms of aSyn have been observed not only in the central nervous system (CNS) but also in peripheral tissues and biofluids such as cerebrospinal fluid, plasma, saliva, olfactory mucosa, skin, salivary glands, retina, adrenal medulla, heart, and gastrointestinal tract in PD cases. Interestingly, the possibility to detect and quantify aSyn conformers in biofluids and in tissue biopsies reflects on the multisystem nature of the disorder and represents a promising diagnostic approach at the preclinical stage of PD, when neurodegeneration commences. Randomized controlled trials, cohort and cross-sectional studies and the validation of PD-specific screening questionnaires together with non-invasive and painless sample collection procedures and sophisticated technology for aSyn measurement can contribute to the achievement of timely diagnosis, monitoring and management of PD.

Carrying out robust clinical trials in large populations with the careful application of objective biomarkers and assessment of confounding factors will be essential to consolidate the therapeutic potentials of antioxidants for PD.

Author Contributions

Conceptualization, C.C. and G.M.M.; Writing—original draft, C.C. and G.M.M.; Writing—review and editing, C.C. and G.M.M.; Visualization C.C. and G.M.M.; Supervision C.C. and G.M.M. All authors have read and agreed to the published version of the manuscript.

Institutional Review Board Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Acknowledgments

Figures have been created with Biorender.com.

References

1. T.J. Stevenson; H.C. Murray; C. Turner; R.L.M. Faull; B.V. Dieriks; M.A. Curtis a-Synuclein Inclusions Are Abundant in Non-Neuronal Cells in the Anterior Olfactory Nucleus of the Parkinson’s Disease Olfactory Bulb., 2020, 10,p. 6682. DOI: https://doi.org/10.1038/s41598-020-63412-x. PMID: https://www.ncbi.nlm.nih.gov/pubmed/32317654.

2. F.N. Emamzadeh; A. Surguchov Parkinson’s Disease: Biomarkers, Treatment, and Risk Factors., 2018, 12, 612. DOI: https://doi.org/10.3389/fnins.2018.00612. PMID: https://www.ncbi.nlm.nih.gov/pubmed/30214392.

3. R. Durcan; L. Wiblin; R.A. Lawson; T.K. Khoo; A.J. Yarnall; G.W. Duncan; D.J. Brooks; N. Pavese; D.J. Burn Prevalence and Duration of Non-Motor Symptoms in Prodromal Parkinson’s Disease., 2019, 26,pp. 979-985. DOI: https://doi.org/10.1111/ene.13919. PMID: https://www.ncbi.nlm.nih.gov/pubmed/30706593.

4. A.H.V. Schapira; K.R. Chaudhuri; P. Jenner Non-Motor Features of Parkinson Disease., 2017, 18,pp. 435-450. DOI: https://doi.org/10.1038/nrn.2017.62. PMID: https://www.ncbi.nlm.nih.gov/pubmed/28592904.

5. C.C.G. Alonso; F.G. Silva; L.O.P. Costa; S.M.S.F. Freitas Smell Tests Can Discriminate Parkinson’s Disease Patients from Healthy Individuals: A Meta-Analysis., 2021, 211,p. 107024. DOI: https://doi.org/10.1016/j.clineuro.2021.107024. PMID: https://www.ncbi.nlm.nih.gov/pubmed/34823156.

6. B. Iravani; A. Arshamian; M. Schaefer; P. Svenningsson; J.N. Lundström A Non-Invasive Olfactory Bulb Measure Dissociates Parkinson’s Patients from Healthy Controls and Discloses Disease Duration., 2021, 7,p. 75. DOI: https://doi.org/10.1038/s41531-021-00220-8.

7. J. Hunt; E.J. Coulson; R. Rajnarayanan; H. Oster; A. Videnovic; O. Rawashdeh Sleep and Circadian Rhythms in Parkinson’s Disease and Preclinical Models., 2022, 17,p. 2. DOI: https://doi.org/10.1186/s13024-021-00504-w.

8. S.-C. Gu; R. Shi; C. Gao; X.-L. Yuan; Y. Wu; Z.-G. Liu; C.-D. Wang; S.-R. Zhao; X. Chen; C.-X. Yuan et al. Autonomic Function and Motor Subtypes in Parkinson’s Disease: A Multicentre Cross-Sectional Study., 2023, 13,p. 14548. DOI: https://doi.org/10.1038/s41598-023-41662-9. PMID: https://www.ncbi.nlm.nih.gov/pubmed/37666916.

9. A. Fanciulli; G.K. Wenning Autonomic Failure: A Neglected Presentation of Parkinson’s Disease., 2021, 20,pp. 781-782. DOI: https://doi.org/10.1016/S1474-4422(21)00292-1.

10. O.M.A. El-Agnaf; S.A. Salem; K.E. Paleologou; M.D. Curran; M.J. Gibson; J.A. Court; M.G. Schlossmacher; D. Allsop Detection of Oligomeric Forms of a-Synuclein Protein in Human Plasma as a Potential Biomarker for Parkinson’s Disease., 2006, 20,pp. 419-425. DOI: https://doi.org/10.1096/fj.03-1449com.

11. K. Kosaka Latest Concept of Lewy Body Disease., 2014, 68,pp. 391-394. DOI: https://doi.org/10.1111/pcn.12179. PMID: https://www.ncbi.nlm.nih.gov/pubmed/24597591.

12. C. Soto; S. Pritzkow Protein Misfolding, Aggregation, and Conformational Strains in Neurodegenerative Diseases., 2018, 21,pp. 1332-1340. DOI: https://doi.org/10.1038/s41593-018-0235-9.

13. R.E. Burke; W.T. Dauer; J.P.G. Vonsattel A Critical Evaluation of the Braak Staging Scheme for Parkinson’s Disease., 2008, 64,pp. 485-491. DOI: https://doi.org/10.1002/ana.21541. PMID: https://www.ncbi.nlm.nih.gov/pubmed/19067353.

14. N. Pyatigorskaya; M. Mongin; R. Valabregue; L. Yahia-Cherif; C. Ewenczyk; C. Poupon; E. Debellemaniere; M. Vidailhet; I. Arnulf; S. Lehéricy Medulla Oblongata Damage and Cardiac Autonomic Dysfunction in Parkinson Disease., 2016, 87,pp. 2540-2545. DOI: https://doi.org/10.1212/WNL.0000000000003426. PMID: https://www.ncbi.nlm.nih.gov/pubmed/27837003.

15. H. Braak; K. Del Tredici; U. Rüb; R.A.I. de Vos; E.N.H. Jansen Steur; E. Braak Staging of Brain Pathology Related to Sporadic Parkinson’s Disease., 2003, 24,pp. 197-211. DOI: https://doi.org/10.1016/S0197-4580(02)00065-9. PMID: https://www.ncbi.nlm.nih.gov/pubmed/12498954.

16. J. Dams; U. Siebert; B. Bornschein; J. Volkmann; G. Deuschl; W.H. Oertel; R. Dodel; J. Reese Cost-Effectiveness of Deep Brain Stimulation in Patients with Parkinson’s Disease., 2013, 28,pp. 763-771. DOI: https://doi.org/10.1002/mds.25407.

17. R. Qi; X. Geng; B. Huang; Y. Chen; H. Jiang; Y. Zou; W. Wang; Y. Li; Y. Li; L. Yin et al. Outcomes of STN-DBS in PD Patients with Different Rates of Disease Progression over One Year of Follow-Up., 2020, 11, 600. DOI: https://doi.org/10.3389/fneur.2020.00600.

18. T. Fundament; P.R. Eldridge; A.L. Green; A.L. Whone; R.S. Taylor; A.C. Williams; W.M.M. Schuepbach Deep Brain Stimulation for Parkinson’s Disease with Early Motor Complications: A UK Cost-Effectiveness Analysis., 2016, 11, e0159340. DOI: https://doi.org/10.1371/journal.pone.0159340. PMID: https://www.ncbi.nlm.nih.gov/pubmed/27441637.

19. B. Lilleeng; M. Gjerstad; R. Baardsen; I. Dalen; J.P. Larsen Motor Symptoms after Deep Brain Stimulation of the Subthalamic Nucleus., 2015, 131,pp. 298-304. DOI: https://doi.org/10.1111/ane.12342.

20. C. Listik; R.G. Cury; S.C.B. Casagrande; E. Listik; D. Arnaut; N. Santiago; V.A. Da Silva; R. Galhardoni; J.d.L.A. Machado; J.C.d. Almeida et al. Improvement of Non-Motor Symptoms and Quality of Life after Deep Brain Stimulation for Refractory Dystonia: A 1-Year Follow-Up., 2021, 12, 717239. DOI: https://doi.org/10.3389/fneur.2021.717239.

21. Z. Eghlidos; Z. Rahimian; G. Vadiee; S. Jahangiri Effects of Subthalamic Deep Brain Stimulation on Non-Motor Symptoms of Parkinson’s Disease: A Meta-Analysis., 2022, 146,pp. 115-125. DOI: https://doi.org/10.1111/ane.13652. PMID: https://www.ncbi.nlm.nih.gov/pubmed/35611557.

22. F. Alonso-Frech; C. Fernandez-Garcia; V. Gómez-Mayordomo; M.H.G. Monje; C. Delgado-Suarez; C. Villanueva-Iza; M.J. Catalan-Alonso Non-Motor Adverse Effects Avoided by Directional Stimulation in Parkinson’s Disease: A Case Report., 2022, 12, 786166. DOI: https://doi.org/10.3389/fneur.2021.786166. PMID: https://www.ncbi.nlm.nih.gov/pubmed/35173666.

23. N. El Ghazal; H. Nakanishi; A.E. Martinez-Nunez; N.K. Al Sabbakh; O.A. Segun-Omosehin; N.E. Bourdakos; M. Nasser; R.H. Matar; C. Than; O.A. Danoun et al. The Effects of Deep Brain Stimulation on Mood and Quality of Life in Parkinson’s Disease: A Systematic Review and Meta-Analysis., 2023, 15,p. e44177. DOI: https://doi.org/10.7759/cureus.44177. PMID: https://www.ncbi.nlm.nih.gov/pubmed/37753046.

24. S. Gupta; S. Shukla Non-Motor Symptoms in Parkinson’s Disease: Opening New Avenues in Treatment., 2021, 2,p. 100049. DOI: https://doi.org/10.1016/j.crbeha.2021.100049.

25. A.B.R. Leite Silva; R.W. Gonçalves de Oliveira; G.P. Diógenes; M.F. de Castro Aguiar; C.C. Sallem; M.P.P. Lima; L.B. de Albuquerque Filho; S.D. Peixoto de Medeiros; L.L. Penido de Mendonça; P.C. de Santiago Filho et al. Premotor, Nonmotor and Motor Symptoms of Parkinson’s Disease: A New Clinical State of the Art., 2023, 84,p. 101834. DOI: https://doi.org/10.1016/j.arr.2022.101834. PMID: https://www.ncbi.nlm.nih.gov/pubmed/36581178.

26. L. Maroteaux; J. Campanelli; R. Scheller Synuclein: A Neuron-Specific Protein Localized to the Nucleus and Presynaptic Nerve Terminal., 1988, 8,pp. 2804-2815. DOI: https://doi.org/10.1523/JNEUROSCI.08-08-02804.1988. PMID: https://www.ncbi.nlm.nih.gov/pubmed/3411354.

27. J. Burré; S. Vivona; J. Diao; M. Sharma; A.T. Brunger; T.C. Südhof Properties of Native Brain a-Synuclein., 2013, 498,pp. E4-E6. DOI: https://doi.org/10.1038/nature12125. PMID: https://www.ncbi.nlm.nih.gov/pubmed/23765500.

28. E. Cholak; K. Bugge; A. Khondker; K. Gauger; E. Pedraz-Cuesta; M.E. Pedersen; S. Bucciarelli; B. Vestergaard; S.F. Pedersen; M.C. Rheinstädter et al. Avidity within the N-Terminal Anchor Drives a-Synuclein Membrane Interaction and Insertion., 2020, 34,pp. 7462-7482. DOI: https://doi.org/10.1096/fj.202000107R. PMID: https://www.ncbi.nlm.nih.gov/pubmed/32277854.

29. M. Hashimoto; T. Takenouchi; M. Mallory; E. Masliah; A. Takeda; J.G. Culvenor; C.A. McLean; B.C.V. Campbell; C.L. Masters; Q.-X. Li et al. The Role of NAC in Amyloidogenesis in Alzheimer’s Disease., 2000, 156,pp. 734-735. DOI: https://doi.org/10.1016/S0002-9440(10)64777-3.

30. V. Gao; J.A. Briano; L.E. Komer; J. Burré Functional and Pathological Effects of a-Synuclein on Synaptic SNARE Complexes., 2023, 435, 167714. DOI: https://doi.org/10.1016/j.jmb.2022.167714.

31. J. Diao; J. Burré; S. Vivona; D.J. Cipriano; M. Sharma; M. Kyoung; T.C. Südhof; A.T. Brunger Native a-Synuclein Induces Clustering of Synaptic-Vesicle Mimics via Binding to Phospholipids and Synaptobrevin-2/VAMP2., 2013, 2,p. e00592. DOI: https://doi.org/10.7554/eLife.00592. PMID: https://www.ncbi.nlm.nih.gov/pubmed/23638301.

32. R. Bell; R.J. Thrush; M. Castellana-Cruz; M. Oeller; R. Staats; A. Nene; P. Flagmeier; C.K. Xu; S. Satapathy; C. Galvagnion et al. N-Terminal Acetylation of a-Synuclein Slows down Its Aggregation Process and Alters the Morphology of the Resulting Aggregates., 2022, 61,pp. 1743-1756. DOI: https://doi.org/10.1021/acs.biochem.2c00104. PMID: https://www.ncbi.nlm.nih.gov/pubmed/35944093.

33. J.R. Horsley; B. Jovcevski; T.L. Pukala; A.D. Abell Designer D-Peptides Targeting the N-Terminal Region of a-Synuclein to Prevent Parkinsonian-Associated Fibrilization and Cytotoxicity., 2022, 1870, 140826. DOI: https://doi.org/10.1016/j.bbapap.2022.140826. PMID: https://www.ncbi.nlm.nih.gov/pubmed/35926717.

34. I. Hmila; N.N. Vaikath; N.K. Majbour; D. Erskine; I.P. Sudhakaran; V. Gupta; S.S. Ghanem; Z. Islam; M.M. Emara; H.B. Abdesselem et al. Novel Engineered Nanobodies Specific for N-Terminal Region of Alpha-Synuclein Recognize Lewy-Body Pathology and Inhibit In-Vitro Seeded Aggregation and Toxicity., 2022, 289,pp. 4657-4673. DOI: https://doi.org/10.1111/febs.16376.

35. K. Daida; S. Shimonaka; K. Shiba-f*ckushima; J. Ogata; H. Yoshino; A. Okuzumi; T. Hatano; Y. Motoi; T. Hirunagi; M. Katsuno et al. a-Synuclein V15A Variant in Familial Parkinson’s Disease Exhibits a Weaker Lipid-Binding Property., 2022, 37,pp. 2075-2085. DOI: https://doi.org/10.1002/mds.29162. PMID: https://www.ncbi.nlm.nih.gov/pubmed/35894540.

36. S.H. Diaw; M. Borsche; L. Streubel-Gallasch; M. Dulovic-Mahlow; J. Hermes; I. Lenz; P. Seibler; C. Klein; N. Brüggemann; M. Vos et al. Characterization of the Pathogenic a-Synuclein Variant V15A in Parkinson’s Disease., 2023, 9,p. 148. DOI: https://doi.org/10.1038/s41531-023-00584-z. PMID: https://www.ncbi.nlm.nih.gov/pubmed/37903765.

37. D. Emin; Y.P. Zhang; E. Lobanova; A. Miller; X. Li; Z. Xia; H. Dakin; D.I. Sideris; J.Y.L. Lam; R.T. Ranasinghe et al. Small Soluble a-Synuclein Aggregates Are the Toxic Species in Parkinson’s Disease., 2022, 13,p. 5512. DOI: https://doi.org/10.1038/s41467-022-33252-6. PMID: https://www.ncbi.nlm.nih.gov/pubmed/36127374.

38. J.M. Lee; P. Derkinderen; J.H. Kordower; R. Freeman; D.G. Munoz; T. Kremer; W. Zago; S.J. Hutten; C.H. Adler; G.E. Serrano et al. The Search for a Peripheral Biopsy Indicator of a-Synuclein Pathology for Parkinson Disease., 2017, 76,p. nlw103. DOI: https://doi.org/10.1093/jnen/nlw103.

39. M. Chen; D.E. Mor Gut-to-Brain a-Synuclein Transmission in Parkinson’s Disease: Evidence for Prion-like Mechanisms., 2023, 24, 7205. DOI: https://doi.org/10.3390/ijms24087205.

40. F.S. Ruggeri; P. Flagmeier; J.R. Kumita; G. Meisl; D.Y. Chirgadze; M.N. Bongiovanni; T.P.J. Knowles; C.M. Dobson The Influence of Pathogenic Mutations in a-Synuclein on Biophysical and Structural Characteristics of Amyloid Fibrils., 2020, 14,pp. 5213-5222. DOI: https://doi.org/10.1021/acsnano.9b09676.

41. T.J. Krzystek; R. Banerjee; L. Thurston; J. Huang; K. Swinter; S.N. Rahman; T.L. Falzone; S. Gunawardena Differential Mitochondrial Roles for a-Synuclein in DRP1-Dependent Fission and PINK1/Parkin-Mediated Oxidation., 2021, 12,p. 796. DOI: https://doi.org/10.1038/s41419-021-04046-3. PMID: https://www.ncbi.nlm.nih.gov/pubmed/34404758.

42. M. Helwig; A. Ulusoy; A. Rollar; S.A. O’Sullivan; S.S.L. Lee; H. Aboutalebi; R. Pinto-Costa; B. Jevans; M. Klinkenberg; D.A. Di Monte Neuronal Hyperactivity–Induced Oxidant Stress Promotes In Vivo a-Synuclein Brain Spreading., 2022, 8,p. eabn0356. DOI: https://doi.org/10.1126/sciadv.abn0356. PMID: https://www.ncbi.nlm.nih.gov/pubmed/36044566.

43. I. Stojkovska; W.Y. Wani; F. Zunke; N.R. Belur; E.A. Pavlenko; N. Mwenda; K. Sharma; L. Francelle; J.R. Mazzulli Rescue of a-Synuclein Aggregation in Parkinson’s Patient Neurons by Synergistic Enhancement of ER Proteostasis and Protein Trafficking., 2022, 110,pp. 436-451.e11. DOI: https://doi.org/10.1016/j.neuron.2021.10.032. PMID: https://www.ncbi.nlm.nih.gov/pubmed/34793693.

44. N. Kang; X. Han; Z. Li; T. Liu; X. Mi; Y. Li; X. Guo; D. Han; N. Yang Rapamycin Affects the Hippocampal SNARE Complex to Alleviate Cognitive Dysfunction Induced by Surgery in Aged Rats., 2023, 13, 598. DOI: https://doi.org/10.3390/brainsci13040598. PMID: https://www.ncbi.nlm.nih.gov/pubmed/37190563.

45. D. Sepúlveda; M. Cisternas-Olmedo; J. Arcos; M. Nassif; R.L. Vidal Contribution of Autophagy-Lysosomal Pathway in the Exosomal Secretion of Alpha-Synuclein and Its Impact in the Progression of Parkinson’s Disease., 2022, 15, 805087. DOI: https://doi.org/10.3389/fnmol.2022.805087. PMID: https://www.ncbi.nlm.nih.gov/pubmed/35250476.

46. A. Friedman; D. Homma; B. Bloem; L.G. Gibb; K. Amemori; D. Hu; S. Delcasso; T.F. Truong; J. Yang; A.S. Hood et al. Chronic Stress Alters Striosome-Circuit Dynamics, Leading to Aberrant Decision-Making., 2017, 171,pp. 1191-1205.e28. DOI: https://doi.org/10.1016/j.cell.2017.10.017. PMID: https://www.ncbi.nlm.nih.gov/pubmed/29149606.

47. C.R. Fields; N. Bengoa-Vergniory; R. Wade-Martins Targeting Alpha-Synuclein as a Therapy for Parkinson’s Disease., 2019, 12, 299. DOI: https://doi.org/10.3389/fnmol.2019.00299. PMID: https://www.ncbi.nlm.nih.gov/pubmed/31866823.

48. N. Bengoa-Vergniory; R.F. Roberts; R. Wade-Martins; J. Alegre-Abarrategui Alpha-Synuclein Oligomers: A New Hope., 2017, 134,pp. 819-838. DOI: https://doi.org/10.1007/s00401-017-1755-1. PMID: https://www.ncbi.nlm.nih.gov/pubmed/28803412.

49. J. Alegre-Abarrategui; K.R. Brimblecombe; R.F. Roberts; E. Velentza-Almpani; B.S. Tilley; N. Bengoa-Vergniory; C. Proukakis Selective Vulnerability in a-Synucleinopathies., 2019, 138,pp. 681-704. DOI: https://doi.org/10.1007/s00401-019-02010-2.

50. D. Lindqvist; E. Kaufman; L. Brundin; S. Hall; Y. Surova; O. Hansson Non-Motor Symptoms in Patients with Parkinson’s Disease—Correlations with Inflammatory Cytokines in Serum., 2012, 7, e47387. DOI: https://doi.org/10.1371/journal.pone.0047387.

51. R. Kim; H. Kim; J.H. Shin; C.Y. Lee; S.H. Jeon; B. Jeon Serum Inflammatory Markers and Progression of Nonmotor Symptoms in Early Parkinson’s Disease., 2022, 37,pp. 1535-1541. DOI: https://doi.org/10.1002/mds.29056. PMID: https://www.ncbi.nlm.nih.gov/pubmed/35596676.

52. S.S. Chavan; V.A. Pavlov; K.J. Tracey Mechanisms and Therapeutic Relevance of Neuro-Immune Communication., 2017, 46,pp. 927-942. DOI: https://doi.org/10.1016/j.immuni.2017.06.008. PMID: https://www.ncbi.nlm.nih.gov/pubmed/28636960.

53. K.J. Doorn; T. Moors; B. Drukarch; W.D. van de Berg; P.J. Lucassen; A.-M. van Dam Microglial Phenotypes and Toll-like Receptor 2 in the Substantia Nigra and Hippocampus of Incidental Lewy Body Disease Cases and Parkinson’s Disease Patients., 2014, 2,p. 90. DOI: https://doi.org/10.1186/s40478-014-0090-1.

54. J. Drouin-Ouellet Mitochondrial Complex I Deficiency and Parkinson Disease., 2023, 24,p. 193. DOI: https://doi.org/10.1038/s41583-023-00676-y. PMID: https://www.ncbi.nlm.nih.gov/pubmed/36697768.

55. X. Li; L. Xue; J. Sun; Y. Sun; A. Xie Single Nucleotide Polymorphisms in the Toll-like Receptor 2 (TLR2) Gene Are Associated with Sporadic Parkinson’s Disease in the North-Eastern Han Chinese Population., 2017, 656,pp. 72-76. DOI: https://doi.org/10.1016/j.neulet.2017.07.014. PMID: https://www.ncbi.nlm.nih.gov/pubmed/28729076.

56. G.M. Mazzotta; N. Ceccato; C. Conte Synucleinopathies Take Their Toll: Are TLRs a Way to Go?., 2023, 12, 1231. DOI: https://doi.org/10.3390/cells12091231.

57. V. Kumar Toll-like Receptors in the Pathogenesis of Neuroinflammation., 2019, 332,pp. 16-30. DOI: https://doi.org/10.1016/j.jneuroim.2019.03.012. PMID: https://www.ncbi.nlm.nih.gov/pubmed/30928868.

58. A. Kouli; C.B. Horne; C.H. Williams-Gray Toll-like Receptors and Their Therapeutic Potential in Parkinson’s Disease and a-Synucleinopathies., 2019, 81,pp. 41-51. DOI: https://doi.org/10.1016/j.bbi.2019.06.042. PMID: https://www.ncbi.nlm.nih.gov/pubmed/31271873.

59. A. Heidari; N. Yazdanpanah; N. Rezaei The Role of Toll-like Receptors and Neuroinflammation in Parkinson’s Disease., 2022, 19,p. 135. DOI: https://doi.org/10.1186/s12974-022-02496-w.

60. J.M. Yoo; Y. Lin; Y. Heo; Y.-H. Lee Polymorphism in Alpha-Synuclein Oligomers and Its Implications in Toxicity under Disease Conditions., 2022, 9, 959425. DOI: https://doi.org/10.3389/fmolb.2022.959425.

61. J. Ma; J. Gao; J. Wang; A. Xie Prion-like Mechanisms in Parkinson’s Disease., 2019, 13, 552. DOI: https://doi.org/10.3389/fnins.2019.00552. PMID: https://www.ncbi.nlm.nih.gov/pubmed/31275093.

62. A. Jan; N.P. Gonçalves; C.B. Vaegter; P.H. Jensen; N. Ferreira The Prion-like Spreading of Alpha-Synuclein in Parkinson’s Disease: Update on Models and Hypotheses., 2021, 22, 8338. DOI: https://doi.org/10.3390/ijms22158338.

63. P. Garcia; W. Jürgens-Wemheuer; O. Uriarte Huarte; A. Michelucci; A. Masuch; S. Brioschi; A. Weihofen; E. Koncina; D. Coowar; T. Heurtaux et al. Neurodegeneration and Neuroinflammation Are Linked, but Independent of Alpha-synuclein Inclusions, in a Seeding/Spreading Mouse Model of Parkinson’s Disease., 2022, 70,pp. 935-960. DOI: https://doi.org/10.1002/glia.24149. PMID: https://www.ncbi.nlm.nih.gov/pubmed/35092321.

64. Y. Dening; T. Straßl; V. Ruf; P. Dirscherl; A. Chovsepian; A. Stievenard; A. Khairnar; F. Schmidt; F. Giesert; J. Herms et al. Toxicity of Extracellular Alpha-Synuclein Is Independent of Intracellular Alpha-Synuclein., 2022, 12,p. 21951. DOI: https://doi.org/10.1038/s41598-022-25790-2. PMID: https://www.ncbi.nlm.nih.gov/pubmed/36535974.

65. E. Vasili; A. Dominguez-Meijide; M. Flores-León; M. Al-Azzani; A. Kanellidi; R. Melki; L. Stefanis; T.F. Outeiro Endogenous Levels of Alpha-Synuclein Modulate Seeding and Aggregation in Cultured Cells., 2022, 59,pp. 1273-1284. DOI: https://doi.org/10.1007/s12035-021-02713-2.

66. R.J. Karpowicz; J.Q. Trojanowski; V.M.-Y. Lee Transmission of a-Synuclein Seeds in Neurodegenerative Disease: Recent Developments., 2019, 99,pp. 971-981. DOI: https://doi.org/10.1038/s41374-019-0195-z.

67. L. Holper; D. Ben-Shachar; J. Mann Multivariate Meta-Analyses of Mitochondrial Complex I and IV in Major Depressive Disorder, Bipolar Disorder, Schizophrenia, Alzheimer Disease, and Parkinson Disease., 2019, 44,pp. 837-849. DOI: https://doi.org/10.1038/s41386-018-0090-0.

68. V. Tapias; X. Hu; K.C. Luk; L.H. Sanders; V.M. Lee; J.T. Greenamyre Synthetic Alpha-Synuclein Fibrils Cause Mitochondrial Impairment and Selective Dopamine Neurodegeneration in Part via INOS-Mediated Nitric Oxide Production., 2017, 74,pp. 2851-2874. DOI: https://doi.org/10.1007/s00018-017-2541-x.

69. K. Yamamoto; Y. Izumi; M. Arif*cku; T. Kume; H. Sawada a-Synuclein Oligomers Mediate the Aberrant Form of Spike-Induced Calcium Release from IP3 Receptor., 2019, 9,p. 15977. DOI: https://doi.org/10.1038/s41598-019-52135-3.

70. E. Leandrou; I. Chalatsa; D. Anagnostou; C. Machalia; M. Semitekolou; V. Filippa; M. Makridakis; A. Vlahou; E. Anastasiadou; K. Vekrellis et al. a-Synuclein Oligomers Potentiate Neuroinflammatory NF-?B Activity and Induce Cav3.2 Calcium Signaling in Astrocytes., 2024, 13,p. 11. DOI: https://doi.org/10.1186/s40035-024-00401-4.

71. L. Shvachiy; V. Geraldes; T.F. Outeiro Uncovering the Molecular Link Between Lead Toxicity and Parkinson’s Disease., 2023, 39,pp. 321-335. DOI: https://doi.org/10.1089/ars.2022.0076. PMID: https://www.ncbi.nlm.nih.gov/pubmed/36641635.

72. R. Di Maio; P.J. Barrett; E.K. Hoffman; C.W. Barrett; A. Zharikov; A. Borah; X. Hu; J. McCoy; C.T. Chu; E.A. Burton et al. a-Synuclein Binds to TOM20 and Inhibits Mitochondrial Protein Import in Parkinson’s Disease., 2016, 8,p. 342ra78. DOI: https://doi.org/10.1126/scitranslmed.aaf3634. PMID: https://www.ncbi.nlm.nih.gov/pubmed/27280685.

73. L. Shan; H.J. Heusinkveld; K.C. Paul; S. Hughes; S.K.L. Darweesh; B.R. Bloem; J.R. Homberg Towards Improved Screening of Toxins for Parkinson’s Risk., 2023, 9,p. 169. DOI: https://doi.org/10.1038/s41531-023-00615-9. PMID: https://www.ncbi.nlm.nih.gov/pubmed/38114496.

74. S.J. Won; R. Fong; N. Butler; J. Sanchez; Y. Zhang; C. Wong; O. Tambou Nzoutchoum; A. Huynh; J. Pan; R.A. Swanson Neuronal Oxidative Stress Promotes a-Synuclein Aggregation In Vivo., 2022, 11, 2466. DOI: https://doi.org/10.3390/antiox11122466. PMID: https://www.ncbi.nlm.nih.gov/pubmed/36552674.

75. L. Almandoz-Gil; H. Welander; E. Ihse; P.E. Khoonsari; S. Musunuri; C. Lendel; J. Sigvardson; M. Karlsson; M. Ingelsson; K. Kultima et al. Low Molar Excess of 4-Oxo-2-Nonenal and 4-Hydroxy-2-Nonenal Promote Oligomerization of Alpha-Synuclein through Different Pathways., 2017, 110,pp. 421-431. DOI: https://doi.org/10.1016/j.freeradbiomed.2017.07.004. PMID: https://www.ncbi.nlm.nih.gov/pubmed/28690195.

76. M. Sardar Sinha; A.M. Villamil Giraldo; K. Öllinger; M. Hallbeck; L. Civitelli Lipid Vesicles Affect the Aggregation of 4-Hydroxy-2-Nonenal-Modified a-Synuclein Oligomers., 2018, 1864,pp. 3060-3068. DOI: https://doi.org/10.1016/j.bbadis.2018.06.020. PMID: https://www.ncbi.nlm.nih.gov/pubmed/29960040.

77. C. Sahin; E.C. Østerlund; N. Österlund; J. Costeira-Paulo; J.N. Pedersen; G. Christiansen; J. Nielsen; A.L. Grønnemose; S.K. Amstrup; M.K. Tiwari et al. Structural Basis for Dityrosine-Mediated Inhibition of a-Synuclein Fibrillization., 2022, 144,pp. 11949-11954. DOI: https://doi.org/10.1021/jacs.2c03607. PMID: https://www.ncbi.nlm.nih.gov/pubmed/35749730.

78. Y.K. Al-Hilaly; L. Biasetti; B.J.F. Blakeman; S.J. Pollack; S. Zibaee; A. Abdul-Sada; J.R. Thorpe; W.-F. Xue; L.C. Serpell The Involvement of Dityrosine Crosslinking in a-Synuclein Assembly and Deposition in Lewy Bodies in Parkinson’s Disease., 2016, 6,p. 39171. DOI: https://doi.org/10.1038/srep39171. PMID: https://www.ncbi.nlm.nih.gov/pubmed/27982082.

79. M.M. Wördehoff; H. Shaykhalishahi; L. Groß; L. Gremer; M. Stoldt; A.K. Buell; D. Willbold; W. Hoyer Opposed Effects of Dityrosine Formation in Soluble and Aggregated a-Synuclein on Fibril Growth., 2017, 429,pp. 3018-3030. DOI: https://doi.org/10.1016/j.jmb.2017.09.005.

80. A.K. Buell; C. Galvagnion; R. Gaspar; E. Sparr; M. Vendruscolo; T.P.J. Knowles; S. Linse; C.M. Dobson Solution Conditions Determine the Relative Importance of Nucleation and Growth Processes in a-Synuclein Aggregation., 2014, 111,pp. 7671-7676. DOI: https://doi.org/10.1073/pnas.1315346111.

81. N. Mehta; P. Marwah; D. Njus Are Proteinopathy and Oxidative Stress Two Sides of the Same Coin?., 2019, 8, 59. DOI: https://doi.org/10.3390/cells8010059. PMID: https://www.ncbi.nlm.nih.gov/pubmed/30654525.

82. M.G. Stykel; K.M. Humphries; E. Kamski-Hennekam; B. Buchner-Duby; N. Porte-Trachsel; T. Ryan; C.L. Coackley; V.V. Bamm; G. Harauz; S.D. Ryan a-Synuclein Mutation Impairs Processing of Endomembrane Compartments and Promotes Exocytosis and Seeding of a-Synuclein Pathology., 2021, 35,p. 109099. DOI: https://doi.org/10.1016/j.celrep.2021.109099.

83. R. Kumar; D.K. Jangir; G. Verma; S. Shekhar; P. Hanpude; S. Kumar; R. Kumari; N. Singh; N. Sarovar Bhavesh; N. Ranjan Jana et al. S-Nitrosylation of UCHL1 Induces Its Structural Instability and Promotes a-Synuclein Aggregation., 2017, 7,p. 44558. DOI: https://doi.org/10.1038/srep44558. PMID: https://www.ncbi.nlm.nih.gov/pubmed/28300150.

84. A. Grimm; A. Eckert Brain Aging and Neurodegeneration: From a Mitochondrial Point of View., 2017, 143,pp. 418-431. DOI: https://doi.org/10.1111/jnc.14037. PMID: https://www.ncbi.nlm.nih.gov/pubmed/28397282.

85. J.-S. Park; B. Koentjoro; R.L. Davis; C.M. Sue Loss of ATP13A2 Impairs Glycolytic Function in Kufor-Rakeb Syndrome Patient-Derived Cell Models., 2016, 27,pp. 67-73. DOI: https://doi.org/10.1016/j.parkreldis.2016.03.018. PMID: https://www.ncbi.nlm.nih.gov/pubmed/27039055.

86. G.K. Ganjam; K. Bolte; L.A. Matschke; S. Neitemeier; A.M. Dolga; M. Höllerhage; G.U. Höglinger; A. Adamczyk; N. Decher; W.H. Oertel et al. Mitochondrial Damage by a-Synuclein Causes Cell Death in Human Dopaminergic Neurons., 2019, 10,p. 865. DOI: https://doi.org/10.1038/s41419-019-2091-2. PMID: https://www.ncbi.nlm.nih.gov/pubmed/31727879.

87. P.P. Michel; E.C. Hirsch; S. Hunot Understanding Dopaminergic Cell Death Pathways in Parkinson Disease., 2016, 90,pp. 675-691. DOI: https://doi.org/10.1016/j.neuron.2016.03.038. PMID: https://www.ncbi.nlm.nih.gov/pubmed/27196972.

88. Š. Lehtonen; T.-M. Sonninen; S. Wojciechowski; G. Goldsteins; J. Koistinaho Dysfunction of Cellular Proteostasis in Parkinson’s Disease., 2019, 13, 457. DOI: https://doi.org/10.3389/fnins.2019.00457. PMID: https://www.ncbi.nlm.nih.gov/pubmed/31133790.

89. M. Gómez-Benito; N. Granado; P. García-Sanz; A. Michel; M. Dumoulin; R. Moratalla Modeling Parkinson’s Disease with the Alpha-Synuclein Protein., 2020, 11, 356. DOI: https://doi.org/10.3389/fphar.2020.00356.

90. M.H.R. Ludtmann; P.R. Angelova; M.H. Horrocks; M.L. Choi; M. Rodrigues; A.Y. Baev; A.V. Berezhnov; Z. Yao; D. Little; B. Banushi et al. a-Synuclein Oligomers Interact with ATP Synthase and Open the Permeability Transition Pore in Parkinson’s Disease., 2018, 9,p. 2293. DOI: https://doi.org/10.1038/s41467-018-04422-2.

91. X.-L. Wang; S.-T. Feng; Y.-T. Wang; Y.-H. Yuan; Z.-P. Li; N.-H. Chen; Z.-Z. Wang; Y. Zhang Mitophagy, a Form of Selective Autophagy, Plays an Essential Role in Mitochondrial Dynamics of Parkinson’s Disease., 2022, 42,pp. 1321-1339. DOI: https://doi.org/10.1007/s10571-021-01039-w. PMID: https://www.ncbi.nlm.nih.gov/pubmed/33528716.

92. D.Y. Kim; J.Y. Shin; J.E. Lee; H.N. Kim; S.J. Chung; H.S. Yoo; S.J. Kim; H.J. Cho; E.-J. Lee; S.J. Nam et al. A Selective ER-Phagy Exerts Neuroprotective Effects via Modulation of a-Synuclein Clearance in Parkinsonian Models., 2023, 120,p. e2221929120. DOI: https://doi.org/10.1073/pnas.2221929120. PMID: https://www.ncbi.nlm.nih.gov/pubmed/37669380.

93. S. Bido; S. Muggeo; L. Massimino; M.J. Marzi; S.G. Giannelli; E. Melacini; M. Nannoni; D. Gambarè; E. Bellini; G. Ordazzo et al. Microglia-Specific Overexpression of a-Synuclein Leads to Severe Dopaminergic Neurodegeneration by Phagocytic Exhaustion and Oxidative Toxicity., 2021, 12,p. 6237. DOI: https://doi.org/10.1038/s41467-021-26519-x. PMID: https://www.ncbi.nlm.nih.gov/pubmed/34716339.

94. W. Haenseler; F. Zambon; H. Lee; J. Vowles; F. Rinaldi; G. Duggal; H. Houlden; K. Gwinn; S. Wray; K.C. Luk et al. Excess a-Synuclein Compromises Phagocytosis in IPSC-Derived Macrophages., 2017, 7,p. 9003. DOI: https://doi.org/10.1038/s41598-017-09362-3. PMID: https://www.ncbi.nlm.nih.gov/pubmed/28827786.

95. C. Peng; J.Q. Trojanowski; V.M.-Y. Lee Protein Transmission in Neurodegenerative Disease., 2020, 16,pp. 199-212. DOI: https://doi.org/10.1038/s41582-020-0333-7.

96. S. Isik; B. Yeman Kiyak; R. Akbayir; R. Seyhali; T. Arpaci Microglia Mediated Neuroinflammation in Parkinson’s Disease., 2023, 12, 1012. DOI: https://doi.org/10.3390/cells12071012.

97. A.F. Pike; I. Szabò; R. Veerhuis; L. Bubacco The Potential Convergence of NLRP3 Inflammasome, Potassium, and Dopamine Mechanisms in Parkinson’s Disease., 2022, 8,p. 32. DOI: https://doi.org/10.1038/s41531-022-00293-z. PMID: https://www.ncbi.nlm.nih.gov/pubmed/35332154.

98. Y. Murakami; T. Ishibashi; E. Tomita; Y. Imamura; T. Tashiro; K. Watcharanurak; M. Nishikawa; Y. Takahashi; Y. Takakura; S. Mitani et al. Depressive Symptoms as a Side Effect of Interferon-a Therapy Induced by Induction of Indoleamine 2,3-Dioxygenase 1., 2016, 6,p. 29920. DOI: https://doi.org/10.1038/srep29920.

99. Y. Zhang; H. Shi; G. Yang; Y. Yang; W. Li; M. Song; M. Shao; X. Su; L. Lv Associations between Expression of Indoleamine 2, 3-Dioxygenase Enzyme and Inflammatory Cytokines in Patients with First-Episode Drug-Naive Schizophrenia., 2021, 11,p. 595. DOI: https://doi.org/10.1038/s41398-021-01688-x.

100. R.K. Sodhi; Y. Bansal; R. Singh; P. Saroj; R. Bhandari; B. Kumar; A. Kuhad IDO-1 Inhibition Protects against Neuroinflammation, Oxidative Stress and Mitochondrial Dysfunction in 6-OHDA Induced Murine Model of Parkinson’s Disease., 2021, 84,pp. 184-197. DOI: https://doi.org/10.1016/j.neuro.2021.03.009.

101. B.d.S. Oliveira; E.C.d.B. Toscano; L.K.S. Abreu; H.d.B. Fernandes; R.F. Amorim; R.N. Ferreira; C.A. Machado; B.C. Carvalho; M.C.M. da Silva; A.C.P. de Oliveira et al. Nigrostriatal Inflammation Is Associated with Nonmotor Symptoms in an Experimental Model of Prodromal Parkinson’s Disease., 2024, 549,pp. 65-75. DOI: https://doi.org/10.1016/j.neuroscience.2024.05.011. PMID: https://www.ncbi.nlm.nih.gov/pubmed/38750924.

102. F. Rahmani; A. Saghazadeh; M. Rahmani; A.L. Teixeira; N. Rezaei; V. Aghamollaii; H.E. Ardebili Plasma Levels of Brain-Derived Neurotrophic Factor in Patients with Parkinson Disease: A Systematic Review and Meta-Analysis., 2019, 1704,pp. 127-136. DOI: https://doi.org/10.1016/j.brainres.2018.10.006. PMID: https://www.ncbi.nlm.nih.gov/pubmed/30296429.

103. A.M. Hemmerle; J.W. Dickerson; J.P. Herman; K.B. Seroogy Stress Exacerbates Experimental Parkinson’s Disease., 2014, 19,pp. 638-640. DOI: https://doi.org/10.1038/mp.2013.108. PMID: https://www.ncbi.nlm.nih.gov/pubmed/23999522.

104. Y.-Y. Tan; P. Jenner; S.-D. Chen Monoamine Oxidase-B Inhibitors for the Treatment of Parkinson’s Disease: Past, Present, and Future., 2022, 12,pp. 477-493. DOI: https://doi.org/10.3233/JPD-212976. PMID: https://www.ncbi.nlm.nih.gov/pubmed/34957948.

105. Y. Fu; Y. He; K. Phan; S. Bhatia; R. Pickford; P. Wu; N. Dzamko; G.M. Halliday; W.S. Kim Increased Unsaturated Lipids Underlie Lipid Peroxidation in Synucleinopathy Brain., 2022, 10,p. 165. DOI: https://doi.org/10.1186/s40478-022-01469-7. PMID: https://www.ncbi.nlm.nih.gov/pubmed/36376990.

106. Y. Li; T. Zhao; J. Li; M. Xia; Y. Li; X. Wang; C. Liu; T. Zheng; R. Chen; D. Kan et al. Oxidative Stress and 4-Hydroxy-2-Nonenal (4-HNE): Implications in the Pathogenesis and Treatment of Aging-Related Diseases., 2022, 2022,p. 2233906. DOI: https://doi.org/10.1155/2022/2233906. PMID: https://www.ncbi.nlm.nih.gov/pubmed/35411309.

107. G. De Franceschi; E. Frare; L. Bubacco; S. Mammi; A. Fontana; P.P. de Laureto Molecular Insights into the Interaction between a-Synuclein and Docosahexaenoic Acid., 2009, 394,pp. 94-107. DOI: https://doi.org/10.1016/j.jmb.2009.09.008. PMID: https://www.ncbi.nlm.nih.gov/pubmed/19747490.

108. G. De Franceschi; E. Frare; M. Pivato; A. Relini; A. Penco; E. Greggio; L. Bubacco; A. Fontana; P.P. de Laureto Structural and Morphological Characterization of Aggregated Species of a-Synuclein Induced by Docosahexaenoic Acid., 2011, 286,pp. 22262-22274. DOI: https://doi.org/10.1074/jbc.M110.202937.

109. Y. Cai; C. Lendel; L. Österlund; A. Kasrayan; L. Lannfelt; M. Ingelsson; F. Nikolajeff; M. Karlsson; J. Bergström Changes in Secondary Structure of a-Synuclein during Oligomerization Induced by Reactive Aldehydes., 2015, 464,pp. 336-341. DOI: https://doi.org/10.1016/j.bbrc.2015.06.154.

110. M.F. Beal; J. Chiluwal; N.Y. Calingasan; G.L. Milne; M.S. Shchepinov; V. Tapias Isotope-Reinforced Polyunsaturated Fatty Acids Improve Parkinson’s Disease-like Phenotype in Rats Overexpressing a-Synuclein., 2020, 8,p. 220. DOI: https://doi.org/10.1186/s40478-020-01090-6.

111. C. Galvagnion The Role of Lipids Interacting with a-Synuclein in the Pathogenesis of Parkinson’s Disease., 2017, 7,pp. 433-450. DOI: https://doi.org/10.3233/JPD-171103. PMID: https://www.ncbi.nlm.nih.gov/pubmed/28671142.

112. S.R. Bonam; C. Tranchant; S. Muller Autophagy-Lysosomal Pathway as Potential Therapeutic Target in Parkinson’s Disease., 2021, 10, 3547. DOI: https://doi.org/10.3390/cells10123547. PMID: https://www.ncbi.nlm.nih.gov/pubmed/34944054.

113. I. Kinnart; L. Manders; T. Heyninck; D. Imberechts; R. Praschberger; N. Schoovaerts; C. Verfaillie; P. Verstreken; W. Vandenberghe Elevated a-Synuclein Levels Inhibit Mitophagic Flux., 2024, 10,p. 80. DOI: https://doi.org/10.1038/s41531-024-00696-0.

114. M.G. Stykel; S.D. Ryan Nitrosative Stress in Parkinson’s Disease., 2022, 8,p. 104. DOI: https://doi.org/10.1038/s41531-022-00370-3. PMID: https://www.ncbi.nlm.nih.gov/pubmed/35953517.

115. N. Bisi; L. Feni; K. Peqini; H. Pérez-Peña; S. Ongeri; S. Pieraccini; S. Pellegrino a-Synuclein: An All-Inclusive Trip Around Its Structure, Influencing Factors and Applied Techniques., 2021, 9, 666585. DOI: https://doi.org/10.3389/fchem.2021.666585. PMID: https://www.ncbi.nlm.nih.gov/pubmed/34307295.

116. N. Lorenzen; L. Lemminger; J.N. Pedersen; S.B. Nielsen; D.E. Otzen The N-Terminus of a-Synuclein Is Essential for Both Monomeric and Oligomeric Interactions with Membranes., 2014, 588,pp. 497-502. DOI: https://doi.org/10.1016/j.febslet.2013.12.015. PMID: https://www.ncbi.nlm.nih.gov/pubmed/24374342.

117. D.L. Abeyawardhane; R.D. Fernández; C.J. Murgas; D.R. Heitger; A.K. Forney; M.K. Crozier; H.R. Lucas Iron Redox Chemistry Promotes Antiparallel Oligomerization of a-Synuclein., 2018, 140,pp. 5028-5032. DOI: https://doi.org/10.1021/jacs.8b02013. PMID: https://www.ncbi.nlm.nih.gov/pubmed/29608844.

118. C.J. Chang Bioinorganic Life and Neural Activity: Toward a Chemistry of Consciousness?., 2017, 50,pp. 535-538. DOI: https://doi.org/10.1021/acs.accounts.6b00531. PMID: https://www.ncbi.nlm.nih.gov/pubmed/28945425.

119. M. Bisaglia; L. Bubacco Copper Ions and Parkinson’s Disease: Why Is Homeostasis So Relevant?., 2020, 10, 195. DOI: https://doi.org/10.3390/biom10020195. PMID: https://www.ncbi.nlm.nih.gov/pubmed/32013126.

120. C. Exley; M.J. Mold Aluminium in Human Brain Tissue: How Much Is Too Much?., 2019, 24,pp. 1279-1282. DOI: https://doi.org/10.1007/s00775-019-01710-0.

121. M.J. Mold; C. Exley Aluminium Co-Localises with Biondi Ring Tangles in Parkinson’s Disease and Epilepsy., 2022, 12,p. 1465. DOI: https://doi.org/10.1038/s41598-022-05627-8.

122. A. Maillet; E. Météreau; L. Tremblay; E. Favre; H. Klinger; E. Lhommée; D. Le Bars; A. Castrioto; S. Prange; V. Sgambato et al. Serotonergic and Dopaminergic Lesions Underlying Parkinsonian Neuropsychiatric Signs., 2021, 36,pp. 2888-2900. DOI: https://doi.org/10.1002/mds.28722. PMID: https://www.ncbi.nlm.nih.gov/pubmed/34494685.

123. S. Hayley; F. Vahid-Ansari; H. Sun; P.R. Albert Mood Disturbances in Parkinson’s Disease: From Prodromal Origins to Application of Animal Models., 2023, 181, 106115. DOI: https://doi.org/10.1016/j.nbd.2023.106115.

124. Y. Bang; J. Lim; H.J. Choi Recent Advances in the Pathology of Prodromal Non-Motor Symptoms Olfactory Deficit and Depression in Parkinson’s Disease: Clues to Early Diagnosis and Effective Treatment., 2021, 44,pp. 588-604. DOI: https://doi.org/10.1007/s12272-021-01337-3.

125. F. Stocchi; A. Antonini; P. Barone; G. Bellelli; A. fa*giolini; L. Ferini Strambi; S. Sorbi; A. Padovani Exploring Depression in Parkinson’s Disease: An Italian Delphi Consensus on Phenomenology, Diagnosis, and Management., 2023, 44,pp. 3123-3131. DOI: https://doi.org/10.1007/s10072-023-06740-w.

126. M. Banwinkler; H. Theis; S. Prange; T. van Eimeren Imaging the Limbic System in Parkinson’s Disease—A Review of Limbic Pathology and Clinical Symptoms., 2022, 12, 1248. DOI: https://doi.org/10.3390/brainsci12091248.

127. S. Prange; E. Metereau; A. Maillet; E. Lhommée; H. Klinger; P. Pelissier; D. Ibarrola; R.A. Heckemann; A. Castrioto; L. Tremblay et al. Early Limbic Microstructural Alterations in Apathy and Depression in de Novo Parkinson’s Disease., 2019, 34,pp. 1644-1654. DOI: https://doi.org/10.1002/mds.27793. PMID: https://www.ncbi.nlm.nih.gov/pubmed/31309609.

128. A.M. Buchanan; S. Mena; I. Choukari; A. Vasa; J.N. Crawford; J. Fadel; N. Maxwell; L. Reagan; A. Cruikshank; J. Best et al. Serotonin as a Biomarker of Toxin-Induced Parkinsonism., 2024, 30,p. 33. DOI: https://doi.org/10.1186/s10020-023-00773-9. PMID: https://www.ncbi.nlm.nih.gov/pubmed/38429661.

129. Q. Tong; L. Zhang; Y. Yuan; S. Jiang; R. Zhang; Q. Xu; J. Ding; D. Li; X. Zhou; K. Zhang Reduced Plasma Serotonin and 5-Hydroxyindoleacetic Acid Levels in Parkinson’s Disease Are Associated with Nonmotor Symptoms., 2015, 21,pp. 882-887. DOI: https://doi.org/10.1016/j.parkreldis.2015.05.016.

130. T. Du; G. Li; H. Luo; Y. Pan; Q. Xu; K. Ma Hippocampal Alpha-Synuclein Mediates Depressive-like Behaviors., 2021, 95,pp. 226-237. DOI: https://doi.org/10.1016/j.bbi.2021.03.020.

131. K. Kamagata; T. Nakatsuka; R. Sakakibara; Y. Tsuyusaki; T. Takamura; K. Sato; M. Suzuki; M. Hori; K.K. Kumamaru; T. Inaoka et al. Diagnostic Imaging of Dementia with Lewy Bodies by Susceptibility-Weighted Imaging of Nigrosomes versus Striatal Dopamine Transporter Single-Photon Emission Computed Tomography: A Retrospective Observational Study., 2017, 59,pp. 89-98. DOI: https://doi.org/10.1007/s00234-016-1773-z. PMID: https://www.ncbi.nlm.nih.gov/pubmed/28035426.

132. L.M. Butkovich; M.C. Houser; T. Chalermpalanupap; K.A. Porter-Stransky; A.F. Iannitelli; J.S. Boles; G.M. Lloyd; A.S. Coomes; L.N. Eidson; M.E. De Sousa Rodrigues et al. Transgenic Mice Expressing Human a-Synuclein in Noradrenergic Neurons Develop Locus Ceruleus Pathology and Nonmotor Features of Parkinson’s Disease., 2020, 40,pp. 7559-7576. DOI: https://doi.org/10.1523/JNEUROSCI.1468-19.2020. PMID: https://www.ncbi.nlm.nih.gov/pubmed/32868457.

133. L. Miquel-Rio; D. Alarcón-Arís; M. Torres-López; V. Cóppola-Segovia; R. Pavia-Collado; V. Paz; E. Ruiz-Bronchal; L. Campa; C. Casal; A. Montefeltro et al. Human a-Synuclein Overexpression in Mouse Serotonin Neurons Triggers a Depressive-like Phenotype. Rescue by Oligonucleotide Therapy., 2022, 12,p. 79. DOI: https://doi.org/10.1038/s41398-022-01842-z. PMID: https://www.ncbi.nlm.nih.gov/pubmed/35210396.

134. N.N.W. Dissanayaka; E. White; J.D. O’Sullivan; R. Marsh; P.A. Silburn; D.A. Copland; G.D. Mellick; G.J. Byrne Characteristics and Treatment of Anxiety Disorders in Parkinson’s Disease., 2015, 2,pp. 155-162. DOI: https://doi.org/10.1002/mdc3.12157. PMID: https://www.ncbi.nlm.nih.gov/pubmed/30363816.

135. E.M. Khedr; A.A. Abdelrahman; Y. Elserogy; A.F. Zaki; A. Gamea Depression and Anxiety among Patients with Parkinson’s Disease: Frequency, Risk Factors, and Impact on Quality of Life., 2020, 56,p. 116. DOI: https://doi.org/10.1186/s41983-020-00253-5.

136. T.T. Lai; B. Gericke; M. Feja; M. Conoscenti; M. Zelikowsky; F. Richter Anxiety in Synucleinopathies: Neuronal Circuitry, Underlying Pathomechanisms and Current Therapeutic Strategies., 2023, 9,p. 97. DOI: https://doi.org/10.1038/s41531-023-00547-4. PMID: https://www.ncbi.nlm.nih.gov/pubmed/37349373.

137. K. Kalinderi; V. Papaliagkas; L. Fidani Current Genetic Data on Depression and Anxiety in Parkinson’s Disease Patients., 2024, 118,p. 105922. DOI: https://doi.org/10.1016/j.parkreldis.2023.105922. PMID: https://www.ncbi.nlm.nih.gov/pubmed/37935601.

138. W. Wang; N. Song; F. Jia; T. Tang; W. Bao; C. Zuo; J. Xie; H. Jiang Genomic DNA Levels of Mutant Alpha-Synuclein Correlate with Non-Motor Symptoms in an A53T Parkinson’s Disease Mouse Model., 2018, 114,pp. 71-79. DOI: https://doi.org/10.1016/j.neuint.2018.01.006. PMID: https://www.ncbi.nlm.nih.gov/pubmed/29355568.

139. J. Deusser; S. Schmidt; B. Ettle; S. Plötz; S. Huber; C.P. Müller; E. Masliah; J. Winkler; Z. Kohl Serotonergic Dysfunction in the A53T Alpha-Synuclein Mouse Model of Parkinson’s Disease., 2015, 135,pp. 589-597. DOI: https://doi.org/10.1111/jnc.13253.

140. L.E. Stoyka; A.E. Arrant; D.R. Thrasher; D.L. Russell; J. Freire; C.L. Mahoney; A. Narayanan; A.G. Dib; D.G. Standaert; L.A. Volpicelli-Daley Behavioral Defects Associated with Amygdala and Cortical Dysfunction in Mice with Seeded a-Synuclein Inclusions., 2020, 134, 104708. DOI: https://doi.org/10.1016/j.nbd.2019.104708.

141. N. Uemura; J. Ueda; T. Yoshihara; M. Ikuno; M.T. Uemura; H. Yamakado; M. Asano; J.Q. Trojanowski; R. Takahashi a-Synuclein Spread from Olfactory Bulb Causes Hyposmia, Anxiety, and Memory Loss in BAC-SNCA Mice., 2021, 36,pp. 2036-2047. DOI: https://doi.org/10.1002/mds.28512. PMID: https://www.ncbi.nlm.nih.gov/pubmed/33547846.

142. H. Fu; A. Possenti; R. Freer; Y. Nakano; N.C. Hernandez Villegas; M. Tang; P.V.M. Cauhy; B.A. Lassus; S. Chen; S.L. Fowler et al. A Tau Homeostasis Signature Is Linked with the Cellular and Regional Vulnerability of Excitatory Neurons to Tau Pathology., 2019, 22,pp. 47-56. DOI: https://doi.org/10.1038/s41593-018-0298-7. PMID: https://www.ncbi.nlm.nih.gov/pubmed/30559469.

143. G.S. Virdi; M.L. Choi; J.R. Evans; Z. Yao; D. Athauda; S. Strohbuecker; R.S. Nirujogi; A.I. Wernick; N. Pelegrina-Hidalgo; C. Leighton et al. Protein Aggregation and Calcium Dysregulation Are Hallmarks of Familial Parkinson’s Disease in Midbrain Dopaminergic Neurons., 2022, 8,p. 162. DOI: https://doi.org/10.1038/s41531-022-00423-7. PMID: https://www.ncbi.nlm.nih.gov/pubmed/36424392.

144. A. Maillet; P. Krack; E. Lhommée; E. Météreau; H. Klinger; E. Favre; D. Le Bars; E. Schmitt; A. Bichon; P. Pelissier et al. The Prominent Role of Serotonergic Degeneration in Apathy, Anxiety and Depression in de Novo Parkinson’s Disease., 2016, 139,pp. 2486-2502. DOI: https://doi.org/10.1093/brain/aww162. PMID: https://www.ncbi.nlm.nih.gov/pubmed/27538418.

145. P. Wichit; S. Thanprasertsuk; O. Phokaewvarangkul; R. Bhidayasiri; S. Bongsebandhu-phubhakdi Monoamine Levels and Parkinson’s Disease Progression: Evidence from a High-Performance Liquid Chromatography Study., 2021, 15, 605887. DOI: https://doi.org/10.3389/fnins.2021.605887. PMID: https://www.ncbi.nlm.nih.gov/pubmed/34393700.

146. L. Batzu; A. Podlewska; L. Gibson; K.R. Chaudhuri; D. Aarsland A General Clinical Overview of the Non-Motor Symptoms in Parkinson’s Disease: Neuropsychiatric Symptoms., 2024, 174,pp. 59-97. PMID: https://www.ncbi.nlm.nih.gov/pubmed/38341232.

147. R.N. Taddei; S. Cankaya; S. Dhaliwal; K.R. Chaudhuri Management of Psychosis in Parkinson’s Disease: Emphasizing Clinical Subtypes and Pathophysiological Mechanisms of the Condition., 2017, 2017,p. 3256542. DOI: https://doi.org/10.1155/2017/3256542. PMID: https://www.ncbi.nlm.nih.gov/pubmed/29104810.

148. A. Polissidis; M. Koronaiou; V. Kollia; E. Koronaiou; M. Nakos-Bimpos; M. Bogiongko; S. Vrettou; K. Karali; N. Casadei; O. Riess et al. Psychosis-like Behavior and Hyperdopaminergic Dysregulation in Human a-Synuclein BAC Transgenic Rats., 2021, 36,pp. 716-728. DOI: https://doi.org/10.1002/mds.28383.

149. J. Pagonabarraga; H. Bejr-Kasem; S. Martinez-Horta; J. Kulisevsky Parkinson Disease Psychosis: From Phenomenology to Neurobiological Mechanisms., 2024, 20,pp. 135-150. DOI: https://doi.org/10.1038/s41582-023-00918-8.

150. R. Savica; B.R. Grossardt; J.H. Bower; J.E. Ahlskog; M.M. Mielke; W.A. Rocca Incidence and Time Trends of Drug-Induced Parkinsonism: A 30-Year Population-Based Study., 2017, 32,pp. 227-234. DOI: https://doi.org/10.1002/mds.26839.

151. D. Göverti; N. Büyüklüoglu; R. Nazik Yüksel; H. Kaya; Ç. Yücel; E. Göka Decreased Serum Levels of a-Synuclein in Patients with Schizophrenia and Their Unaffected Siblings., 2023, 17,pp. 1079-1086. DOI: https://doi.org/10.1111/eip.13398.

152. Ö.F. Demirel; I. Cetin; S. Turan; T. Saglam; N. Yildiz; A. Duran Decreased Expression of a-Synuclein, Nogo-A and UCH-L1 in Patients with Schizophrenia: A Preliminary Serum Study., 2017, 14,p. 344. DOI: https://doi.org/10.4306/pi.2017.14.3.344.

153. M.R. Noori-Daloii; M. Kheirollahi; P. Mahbod; F. Mohammadi; A.N. Astaneh; M.R. Zarindast; C. Azimi; M.R. Mohammadi Alpha- and Beta-Synucleins MRNA Expression in Lymphocytes of Schizophrenia Patients., 2010, 14,pp. 725-729. DOI: https://doi.org/10.1089/gtmb.2010.0050.

154. S. Takamura; A. Ikeda; K. Nishioka; H. Furuya; M. Tashiro; T. Matsushima; Y. Li; H. Yoshino; M. Funayama; S. Morinobu et al. Schizophrenia as a Prodromal Symptom in a Patient Harboring SNCA Duplication., 2016, 25,pp. 108-109. DOI: https://doi.org/10.1016/j.parkreldis.2016.01.028.

155. F.E.C. Mosquera; M.C. Guevara-Montoya; V. Serna-Ramirez; Y. Liscano Neuroinflammation and Schizophrenia: New Therapeutic Strategies through Psychobiotics, Nanotechnology, and Artificial Intelligence (AI)., 2024, 14, 391. DOI: https://doi.org/10.3390/jpm14040391.

156. M. Gerlach; M. Sharma; M. Romanos; K.-P. Lesch; S. Walitza; H.A. Conzelmann; R. Krüger; T.J. Renner Family-Based Association Study on Functional a-Synuclein Polymorphisms in Attention-Deficit/Hyperactivity Disorder., 2019, 11,pp. 107-111. DOI: https://doi.org/10.1007/s12402-019-00286-8. PMID: https://www.ncbi.nlm.nih.gov/pubmed/30927235.

157. D. Erskine; A.J. Thomas; J.-P. Taylor; M.A. Savage; J. Attems; I.G. McKeith; C.M. Morris; A.A. Khundakar Neuronal Loss and A-Synuclein Pathology in the Superior Colliculus and Its Relationship to Visual Hallucinations in Dementia with Lewy Bodies., 2017, 25,pp. 595-604. DOI: https://doi.org/10.1016/j.jagp.2017.01.005. PMID: https://www.ncbi.nlm.nih.gov/pubmed/28190674.

158. T. Usenko; A. Bezrukova; K. Basharova; G. Baydakova; E. Shagimardanova; N. Blatt; A. Rizvanov; O. Limankin; M. Novitskiy; N. Shnayder et al. Altered Sphingolipid Hydrolase Activities and Alpha-Synuclein Level in Late-Onset Schizophrenia., 2023, 14, 30. DOI: https://doi.org/10.3390/metabo14010030. PMID: https://www.ncbi.nlm.nih.gov/pubmed/38248833.

159. A.K. Gupta; R. Pokhriyal; U. Das; M.I. Khan; D. Ratna Kumar; R. Gupta; R.K. Chadda; R. Ramachandran; V. Goyal; M. Tripathi et al. Evaluation of a-Synuclein and Apolipoprotein E as Potential Biomarkers in Cerebrospinal Fluid to Monitor Pharmacotherapeutic Efficacy in Dopamine Dictated Disease States of Parkinson’s Disease and Schizophrenia., 2019, 15,pp. 2073-2085. DOI: https://doi.org/10.2147/NDT.S205550.

160. C.A. Altar; L.W. Jurata; V. Charles; A. Lemire; P. Liu; Y. Bukhman; T.A. Young; J. Bullard; H. Yokoe; M.J. Webster et al. Deficient Hippocampal Neuron Expression of Proteasome, Ubiquitin, and Mitochondrial Genes in Multiple Schizophrenia Cohorts., 2005, 58,pp. 85-96. DOI: https://doi.org/10.1016/j.biopsych.2005.03.031.

161. E. Cuvelier; M. Méquinion; C. Leghay; W. Sibran; A. Stievenard; A. Sarchione; M.-A. Bonte; C. Vanbesien-Mailliot; O. Viltart; K. Saitoski et al. Overexpression of Wild-Type Human Alpha-Synuclein Causes Metabolism Abnormalities in Thy1-ASYN Transgenic Mice., 2018, 11, 321. DOI: https://doi.org/10.3389/fnmol.2018.00321. PMID: https://www.ncbi.nlm.nih.gov/pubmed/30333721.

162. T. Stewart; C. Liu; C. Ginghina; K.C. Cain; P. Auinger; B. Cholerton; M. Shi; J. Zhang Cerebrospinal Fluid a-Synuclein Predicts Cognitive Decline in Parkinson Disease Progression in the DATATOP Cohort., 2014, 184,pp. 966-975. DOI: https://doi.org/10.1016/j.ajpath.2013.12.007. PMID: https://www.ncbi.nlm.nih.gov/pubmed/24625392.

163. S. Hall; Y. Surova; A. Öhrfelt; H. Zetterberg; D. Lindqvist; O. Hansson CSF Biomarkers and Clinical Progression of Parkinson Disease., 2015, 84,pp. 57-63. DOI: https://doi.org/10.1212/WNL.0000000000001098. PMID: https://www.ncbi.nlm.nih.gov/pubmed/25411441.

164. C.-H. Lin; S.-Y. Yang; H.-E. Horng; C.-C. Yang; J.-J. Chieh; H.-H. Chen; B.-H. Liu; M.-J. Chiu Plasma a-Synuclein Predicts Cognitive Decline in Parkinson’s Disease., 2017, 88,pp. 818-824. DOI: https://doi.org/10.1136/jnnp-2016-314857.

165. T.-T. Yang; C.-G. Liu; S.-C. Gao; Y. Zhang; P.-C. Wang The Serum Exosome Derived MicroRNA-135a, -193b, and -384 Were Potential Alzheimer’s Disease Biomarkers., 2018, 31,pp. 87-96. PMID: https://www.ncbi.nlm.nih.gov/pubmed/29606187.

166. L. Han; Y. Tang; X. Bai; X. Liang; Y. Fan; Y. Shen; F. Huang; J. Wang Association of the Serum MicroRNA-29 Family with Cognitive Impairment in Parkinson’s Disease., 2020, 12,pp. 13518-13528. DOI: https://doi.org/10.18632/aging.103458. PMID: https://www.ncbi.nlm.nih.gov/pubmed/32649312.

167. S. Li; Z. Lei; T. Sun The Role of MicroRNAs in Neurodegenerative Diseases: A Review., 2023, 39,pp. 53-83. DOI: https://doi.org/10.1007/s10565-022-09761-x.

168. F. Stabile; G. Torromino; S. Rajendran; G. Del Vecchio; C. Presutti; C. Mannironi; E. De Leonibus; A. Mele; A. Rinaldi Short-Term Memory Deficit Associates with MiR-153-3p Upregulation in the Hippocampus of Middle-Aged Mice., 2024, 61,pp. 3031-3041. DOI: https://doi.org/10.1007/s12035-023-03770-5.

169. S. Palmqvist; M. Rossi; S. Hall; C. Quadalti; N. Mattsson-Carlgren; S. Dellavalle; P. Tideman; J.B. Pereira; M.H. Nilsson; A. Mammana et al. Cognitive Effects of Lewy Body Pathology in Clinically Unimpaired Individuals., 2023, 29,pp. 1971-1978. DOI: https://doi.org/10.1038/s41591-023-02450-0.

170. M.F. Manchinu; M. Pala; M.F. Palmas; M.A. Diana; A. Maschio; M. Etzi; A. Pisanu; F.I. Diana; J. Marongiu; S. Mansueto et al. Region-Specific Changes in Gene Expression Are Associated with Cognitive Deficits in the Alpha-Synuclein-Induced Model of Parkinson’s Disease: A Transcriptomic Profiling Study., 2024, 372,p. 114651. DOI: https://doi.org/10.1016/j.expneurol.2023.114651.

171. K. Rosborough; N. Patel; L.V. Kalia a-Synuclein and Parkinsonism: Updates and Future Perspectives., 2017, 17,p. 31. DOI: https://doi.org/10.1007/s11910-017-0737-y. PMID: https://www.ncbi.nlm.nih.gov/pubmed/28324300.

172. E. Doxakis Post-Transcriptional Regulation of a-Synuclein Expression by Mir-7 and Mir-153., 2010, 285,pp. 12726-12734. DOI: https://doi.org/10.1074/jbc.M109.086827. PMID: https://www.ncbi.nlm.nih.gov/pubmed/20106983.

173. N. Fussi; M. Höllerhage; T. Chakroun; N.-P. Nykänen; T.W. Rösler; T. Koeglsperger; W. Wurst; C. Behrends; G.U. Höglinger Exosomal Secretion of a-Synuclein as Protective Mechanism after Upstream Blockage of Macroautophagy., 2018, 9,p. 757. DOI: https://doi.org/10.1038/s41419-018-0816-2.

174. J.R. Mazzulli; Y.-H. Xu; Y. Sun; A.L. Knight; P.J. McLean; G.A. Caldwell; E. Sidransky; G.A. Grabowski; D. Krainc Gaucher Disease Glucocerebrosidase and a-Synuclein Form a Bidirectional Pathogenic Loop in Synucleinopathies., 2011, 146,pp. 37-52. DOI: https://doi.org/10.1016/j.cell.2011.06.001. PMID: https://www.ncbi.nlm.nih.gov/pubmed/21700325.

175. J. Lu; S. Zhang; X. Ma; C. Jia; Z. Liu; C. Huang; C. Liu; D. Li Structural Basis of the Interplay between a-Synuclein and Tau in Regulating Pathological Amyloid Aggregation., 2020, 295,pp. 7470-7480. DOI: https://doi.org/10.1074/jbc.RA119.012284. PMID: https://www.ncbi.nlm.nih.gov/pubmed/32291284.

176. M.J. Lim; S.L. Boschen; A. Kurti; M. Castanedes Casey; V.R. Phillips; J.D. Fryer; D. Dickson; K.R. Jansen-West; L. Petrucelli; M. Delenclos et al. Investigating the Pathogenic Interplay of Alpha-Synuclein, Tau, and Amyloid Beta in Lewy Body Dementia: Insights from Viral-Mediated Overexpression in Transgenic Mouse Models., 2023, 11, 2863. DOI: https://doi.org/10.3390/biomedicines11102863. PMID: https://www.ncbi.nlm.nih.gov/pubmed/37893236.

177. A. Prasad; V. Bharathi; V. Sivalingam; A. Girdhar; B.K. Patel Molecular Mechanisms of TDP-43 Misfolding and Pathology in Amyotrophic Lateral Sclerosis., 2019, 12, 25. DOI: https://doi.org/10.3389/fnmol.2019.00025.

178. A. Licht-Murava; S.M. Meadows; F. Palaguachi; S.C. Song; S. Jackvony; Y. Bram; C. Zhou; R.E. Schwartz; R.C. Froemke; A.L. Orr et al. Astrocytic TDP-43 Dysregulation Impairs Memory by Modulating Antiviral Pathways and Interferon-Inducible Chemokines., 2023, 9,p. eade1282. DOI: https://doi.org/10.1126/sciadv.ade1282. PMID: https://www.ncbi.nlm.nih.gov/pubmed/37075107.

179. B. Ford Pain in Parkinson’s Disease., 2010, 25,pp. S98-S103. DOI: https://doi.org/10.1002/mds.22716.

180. Y. Buhidma; C. Hobbs; M. Malcangio; S. Duty Periaqueductal Grey and Spinal Cord Pathology Contribute to Pain in Parkinson’s Disease., 2023, 9,p. 69. DOI: https://doi.org/10.1038/s41531-023-00510-3.

181. N. Ferreira; N.P. Gonçalves; A. Jan; N.M. Jensen; A. van der Laan; S. Mohseni; C.B. Vægter; P.H. Jensen Trans-Synaptic Spreading of Alpha-Synuclein Pathology through Sensory Afferents Leads to Sensory Nerve Degeneration and Neuropathic Pain., 2021, 9,p. 31. DOI: https://doi.org/10.1186/s40478-021-01131-8. PMID: https://www.ncbi.nlm.nih.gov/pubmed/33632316.

182. D. Li; X. Song; H. Huang; H. Huang; Z. Ye Association of Parkinson’s Disease-Related Pain with Plasma Interleukin-1, Interleukin-6, Interleukin-10, and Tumour Necrosis Factor-a., 2018, 683,pp. 181-184. DOI: https://doi.org/10.1016/j.neulet.2018.07.027. PMID: https://www.ncbi.nlm.nih.gov/pubmed/30063943.

183. M. Möller; C.V. Möser; U. Weiß; E. Niederberger The Role of AlphaSynuclein in Mouse Models of Acute, Inflammatory and Neuropathic Pain., 2022, 11, 1967. DOI: https://doi.org/10.3390/cells11121967. PMID: https://www.ncbi.nlm.nih.gov/pubmed/35741096.

184. T. Chen; J. Li; D. Chao; H.K. Sandhu; X. Liao; J. Zhao; G. Wen; Y. Xia d-Opioid Receptor Activation Reduces a-Synuclein Overexpression and Oligomer Formation Induced by MPP+ and/or Hypoxia., 2014, 255,pp. 127-136. DOI: https://doi.org/10.1016/j.expneurol.2014.02.022. PMID: https://www.ncbi.nlm.nih.gov/pubmed/24613828.

185. M.-H. Yi; J. Shin; N. Shin; Y. Yin; S.Y. Lee; C.-S. Kim; S.R. Kim; E. Zhang; D.W. Kim PINK1 Mediates Spinal Cord Mitophagy in Neuropathic Pain., 2019, 12,pp. 1685-1699. DOI: https://doi.org/10.2147/JPR.S198730. PMID: https://www.ncbi.nlm.nih.gov/pubmed/31239755.

186. S. Jinn; R.E. Drolet; P.E. Cramer; A.H.-K. Wong; D.M. Toolan; C.A. Gretzula; B. Voleti; G. Vassileva; J. Disa; M. Tadin-Strapps et al. TMEM175 Deficiency Impairs Lysosomal and Mitochondrial Function and Increases a-Synuclein Aggregation., 2017, 114,pp. 2389-2394. DOI: https://doi.org/10.1073/pnas.1616332114. PMID: https://www.ncbi.nlm.nih.gov/pubmed/28193887.

187. D. Berg; R.B. Postuma; C.H. Adler; B.R. Bloem; P. Chan; B. Dubois; T. Gasser; C.G. Goetz; G. Halliday; L. Joseph et al. MDS Research Criteria for Prodromal Parkinson’s Disease., 2015, 30,pp. 1600-1611. DOI: https://doi.org/10.1002/mds.26431. PMID: https://www.ncbi.nlm.nih.gov/pubmed/26474317.

188. T.G. Beach; C.L. White; C.L. Hladik; M.N. Sabbagh; D.J. Connor; H.A. Shill; L.I. Sue; J. Sasse; J. Bachalakuri; J. Henry-Watson et al. Olfactory Bulb a-Synucleinopathy Has High Specificity and Sensitivity for Lewy Body Disorders., 2009, 117,p. 169. DOI: https://doi.org/10.1007/s00401-008-0450-7. PMID: https://www.ncbi.nlm.nih.gov/pubmed/18982334.

189. D.M. Mason; Y. Wang; T.N. Bhatia; K.M. Miner; S.A. Trbojevic; J.F. Stolz; K.C. Luk; R.K. Leak The Center of Olfactory Bulb-seeded a-Synucleinopathy Is the Limbic System and the Ensuing Pathology Is Higher in Male than in Female Mice., 2019, 29,pp. 741-770. DOI: https://doi.org/10.1111/bpa.12718.

190. M.E. Johnson; L. Bergkvist; G. Mercado; L. Stetzik; L. Meyerdirk; E. Wolfrum; Z. Madaj; P. Brundin; D.W. Wesson Deficits in Olfactory Sensitivity in a Mouse Model of Parkinson’s Disease Revealed by Plethysmography of Odor-Evoked Sniffing., 2020, 10,p. 9242. DOI: https://doi.org/10.1038/s41598-020-66201-8.

191. E. Martin-Lopez; D.J. Vidyadhara; T. Liberia; S.J. Meller; L.E. Harmon; R.M. Hsu; N. Spence; B. Brennan; K. Han; B. Yücel et al. a-Synuclein Pathology and Reduced Neurogenesis in the Olfactory System Affect Olfaction in a Mouse Model of Parkinson’s Disease., 2023, 43,pp. 1051-1071. DOI: https://doi.org/10.1523/JNEUROSCI.1526-22.2022. PMID: https://www.ncbi.nlm.nih.gov/pubmed/36596700.

192. F. Chen; W. Liu; P. Liu; Z. Wang; Y. Zhou; X. Liu; A. Li a-Synuclein Aggregation in the Olfactory Bulb Induces Olfactory Deficits by Perturbing Granule Cells and Granular–Mitral Synaptic Transmission., 2021, 7,p. 114. DOI: https://doi.org/10.1038/s41531-021-00259-7. PMID: https://www.ncbi.nlm.nih.gov/pubmed/34903719.

193. X.-Y. Liu; K. Wang; X.-H. Deng; Y.-H. Wei; R. Guo; S.-F. Liu; Y.-F. Zhu; J.-J. Zhong; J.-Y. Zheng; M.-D. Wang et al. Amelioration of Olfactory Dysfunction in a Mouse Model of Parkinson’s Disease via Enhancing GABAergic Signaling., 2023, 13, 101. DOI: https://doi.org/10.1186/s13578-023-01049-9. PMID: https://www.ncbi.nlm.nih.gov/pubmed/37270503.

194. S. Zhang; Q. Xiao; W. Le Olfactory Dysfunction and Neurotransmitter Disturbance in Olfactory Bulb of Transgenic Mice Expressing Human A53T Mutant a-Synuclein., 2015, 10, e0119928. DOI: https://doi.org/10.1371/journal.pone.0119928. PMID: https://www.ncbi.nlm.nih.gov/pubmed/25799501.

195. H. Niu; Q. Wang; W. Zhao; J. Liu; D. Wang; B. Muhammad; X. Liu; N. Quan; H. Zhang; F. Zhang et al. IL-1ß/IL-1R1 Signaling Induced by Intranasal Lipopolysaccharide Infusion Regulates Alpha-Synuclein Pathology in the Olfactory Bulb, Substantia Nigra and Striatum., 2020, 30,pp. 1102-1118. DOI: https://doi.org/10.1111/bpa.12886. PMID: https://www.ncbi.nlm.nih.gov/pubmed/32678959.

196. C.N. Martyn Infection in Childhood and Neurological Diseases in Adult Life., 1997, 53,pp. 24-39. DOI: https://doi.org/10.1093/oxfordjournals.bmb.a011603. PMID: https://www.ncbi.nlm.nih.gov/pubmed/9158282.

197. X.-L. Bu; X. Wang; Y. Xiang; L.-L. Shen; Q.-H. Wang; Y.-H. Liu; S.-S. Jiao; Y.-R. Wang; H.-Y. Cao; X. Yi et al. The Association between Infectious Burden and Parkinson’s Disease: A Case-Control Study., 2015, 21,pp. 877-881. DOI: https://doi.org/10.1016/j.parkreldis.2015.05.015. PMID: https://www.ncbi.nlm.nih.gov/pubmed/26037459.

198. M. Campolo; I. Paterniti; R. Siracusa; A. Filippone; E. Esposito; S. Cuzzocrea TLR4 Absence Reduces Neuroinflammation and Inflammasome Activation in Parkinson’s Diseases in Vivo Model., 2019, 76,pp. 236-247. DOI: https://doi.org/10.1016/j.bbi.2018.12.003. PMID: https://www.ncbi.nlm.nih.gov/pubmed/30550933.

199. F. Nieto-Escamez; E. Obrero-Gaitán; I. Cortés-Pérez Visual Dysfunction in Parkinson’s Disease., 2023, 13, 1173. DOI: https://doi.org/10.3390/brainsci13081173.

200. I. Ortuño-Lizarán; G. Esquiva; T.G. Beach; G.E. Serrano; C.H. Adler; P. Lax; N. Cuenca Degeneration of Human Photosensitive Retinal Ganglion Cells May Explain Sleep and Circadian Rhythms Disorders in Parkinson’s Disease., 2018, 6,p. 90. DOI: https://doi.org/10.1186/s40478-018-0596-z.

201. K.K.N. Tran; V.H.Y. Wong; A. Hoang; D.I. Finkelstein; B.V. Bui; C.T.O. Nguyen Retinal Alpha-Synuclein Accumulation Correlates with Retinal Dysfunction and Structural Thinning in the A53T Mouse Model of Parkinson’s Disease., 2023, 17, 1146979. DOI: https://doi.org/10.3389/fnins.2023.1146979. PMID: https://www.ncbi.nlm.nih.gov/pubmed/37214398.

202. E. Marrocco; A. Indrieri; F. Esposito; V. Tarallo; A. Carboncino; F.G. Alvino; S. De Falco; B. Franco; M. De Risi; E. De Leonibus a-Synuclein Overexpression in the Retina Leads to Vision Impairment and Degeneration of Dopaminergic Amacrine Cells., 2020, 10,p. 9619. DOI: https://doi.org/10.1038/s41598-020-66497-6. PMID: https://www.ncbi.nlm.nih.gov/pubmed/32541823.

203. D. Pérez-Acuña; K.H. Rhee; S.J. Shin; J. Ahn; J.-Y. Lee; S.-J. Lee Retina-to-Brain Spreading of a-Synuclein after Intravitreal Injection of Preformed Fibrils., 2023, 11,p. 83. DOI: https://doi.org/10.1186/s40478-023-01575-0. PMID: https://www.ncbi.nlm.nih.gov/pubmed/37210559.

204. I. Ortuño-Lizarán; X. Sánchez-Sáez; P. Lax; G.E. Serrano; T.G. Beach; C.H. Adler; N. Cuenca Dopaminergic Retinal Cell Loss and Visual Dysfunction in Parkinson Disease., 2020, 88,pp. 893-906. DOI: https://doi.org/10.1002/ana.25897. PMID: https://www.ncbi.nlm.nih.gov/pubmed/32881029.

205. S. Baksi; N. Singh a-Synuclein Impairs Ferritinophagy in the Retinal Pigment Epithelium: Implications for Retinal Iron Dyshomeostasis in Parkinson’s Disease., 2017, 7,p. 12843. DOI: https://doi.org/10.1038/s41598-017-12862-x. PMID: https://www.ncbi.nlm.nih.gov/pubmed/28993630.

206. M.-A. Samizadeh; H. Fallah; M. Toomarisahzabi; F. Rezaei; M. Rahimi-Danesh; S. Akhondzadeh; S. Vaseghi Parkinson’s Disease: A Narrative Review on Potential Molecular Mechanisms of Sleep Disturbances, REM Behavior Disorder, and Melatonin., 2023, 13, 914. DOI: https://doi.org/10.3390/brainsci13060914. PMID: https://www.ncbi.nlm.nih.gov/pubmed/37371392.

207. A. Roguski; D. Rayment; A.L. Whone; M.W. Jones; M. Rolinski A Neurologist’s Guide to REM Sleep Behavior Disorder., 2020, 11, 610. DOI: https://doi.org/10.3389/fneur.2020.00610. PMID: https://www.ncbi.nlm.nih.gov/pubmed/32733361.

208. P.-C. Chan; H.-H. Lee; C.-T. Hong; C.-J. Hu; D. Wu REM Sleep Behavior Disorder (RBD) in Dementia with Lewy Bodies (DLB)., 2018, 2018,p. 9421098. DOI: https://doi.org/10.1155/2018/9421098. PMID: https://www.ncbi.nlm.nih.gov/pubmed/30018672.

209. A. Schaffrath; S. Schleyken; A. Seger; H. Jergas; P. Özdüzenciler; M. Pils; L. Blömeke; A. Cousin; J. Willbold; T. Bujnicki et al. Patients with Isolated REM-Sleep Behavior Disorder Have Elevated Levels of Alpha-Synuclein Aggregates in Stool., 2023, 9,p. 14. DOI: https://doi.org/10.1038/s41531-023-00458-4.

210. T. Taguchi; M. Ikuno; M. Hondo; L.K. Parajuli; K. Taguchi; J. Ueda; M. Sawamura; S. Okuda; E. Nakanishi; J. Hara et al. a-Synuclein BAC Transgenic Mice Exhibit RBD-like Behaviour and Hyposmia: A Prodromal Parkinson’s Disease Model., 2020, 143,pp. 249-265. DOI: https://doi.org/10.1093/brain/awz380.

211. A. Iranzo; A. Fernández-Arcos; E. Tolosa; M. Serradell; J.L. Molinuevo; F. Valldeoriola; E. Gelpi; I. Vilaseca; R. Sánchez-Valle; A. Lladó et al. Neurodegenerative Disorder Risk in Idiopathic REM Sleep Behavior Disorder: Study in 174 Patients., 2014, 9, e89741. DOI: https://doi.org/10.1371/journal.pone.0089741. PMID: https://www.ncbi.nlm.nih.gov/pubmed/24587002.

212. D.P. Breen; R. Vuono; U. Nawarathna; K. Fisher; J.M. Shneerson; A.B. Reddy; R.A. Barker Sleep and Circadian Rhythm Regulation in Early Parkinson Disease., 2014, 71,p. 589. DOI: https://doi.org/10.1001/jamaneurol.2014.65. PMID: https://www.ncbi.nlm.nih.gov/pubmed/24687146.

213. S. Diaconu; L. Irincu; L. Ungureanu; B. Ciopleia?; D. ?în?; C. Falup-Pecurariu Restless Legs Syndrome in Parkinson’s Disease., 2023, 13, 915. DOI: https://doi.org/10.3390/jpm13060915. PMID: https://www.ncbi.nlm.nih.gov/pubmed/37373904.

214. S. Azmin; A.M. Khairul Anuar; W.Y. Nafisah; H.J. Tan; A.A. Raymond; O. Hanita; S.A. Shah; M.I. Norlinah Restless Legs Syndrome and Its Associated Risk Factors in Parkinson’s Disease., 2013, 2013,p. 535613. DOI: https://doi.org/10.1155/2013/535613. PMID: https://www.ncbi.nlm.nih.gov/pubmed/24455416.

215. S. Chung; N.I. Bohnen; R.L. Albin; K.A. Frey; M.L.T.M. Müller; R.D. Chervin Insomnia and Sleepiness in Parkinson Disease: Associations with Symptoms and Comorbidities., 2013, 09,pp. 1131-1137. DOI: https://doi.org/10.5664/jcsm.3150. PMID: https://www.ncbi.nlm.nih.gov/pubmed/24235893.

216. H. Braak; E. Ghebremedhin; U. Rüb; H. Bratzke; K. Del Tredici Stages in the Development of Parkinson’s Disease-Related Pathology., 2004, 318,pp. 121-134. DOI: https://doi.org/10.1007/s00441-004-0956-9. PMID: https://www.ncbi.nlm.nih.gov/pubmed/15338272.

217. J. Lu; D. Sherman; M. Devor; C.B. Saper A Putative Flip–Flop Switch for Control of REM Sleep., 2006, 441,pp. 589-594. DOI: https://doi.org/10.1038/nature04767. PMID: https://www.ncbi.nlm.nih.gov/pubmed/16688184.

218. M.M.S. Lima; M.L. Andersen; A.B. Reksidler; M.A.B.F. Vital; S. Tufik The Role of the Substantia Nigra Pars Compacta in Regulating Sleep Patterns in Rats., 2007, 2, e513. DOI: https://doi.org/10.1371/journal.pone.0000513. PMID: https://www.ncbi.nlm.nih.gov/pubmed/17551593.

219. L.D. Willison; T. Kudo; D.H. Loh; D. Kuljis; C.S. Colwell Circadian Dysfunction May Be a Key Component of the Non-Motor Symptoms of Parkinson’s Disease: Insights from a Transgenic Mouse Model., 2013, 243,pp. 57-66. DOI: https://doi.org/10.1016/j.expneurol.2013.01.014.

220. K.A. McDowell; D. Shin; K.P. Roos; M.-F. Chesselet Sleep Dysfunction and EEG Alterations in Mice Overexpressing Alpha-Synuclein., 2018, 4,pp. 531-539. DOI: https://doi.org/10.3233/JPD-140374.

221. M.B. Gajula Balija; C. Griesinger; A. Herzig; M. Zweckstetter; H. Jäckle Pre-Fibrillar a-Synuclein Mutants Cause Parkinson’s Disease-like Non-Motor Symptoms in Drosophila., 2011, 6, e24701. DOI: https://doi.org/10.1371/journal.pone.0024701. PMID: https://www.ncbi.nlm.nih.gov/pubmed/21931820.

222. N.R. Dhanushkodi; S.B. Abul Khair; M.T. Ardah; M.E. Haque ATP13A2 Gene Silencing in Drosophila Affects Autophagic Degradation of A53T Mutant a-Synuclein., 2023, 24, 1775. DOI: https://doi.org/10.3390/ijms24021775.

223. S.B. Abul Khair; N.R. Dhanushkodi; M.T. Ardah; W. Chen; Y. Yang; M.E. Haque Silencing of Glucocerebrosidase Gene in Drosophila Enhances the Aggregation of Parkinson’s Disease Associated a-Synuclein Mutant A53T and Affects Locomotor Activity., 2018, 12, 81. DOI: https://doi.org/10.3389/fnins.2018.00081. PMID: https://www.ncbi.nlm.nih.gov/pubmed/29503608.

224. K. Ito; H. Kawasaki; T. Suzuki; T. Takahara; N. Ishida Effects of Kamikihito and Unkei-to on Sleep Behavior of Wild Type and Parkinson Model in Drosophila., 2017, 8, 132. DOI: https://doi.org/10.3389/fpsyt.2017.00132. PMID: https://www.ncbi.nlm.nih.gov/pubmed/28824465.

225. A. Borbély The Two-Process Model of Sleep Regulation: Beginnings and Outlook., 2022, 31,p. e13598. DOI: https://doi.org/10.1111/jsr.13598. PMID: https://www.ncbi.nlm.nih.gov/pubmed/35502706.

226. A. Patke; M.W. Young; S. Axelrod Molecular Mechanisms and Physiological Importance of Circadian Rhythms., 2020, 21,pp. 67-84. DOI: https://doi.org/10.1038/s41580-019-0179-2. PMID: https://www.ncbi.nlm.nih.gov/pubmed/31768006.

227. A. Videnovic; C. Noble; K.J. Reid; J. Peng; F.W. Turek; A. Marconi; A.W. Rademaker; T. Simuni; C. Zadikoff; P.C. Zee Circadian Melatonin Rhythm and Excessive Daytime Sleepiness in Parkinson Disease., 2014, 71,p. 463. DOI: https://doi.org/10.1001/jamaneurol.2013.6239. PMID: https://www.ncbi.nlm.nih.gov/pubmed/24566763.

228. C.B. Peek; A.H. Affinati; K.M. Ramsey; H.-Y. Kuo; W. Yu; L.A. Sena; O. Ilkayeva; B. Marcheva; Y. Kobayashi; C. Omura et al. Circadian Clock NAD+ Cycle Drives Mitochondrial Oxidative Metabolism in Mice., 2013, 342,p. 1243417. DOI: https://doi.org/10.1126/science.1243417. PMID: https://www.ncbi.nlm.nih.gov/pubmed/24051248.

229. P. Rathor; R. Ch Metabolic Basis of Circadian Dysfunction in Parkinson’s Disease., 2023, 12, 1294. DOI: https://doi.org/10.3390/biology12101294.

230. T. Warnecke; K.-H. Schäfer; I. Claus; K. Del Tredici; W.H. Jost Gastrointestinal Involvement in Parkinson’s Disease: Pathophysiology, Diagnosis, and Management., 2022, 8,p. 31. DOI: https://doi.org/10.1038/s41531-022-00295-x.

231. E. Camerucci; A.F. Mullan; J.H. Bower; A.E. Bharucha; P. Turcano; C.D. Stang; E.E. Benarroch; B.F. Boeve; J.E. Ahlskog; R. Savica Lifelong Constipation in Parkinson’s Disease and Other Clinically Defined Alpha-Synucleinopathies: A Population-Based Study in Southeast Minnesota., 2023, 107,p. 105244. DOI: https://doi.org/10.1016/j.parkreldis.2022.105244. PMID: https://www.ncbi.nlm.nih.gov/pubmed/36630736.

232. P. Borghammer; J. Horsager; K. Andersen; N. Van Den Berge; A. Raunio; S. Murayama; L. Parkkinen; L. Myllykangas Neuropathological Evidence of Body-First vs. Brain-First Lewy Body Disease., 2021, 161, 105557. DOI: https://doi.org/10.1016/j.nbd.2021.105557. PMID: https://www.ncbi.nlm.nih.gov/pubmed/34763110.

233. J. Horsager; K.B. Andersen; K. Knudsen; C. Skjærbæk; T.D. Fedorova; N. Okkels; E. Schaeffer; S.K. Bonkat; J. Geday; M. Otto et al. Brain-First versus Body-First Parkinson’s Disease: A Multimodal Imaging Case-Control Study., 2020, 143,pp. 3077-3088. DOI: https://doi.org/10.1093/brain/awaa238. PMID: https://www.ncbi.nlm.nih.gov/pubmed/32830221.

234. P. Borghammer; M.K. Just; J. Horsager; C. Skjærbæk; A. Raunio; E.H. Kok; S. Savola; S. Murayama; Y. Saito; L. Myllykangas et al. A Postmortem Study Suggests a Revision of the Dual-Hit Hypothesis of Parkinson’s Disease., 2022, 8,p. 166. DOI: https://doi.org/10.1038/s41531-022-00436-2. PMID: https://www.ncbi.nlm.nih.gov/pubmed/36450732.

235. Y. Li; Q. Tong; Y. Wang; Y. Cheng; Y. Geng; T. Tian; Y. Yuan; Y. Fan; M. Lu; K. Zhang Phosphorylated a-Synuclein Deposited in Schwann Cells Interacting with TLR2 Mediates Cell Damage and Induces Parkinson’s Disease Autonomic Dysfunction., 2024, 10,p. 52. DOI: https://doi.org/10.1038/s41420-024-01824-8. PMID: https://www.ncbi.nlm.nih.gov/pubmed/38278799.

236. D. Dutta; M. Jana; M. Majumder; S. Mondal; A. Roy; K. Pahan Selective Targeting of the TLR2/MyD88/NF-?B Pathway Reduces a-Synuclein Spreading In Vitro and In Vivo., 2021, 12,p. 5382. DOI: https://doi.org/10.1038/s41467-021-25767-1. PMID: https://www.ncbi.nlm.nih.gov/pubmed/34508096.

237. C. Conte; A. Ingrassia; J. Breve; J.J. Bol; E. Timmermans-Huisman; A.-M. van Dam; T. Beccari; W.D.J. van de Berg Toll-like Receptor 4 Is Upregulated in Parkinson’s Disease Patients and Co-Localizes with PSer129aSyn: A Possible Link with the Pathology., 2023, 12, 1368. DOI: https://doi.org/10.3390/cells12101368.

238. G. Beck; Y. Hori; Y. Hayashi; E. Morii; T. Takehara; H. Mochizuki Detection of Phosphorylated Alpha-Synuclein in the Muscularis Propria of the Gastrointestinal Tract Is a Sensitive Predictor for Parkinson’s Disease., 2020, 2020,p. 4687530. DOI: https://doi.org/10.1155/2020/4687530. PMID: https://www.ncbi.nlm.nih.gov/pubmed/33029342.

239. C. Conte; M. Sichetti; G. Traina Gut–Brain Axis: Focus on Neurodegeneration and Mast Cells., 2020, 10, 1828. DOI: https://doi.org/10.3390/app10051828.

240. S. Ryman; A.A. Vakhtin; S.P. Richardson; H.C. Lin Microbiome–Gut–Brain Dysfunction in Prodromal and Symptomatic Lewy Body Diseases., 2023, 270,pp. 746-758. DOI: https://doi.org/10.1007/s00415-022-11461-9.

241. A.H. Tan; K.H. Chuah; Y.Y. Beh; J.P. Schee; S. Mahadeva; S.-Y. Lim Gastrointestinal Dysfunction in Parkinson’s Disease: Neuro-Gastroenterology Perspectives on a Multifaceted Problem., 2023, 16,pp. 138-151. DOI: https://doi.org/10.14802/jmd.22220.

242. R.A. Liddle Parkinson’s Disease from the Gut., 2018, 1693,pp. 201-206. DOI: https://doi.org/10.1016/j.brainres.2018.01.010. PMID: https://www.ncbi.nlm.nih.gov/pubmed/29360467.

243. J. Xu; L. Wang; X. Chen; W. Le New Understanding on the Pathophysiology and Treatment of Constipation in Parkinson’s Disease., 2022, 14, 917499. DOI: https://doi.org/10.3389/fnagi.2022.917499.

244. L. Rota; C. Pellegrini; L. Benvenuti; L. Antonioli; M. Fornai; C. Blandizzi; A. Cattaneo; E. Colla Constipation, Deficit in Colon Contractions and Alpha-Synuclein Inclusions within the Colon Precede Motor Abnormalities and Neurodegeneration in the Central Nervous System in a Mouse Model of Alpha-Synucleinopathy., 2019, 8,p. 5. DOI: https://doi.org/10.1186/s40035-019-0146-z.

245. F. Scheperjans; V. Aho; P.A.B. Pereira; K. Koskinen; L. Paulin; E. Pekkonen; E. Haapaniemi; S. Kaakkola; J. Eerola-Rautio; M. Pohja et al. Gut Microbiota Are Related to Parkinson’s Disease and Clinical Phenotype., 2015, 30,pp. 350-358. DOI: https://doi.org/10.1002/mds.26069.

246. L. Ortiz de Ora; J.M. Balsamo; K.S. Uyeda; E.N. Bess Discovery of a Gut Bacterial Metabolic Pathway That Drives a-Synuclein Aggregation., 2024, 19,pp. 1011-1021. DOI: https://doi.org/10.1021/acschembio.4c00095.

247. C.-M. Qiao; M.-F. Sun; X.-B. Jia; Y. Li; B.-P. Zhang; L.-P. Zhao; Y. Shi; Z.-L. Zhou; Y.-L. Zhu; C. Cui et al. Sodium Butyrate Exacerbates Parkinson’s Disease by Aggravating Neuroinflammation and Colonic Inflammation in MPTP-Induced Mice Model., 2020, 45,pp. 2128-2142. DOI: https://doi.org/10.1007/s11064-020-03074-3. PMID: https://www.ncbi.nlm.nih.gov/pubmed/32556930.

248. N.U. Mahbub; M.M. Islam; S.-T. Hong; H.-J. Chung Dysbiosis of the Gut Microbiota and Its Effect on a-Synuclein and Prion Protein Misfolding: Consequences for Neurodegeneration., 2024, 14, 1348279. DOI: https://doi.org/10.3389/fcimb.2024.1348279. PMID: https://www.ncbi.nlm.nih.gov/pubmed/38435303.

249. K.B. Bhattacharyya; M. Rosa-Grilo Sexual Dysfunctions in Parkinson’s Disease: An Underrated Problem in a Much Discussed Disorder., 2017, 134,pp. 859-876.

250. W.-X. Duan; F. Wang; J.-Y. Liu; C.-F. Liu Relationship Between Short-Chain Fatty Acids and Parkinson’s Disease: A Review from Pathology to Clinic., 2024, 40,pp. 500-516. DOI: https://doi.org/10.1007/s12264-023-01123-9.

251. V.T.E. Aho; M.C. Houser; P.A.B. Pereira; J. Chang; K. Rudi; L. Paulin; V. Hertzberg; P. Auvinen; M.G. Tansey; F. Scheperjans Relationships of Gut Microbiota, Short-Chain Fatty Acids, Inflammation, and the Gut Barrier in Parkinson’s Disease., 2021, 16,p. 6. DOI: https://doi.org/10.1186/s13024-021-00427-6. PMID: https://www.ncbi.nlm.nih.gov/pubmed/33557896.

252. V. Kakoty; K.C. Sarathlal; S.K. Dubey; C.-H. Yang; R. Taliyan Neuroprotective Effects of Trehalose and Sodium Butyrate on Preformed Fibrillar Form of a-Synuclein-Induced Rat Model of Parkinson’s Disease., 2021, 12,pp. 2643-2660. DOI: https://doi.org/10.1021/acschemneuro.1c00144. PMID: https://www.ncbi.nlm.nih.gov/pubmed/34197084.

253. T.R. Sampson; J.W. Debelius; T. Thron; S. Janssen; G.G. Shastri; Z.E. Ilhan; C. Challis; C.E. Schretter; S. Rocha; V. Gradinaru et al. Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson’s Disease., 2016, 167,pp. 1469-1480.e12. DOI: https://doi.org/10.1016/j.cell.2016.11.018. PMID: https://www.ncbi.nlm.nih.gov/pubmed/27912057.

254. R. Sakakibara; F. Tateno; T. Yamamoto; T. Uchiyama; T. Yamanishi Urological Dysfunction in Synucleinopathies: Epidemiology, Pathophysiology and Management., 2018, 28,pp. 83-101. DOI: https://doi.org/10.1007/s10286-017-0480-0. PMID: https://www.ncbi.nlm.nih.gov/pubmed/29124503.

255. Z. Chen; G. Li; J. Liu Autonomic Dysfunction in Parkinson’s Disease: Implications for Pathophysiology, Diagnosis, and Treatment., 2020, 134, 104700. DOI: https://doi.org/10.1016/j.nbd.2019.104700. PMID: https://www.ncbi.nlm.nih.gov/pubmed/31809788.

256. T.G. Beach; C.H. Adler; L.I. Sue; L. Vedders; L. Lue; C.L. White; H. Akiyama; J.N. Caviness; H.A. Shill; M.N. Sabbagh et al. Multi-Organ Distribution of Phosphorylated a-Synuclein Histopathology in Subjects with Lewy Body Disorders., 2010, 119,pp. 689-702. DOI: https://doi.org/10.1007/s00401-010-0664-3. PMID: https://www.ncbi.nlm.nih.gov/pubmed/20306269.

257. R. Sakakibara; F. Tateno; T. Nagao; T. Yamamoto; T. Uchiyama; T. Yamanishi; M. Yano; M. Kishi; Y. Tsuyusaki; Y. Aiba Bladder Function of Patients with Parkinson’s Disease., 2014, 21,pp. 638-646. DOI: https://doi.org/10.1111/iju.12421.

258. Z. Tkaczynska; A. Pilotto; S. Becker; S. Gräber-Sultan; D. Berg; I. Liepelt-Scarfone Association between Cognitive Impairment and Urinary Dysfunction in Parkinson’s Disease., 2017, 124,pp. 543-550. DOI: https://doi.org/10.1007/s00702-017-1690-2.

259. D. Xu; S. Han; J. Wang; J. Feng Relationship between Lower Urinary Tract Dysfunction and Clinical Features in Chinese Parkinson’s Disease Patients., 2019, 2019,p. 6820937. DOI: https://doi.org/10.1155/2019/6820937.

260. Z. Tkaczynska; S. Becker; W. Maetzler; M. Timmers; L. Van Nueten; P. Sulzer; G. Salvadore; E. Schäffer; K. Brockmann; J. Streffer et al. Executive Function Is Related to the Urinary Urgency in Non-Demented Patients with Parkinson’s Disease., 2020, 12, 55. DOI: https://doi.org/10.3389/fnagi.2020.00055.

261. F. Tateno; R. Sakakibara; T. Ogata; Y. Aiba; O. Takahashi; M. Sugiyama The Relationship between Lower Urinary Tract Function and 123ioflupane Scintigraphy in Drug-Naïve Parkinson’s Disease., 2021, 233,p. 102813. DOI: https://doi.org/10.1016/j.autneu.2021.102813. PMID: https://www.ncbi.nlm.nih.gov/pubmed/33894531.

262. A. Shalash; E. Hamid; H. Elrassas; A.I. Abushouk; H.H. Salem Sexual Dysfunction in Male Patients with Parkinson’s Disease: Related Factors and Impact on Quality of Life., 2020, 41,pp. 2201-2206. DOI: https://doi.org/10.1007/s10072-020-04328-2. PMID: https://www.ncbi.nlm.nih.gov/pubmed/32172403.

263. S. Varanda; J. Ribeiro da Silva; A.S. Costa; C. Amorim de Carvalho; J.N. Alves; M. Rodrigues; G. Carneiro Sexual Dysfunction in Women with Parkinson’s Disease., 2016, 31,pp. 1685-1693. DOI: https://doi.org/10.1002/mds.26739. PMID: https://www.ncbi.nlm.nih.gov/pubmed/27501238.

264. S. Hasan; M.M. Mielke; J.E. Ahlskog; J. Bower; P. Turcano; R. Savica Erectile Dysfunction Preceding Clinically Diagnosed Synucleinopathies: A Case-Control Study in Olmsted County., 2019, 2019,p. 6303945. DOI: https://doi.org/10.1155/2019/6303945. PMID: https://www.ncbi.nlm.nih.gov/pubmed/31093326.

265. R. Shaltiel-Karyo; D. Davidi; Y. Menuchin; M. Frenkel-Pinter; M. Marcus-Kalish; J. Ringo; E. Gazit; D. Segal A Novel, Sensitive Assay for Behavioral Defects in Parkinson’s Disease Model Drosophila., 2012, 2012,p. 697564. DOI: https://doi.org/10.1155/2012/697564.

266. X. Guo; W. Song; K. Chen; X. Chen; Z. Zheng; B. Cao; R. Huang; B. Zhao; Y. Wu; H.-F. Shang Gender and Onset Age-Related Features of Non-Motor Symptoms of Patients with Parkinson’s Disease—A Study from Southwest China., 2013, 19,pp. 961-965. DOI: https://doi.org/10.1016/j.parkreldis.2013.06.009.

267. I.Z. Ben Zion; R. Tessler; L. Cohen; E. Lerer; Y. Raz; R. Bachner-Melman; I. Gritsenko; L. Nemanov; A.H. Zohar; R.H. Belmaker et al. Polymorphisms in the Dopamine D4 Receptor Gene (DRD4) Contribute to Individual Differences in Human Sexual Behavior: Desire, Arousal and Sexual Function., 2006, 11,pp. 782-786. DOI: https://doi.org/10.1038/sj.mp.4001832. PMID: https://www.ncbi.nlm.nih.gov/pubmed/16619053.

268. J. Hatate; K. Miwa; M. Matsumoto; T. Sasaki; Y. Yagita; M. Sakaguchi; K. Kitagawa; H. Mochizuki Association between Cerebral Small Vessel Diseases and Mild Parkinsonian Signs in the Elderly with Vascular Risk Factors., 2016, 26,pp. 29-34. DOI: https://doi.org/10.1016/j.parkreldis.2016.02.011. PMID: https://www.ncbi.nlm.nih.gov/pubmed/26949060.

269. J. Potashkin; X. Huang; C. Becker; H. Chen; T. Foltynie; C. Marras Understanding the Links Between Cardiovascular Disease and Parkinson’s Disease., 2020, 35,pp. 55-74. DOI: https://doi.org/10.1002/mds.27836.

270. Z. Zhou; M. Zhang; Q. Fang; J. Huang Relationship between Parkinson’s Disease and Cardio-Cerebrovascular Diseases: A Mendelian Randomized Study., 2023, 13,p. 20428. DOI: https://doi.org/10.1038/s41598-023-47708-2.

271. Y. Hu; S. Xu Association between Parkinson’s Disease and the Risk of Adverse Cardiovascular Events: A Systematic Review and Meta-Analysis., 2023, 10, 1284826. DOI: https://doi.org/10.3389/fcvm.2023.1284826. PMID: https://www.ncbi.nlm.nih.gov/pubmed/38144366.

272. G. Lamotte; C. Holmes; T. Wu; D.S. Goldstein Long-Term Trends in Myocardial Sympathetic Innervation and Function in Synucleinopathies., 2019, 67,pp. 27-33. DOI: https://doi.org/10.1016/j.parkreldis.2019.09.014. PMID: https://www.ncbi.nlm.nih.gov/pubmed/31621602.

273. S. Jain; D.S. Goldstein Cardiovascular Dysautonomia in Parkinson Disease: From Pathophysiology to Pathogenesis., 2012, 46,pp. 572-580. DOI: https://doi.org/10.1016/j.nbd.2011.10.025. PMID: https://www.ncbi.nlm.nih.gov/pubmed/22094370.

274. R. Isonaka; P. Sullivan; D.S. Goldstein Pathophysiological Significance of Increased a-Synuclein Deposition in Sympathetic Nerves in Parkinson’s Disease: A Post-Mortem Observational Study., 2022, 11,p. 15. DOI: https://doi.org/10.1186/s40035-022-00289-y. PMID: https://www.ncbi.nlm.nih.gov/pubmed/35260194.

275. F.A. Scorza; A.C. Fiorini; C.A. Scorza; J. Finsterer Cardiac Abnormalities in Parkinson’s Disease and Parkinsonism., 2018, 53,pp. 1-5. DOI: https://doi.org/10.1016/j.jocn.2018.04.031. PMID: https://www.ncbi.nlm.nih.gov/pubmed/29706419.

276. D.G. Park; J. Kang; Y.-S. An; J. Chang; J.H. Yoon Association of Plasma a-Synuclein with Cardiac 123I-MIBG Scintigraphy in Early Parkinson’s Disease., 2022, 770,p. 136399. DOI: https://doi.org/10.1016/j.neulet.2021.136399. PMID: https://www.ncbi.nlm.nih.gov/pubmed/34921912.

277. S. Orimo; T. Uchihara; A. Nakamura; F. Mori; A. Kakita; K. Wakabayashi; H. Takahashi Axonal-Synuclein Aggregates Herald Centripetal Degeneration of Cardiac Sympathetic Nerve in Parkinson’s Disease., 2008, 131,pp. 642-650. DOI: https://doi.org/10.1093/brain/awm302. PMID: https://www.ncbi.nlm.nih.gov/pubmed/18079166.

278. K. Javanshiri; T. Drakenberg; M. Haglund; E. Englund Sudden Cardiac Death in Synucleinopathies., 2023, 82,pp. 242-249. DOI: https://doi.org/10.1093/jnen/nlad001. PMID: https://www.ncbi.nlm.nih.gov/pubmed/36668680.

279. L. Grosu; A. Grosu; D. Crisan; A. Zlibut; L. Perju-Dumbrava Parkinson’s Disease and Cardiovascular Involvement: Edifying Insights., 2023, 18, 25. DOI: https://doi.org/10.3892/br.2023.1607.

280. A.C.V. de Guzman; S. Kang; E.J. Kim; J.H. Kim; N. Jang; J.H. Cho; S.S. Choi High-Glucose Diet Attenuates the Dopaminergic Neuronal Function in Elegans, Leading to the Acceleration of the Aging Process., 2022, 7,pp. 32339-32348. DOI: https://doi.org/10.1021/acsomega.2c03384.

281. P. García-Sanz; J.M.F.G. Aerts; R. Moratalla The Role of Cholesterol in a-Synuclein and Lewy Body Pathology in GBA1 Parkinson’s Disease., 2021, 36,pp. 1070-1085. DOI: https://doi.org/10.1002/mds.28396. PMID: https://www.ncbi.nlm.nih.gov/pubmed/33219714.

282. K. Javanshiri; T. Drakenberg; M. Haglund; E. Englund Cardiac Alpha-Synuclein Is Present in Alpha-Synucleinopathies., 2022, 12,pp. 1125-1131. DOI: https://doi.org/10.3233/JPD-223161. PMID: https://www.ncbi.nlm.nih.gov/pubmed/35275559.

Figures

Figure 1: Bidirectional relationship between toxic aSyn species and cellular dysfunctions. Different factors are well known to contribute to the generation of misfolded toxic aSyn species. Additionally, abnormal levels or misfolded forms of aSyn impact several aspects of neuronal function, thus contributing to the pathogenesis of PD. Created with Biorender.com. [Please download the PDF to view the image]

Figure 2: Prodromal non-motor symptoms in synucleinopathies. In the prodromal stage of synucleinopathies, several non-motor symptoms can appear up to 20 years before cardinal motor symptoms manifest. Created with Biorender.com. [Please download the PDF to view the image]

Author Affiliation(s):

[1] Department of Biology, University of Padova, 35131 Padova, Italy

[2] Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy

Author Note(s):

[*] Correspondence: [emailprotected] (G.M.M.); [emailprotected] (C.C.)

DOI: 10.3390/cells13151265

COPYRIGHT 2024 MDPI AG
No portion of this article can be reproduced without the express written permission from the copyright holder.

Copyright 2024 Gale, Cengage Learning. All rights reserved.


Alpha Synuclein Toxicity and Non-Motor Parkinson’s. (2024)

FAQs

Alpha Synuclein Toxicity and Non-Motor Parkinson’s.? ›

αSyn Toxicity. From a pathological point of view, similar to motor symptoms, NMS can occur as a consequence of the formation of intracytoplasmic Lewy bodies and neurites rich in αSyn aggregates in nigral and extranigral areas.

What causes non-motor symptoms of Parkinson's disease? ›

Parkinson's disease (PD) is a multi-systemic disorder that is characterized by a combination of motor and non-motor symptoms (NMS). The dopaminergic neurodegeneration of PD is involved in the genesis of NMS, but other conditions and side effects of levodopa are also associated with NMS.

How does alpha-synuclein affect Parkinson's? ›

α-Synuclein may contribute to PD pathogenesis in a number of ways, but it is generally thought that its aberrant soluble oligomeric conformations, termed protofibrils, are the toxic species that mediate disruption of cellular homeostasis and neuronal death, through effects on various intracellular targets, including ...

What are the spectrum of non-motor symptoms in Parkinson's disease? ›

Sleep disturbances, gastrointestinal dysfunction, bladder dysfunction, and even fatigue (extreme exhaustion) all are described by Dr. Parkinson. What has changed is the growing recognition of the prevalence and importance of non-motor dysfunction in both the diagnosis and management of PD.

What diseases are associated with alpha-synuclein? ›

Abnormal aggregates of α-syn, such as Lewy bodies (LB) and Lewy neurites, and glial cell inclusions have been then implicated in several sporadic neurodegenerative diseases termed α-synucleinopathies, including idiopathic PD, Dementia with Lewy bodies (DLB), Multiple systems atrophy (MSA), Pure autonomic failure (PAF) ...

Which of the following is the most overlooked non-motor symptom of Parkinson's disease? ›

People with PD may experience depression, apathy, and anxiety more frequently than those without Parkinson's. In fact, depression — while common in PD — is often overlooked and undertreated. It is important to be aware of these symptoms, so they can be treated effectively if you experience them.

What else could it be if not Parkinson's? ›

Several conditions fall into the category of Parkinson-like diseases. They can mimic Parkinson's symptoms, such as tremors, slow movements, and stiffness. Examples of conditions similar to Parkinson's include dementia with Lewy bodies, multiple system atrophy, and progressive supranuclear palsy.

What clears alpha-synuclein? ›

Neighboring neurons and glia can engulf and clear extracellular α-synuclein and thus contributing to the regulation of α-synuclein homeostasis in the brain.

Is alpha-synuclein toxic? ›

We show that α-syn oligomers are toxic in vivo and that α-syn oligomers might interact with and potentially disrupt membranes.

Which neurodegenerative disease is most linked to a buildup of alpha-synuclein protein in the brain? ›

Progressive dopaminergic neuronal cell loss in the substantia nigra pars compacta and widespread aggregation of the α-synuclein protein (encoded by the SNCA gene) in the form of Lewy bodies and Lewy neurites are the neuropathological hallmarks of Parkinson's disease.

What is the 5:2:1 rule for Parkinson's? ›

5- (5 times oral levodopa tablet taken/day) 2- (2 hours of OFF time/day) 1- (1 hour/day of troublesome dyskinesia) criteria have been proposed by a Delphi expert consensus panel for diagnosing advanced Parkinson's disease (PD).

What is the most disabling symptom of Parkinson's disease? ›

About half of people with PD report that fatigue is a major problem and one third say it is their most disabling symptom.

What are the odd behaviors of Parkinson's patients? ›

Healthcare providers usually refer to these symptoms as "Parkinson's disease associated psychosis." Psychosis can vary from severe confusion (disordered thinking) to seeing things that aren't there (hallucinations) to believing things that are not true (delusions).

What increases alpha-synuclein? ›

The increase in α-syntotal with the advancement of disease, was attributed by Vivacqua et al. to disease progression causing advanced synaptic and cellular damage that result in α-syn monomer release into the extracellular medium.

Is alpha-synuclein good or bad? ›

The build-up of a protein called alpha-synuclein has been considered one of the hallmarks of Parkinson's; evidence suggests alpha-synuclein may play a role in driving the loss of dopamine neurons (nerve cells) in Parkinson's as this protein forms clumps that puts stress on cells and may lead to their death.

What is the prognosis for synucleinopathy? ›

The results show clearly that the long-term outlook in these diseases depends on the particular disease. Median survival is especially poor in MSA-P (7.5 years) and MSA-C (6.4 years).

What are the non-motor symptoms of Parkinson's disease from pathophysiology to early diagnosis? ›

Neuropsychiatric manifestations include depression, anxiety, psychosis, apathy and fatigue, sleep disorders, cognitive impairment and dementia, impulse control disorders, and others.
  • Depression. ...
  • Anxiety. ...
  • Sleep disorders. ...
  • Psychosis. ...
  • Apathy. ...
  • Fatigue.

What is the non-motor symptoms scale for Parkinson's disease? ›

The Non-Motor Symptoms Scale (NMSS) is a 30-item rater-based scale to assess a wide range of non-motor symptoms in patients with Parkinson's disease (PD). The NMSS measures the severity and frequency of non-motor symptoms across nine dimensions. The scale can be used for patients at all stages of PD.

Which of the following non-motor symptoms can most likely occur as a result of Parkinson's disease (PD)? ›

Fatigue is another very common non-motor symptom of Parkinson's. It can feel like an overwhelming sense of tiredness, low energy, sleepiness, weakness or a loss of stamina when active. Fatigue may appear alongside sleep problems like restless leg syndrome or mood issues like depression and anxiety.

What are non-motor symptoms of Parkinson's disease uptodate? ›

Outline
  • Cognitive dysfunction and dementia.
  • Psychotic symptoms.
  • Mood disorders. Depression. Anxiety. Apathy and abulia.
  • Sleep disorders.
  • Excessive daytime sleepiness.
  • Fatigue.
  • Autonomic dysfunction.
  • Olfactory dysfunction.
Mar 14, 2024

Top Articles
London mayor election: How Sadiq Khan won over London for the third time
London mayor election: Sadiq Khan clinches historic third term
417-990-0201
Splunk Stats Count By Hour
Garrison Blacksmith Bench
Pieology Nutrition Calculator Mobile
Top Financial Advisors in the U.S.
P2P4U Net Soccer
According To The Wall Street Journal Weegy
Which aspects are important in sales |#1 Prospection
OnTrigger Enter, Exit ...
Premier Boating Center Conroe
Maxpreps Field Hockey
Little Rock Arkansas Craigslist
Regal Stone Pokemon Gaia
Seattle Rpz
Dr. med. Uta Krieg-Oehme - Lesen Sie Erfahrungsberichte und vereinbaren Sie einen Termin
Napa Autocare Locator
使用 RHEL 8 时的注意事项 | Red Hat Product Documentation
Ups Access Point Lockers
Graphic Look Inside Jeffrey Dahmer
Atdhe Net
Aliciabibs
Bidevv Evansville In Online Liquid
Sandals Travel Agent Login
Chamberlain College of Nursing | Tuition & Acceptance Rates 2024
Meta Carevr
Cosas Aesthetic Para Decorar Tu Cuarto Para Imprimir
Weather October 15
Movies - EPIC Theatres
Weather Underground Durham
Mchoul Funeral Home Of Fishkill Inc. Services
Shiftwizard Login Johnston
Skroch Funeral Home
Federal Student Aid
AsROck Q1900B ITX und Ramverträglichkeit
Ewwwww Gif
Metra Schedule Ravinia To Chicago
Scottsboro Daily Sentinel Obituaries
Dr Adj Redist Cadv Prin Amex Charge
Trap Candy Strain Leafly
Noaa Marine Weather Forecast By Zone
Oppenheimer Showtimes Near B&B Theatres Liberty Cinema 12
Emily Tosta Butt
Shell Gas Stations Prices
Big Reactors Best Coolant
Ups Authorized Shipping Provider Price Photos
Youravon Com Mi Cuenta
This Doctor Was Vilified After Contracting Ebola. Now He Sees History Repeating Itself With Coronavirus
Every Type of Sentinel in the Marvel Universe
Dmv Kiosk Bakersfield
Latest Posts
Article information

Author: Francesca Jacobs Ret

Last Updated:

Views: 6080

Rating: 4.8 / 5 (68 voted)

Reviews: 83% of readers found this page helpful

Author information

Name: Francesca Jacobs Ret

Birthday: 1996-12-09

Address: Apt. 141 1406 Mitch Summit, New Teganshire, UT 82655-0699

Phone: +2296092334654

Job: Technology Architect

Hobby: Snowboarding, Scouting, Foreign language learning, Dowsing, Baton twirling, Sculpting, Cabaret

Introduction: My name is Francesca Jacobs Ret, I am a innocent, super, beautiful, charming, lucky, gentle, clever person who loves writing and wants to share my knowledge and understanding with you.