Fact-checked by Grok 2 weeks ago

Alpha-synuclein

Alpha-synuclein (α-synuclein) is a small, 14 acidic protein consisting of 140 , natively unfolded in its monomeric form but capable of adopting an α-helical structure upon binding to membranes, and it is predominantly expressed in neurons of the central and peripheral nervous systems. Its structure is divided into three distinct domains: an N-terminal region (residues 1–60) rich in residues that facilitates membrane interactions, a hydrophobic non-amyloid-β component () region (residues 61–95) prone to aggregation, and a C-terminal region (residues 96–140) with acidic residues involved in calcium binding and chaperone-like activities. Under physiological conditions, α-synuclein localizes primarily to presynaptic terminals, where it regulates trafficking, promotes SNARE complex assembly to facilitate release, and contributes to membrane curvature and stabilization, thereby supporting and . It also exhibits multifaceted roles, including activity, of signaling, and suppression of in neurons, though its precise functions remain partially elusive due to the mild synaptic phenotypes observed in α-synuclein models. In pathological contexts, α-synuclein is the primary component of Lewy bodies and Lewy neurites, intraneuronal inclusions that define synucleinopathies such as , , and . Genetic mutations (e.g., A53T, E46K), duplications, or triplications of the SNCA gene encoding α-synuclein lead to familial forms of , while sporadic cases involve its misfolding and aggregation into toxic oligomeric intermediates and β-sheet-rich , which propagate prion-like across neurons and trigger synaptic dysfunction, mitochondrial impairment, and neuroinflammation. Over 90% of α-synuclein in Lewy bodies is hyperphosphorylated at serine 129 (S129), a modification that accelerates formation and is a hallmark of disease progression, with early oligomeric species disrupting striatal even at nanomolar concentrations. These aggregates primarily affect neurons in the , resulting in motor symptoms like bradykinesia and in , which impacts approximately 1% of individuals over age 60 worldwide. Beyond neurodegeneration, α-synuclein serves as a promising for early diagnosis, with altered levels in , , and biopsies detectable years before clinical onset, and the ratio of oligomeric to total α-synuclein showing high specificity for and related disorders. Therapeutic strategies targeting α-synuclein include approaches to clear aggregates, small molecules to inhibit fibrillization (e.g., anle138b), and via antisense , though challenges persist in distinguishing pathological from physiological forms. Ongoing research emphasizes its potential nuclear roles as a chaperone and interactions with proteins like parkin and , underscoring α-synuclein's central position in both health and disease.

Genetics and Discovery

Gene and Mutations

The SNCA gene, located on the short arm of at position 4q22.1, spans approximately 114 kb and consists of six exons that encode the 140-amino acid protein alpha-synuclein. This gene was first identified in as the precursor to the non-amyloid beta component (NAC) of in , with the full-length protein determined through efforts that linked it to presynaptic . The protein is derived directly from the SNCA coding region, serving as the genetic blueprint for alpha-synuclein expression in neural tissues. Pathogenic missense mutations in SNCA are rare but causative of autosomal dominant familial (), typically presenting with early onset and variable clinical features such as rapid progression, , and psychiatric symptoms. The first mutation identified was A53T in 1997, found in Italian and Greek kindreds with linked to pathology. Subsequent discoveries include A30P (1998), which alters membrane binding and promotes fibrillization; E46K (2003), associated with dementia-predominant phenotypes; H50Q (2013), linked to late-onset with good levodopa response; G51D (2013), causing atypical with ; A53E (2013), resulting in early-onset levodopa-responsive ; A53V (2013), characterized by rapid disease progression and multisystem involvement; A30G (2021), identified in Greek families with autosomal dominant and classical parkinsonian features; K58N (2025), a variant linked to typical symptoms in familial cases; G14R (2025), associated with a complex neurodegenerative including . These point mutations, occurring primarily in the C-terminal region, enhance alpha-synuclein aggregation propensity and disrupt normal protein , though their precise mechanisms vary. In addition to point mutations, genomic multiplications of SNCA, including duplications and triplications, cause autosomal dominant with dosage-dependent severity and earlier onset compared to single copy variants. Duplications, first reported in in a British family, lead to 1.5- to 2-fold overexpression of wild-type alpha-synuclein, resulting in heterogeneous phenotypes ranging from late-onset to . Triplications, identified in 2004 in a Swedish-Irish , produce even higher expression levels (2- to 3-fold), correlating with more aggressive early-onset disease, pronounced cognitive decline, and reduced in some carriers. These copy number variations underscore the role of in , with over 50 families documented to date showing non-motor features like . Regulatory elements upstream of SNCA, such as the NACP-Rep1 dinucleotide repeat polymorphism located approximately 8.8 kb from the transcription start site, modulate levels and contribute to risk. The Rep1 element acts as an enhancer, with longer alleles (e.g., 261 bp) increasing transcriptional activity up to threefold compared to shorter variants, leading to elevated alpha-synuclein mRNA and protein in regions vulnerable to . This polymorphism interacts with transcription factors like PARP-1 to fine-tune expression, and its association with sporadic highlights non-coding genetic influences on susceptibility.

Protein Sequence

Alpha-synuclein is a 140-amino acid protein with the primary MDVFMKGLSKAKEGVVAAAEKTKEGVLYVGSKTKEGVVHGVATVAEKTKEQVTNVGGAVVTGVTAVAQKTVEGAGSIAAATGFVKKDQLGKNEEGAPQEGILEDMPVDPDNEAYEMPSEGYQDYEPEA. This can be divided into three distinct regions: the N-terminal (residues 1–60), which is amphipathic and contains seven imperfect 11-residue repeats with the KTKEGV that facilitate binding; the central non-amyloid-β component () region (residues 61–95), which is hydrophobic and encompasses the aggregation-prone core GAVVTGVTAVAQKTVEGAG; and the C-terminal (residues 96–140), which is - and acidic residue-rich, promoting . The protein is subject to several post-translational modifications that influence its stability and function. N-terminal acetylation, occurring on the initiating (position 1) in nearly all cellular alpha-synuclein molecules, enhances structural stability and affinity without altering the overall disordered state. at serine 129 is prevalent, accounting for about 90% of the protein incorporated into pathological Lewy bodies, though it constitutes only 4% or less in soluble forms. at residues 39 and 125 introduces groups that can modulate oligomerization and interactions. Ubiquitination targets residues such as Lys6, Lys10, Lys21, and Lys96, marking the protein for proteasomal degradation and regulating its turnover. Alpha-synuclein exhibits strong evolutionary across vertebrates, sharing over 95% with mammalian orthologs and retaining key functional motifs in non-mammalian like , underscoring its ancient role in synaptic processes. Human-specific features, including subtle variations in the region's hydrophobicity, distinguish it from other and may contribute to its unique aggregation behavior.

Molecular Structure

Native Structure

Alpha-synuclein is a 140-amino-acid protein with a molecular weight of approximately 14.5 kDa and an (pI) of about 4.7, rendering it highly soluble under neutral physiological conditions. As an intrinsically disordered protein (), it lacks a stable tertiary structure in its soluble, unbound form, adopting a dynamic ensemble of conformations characterized by high flexibility and rapid interconversion. This disorder is essential for its physiological roles, allowing transient interactions with binding partners without a rigid scaffold. The protein's sequence can be divided into three distinct regions that contribute to its disordered nature. The N-terminal region (residues 1–60) exhibits amphipathic properties with a propensity for α-helical formation due to seven imperfect 11-residue repeats containing the KTKEGV consensus motif, though it remains largely unstructured in isolation. The central non-amyloid-β component () region (residues 61–95) is hydrophobic and has potential for β-sheet formation, but in the native state, it contributes to the overall lack of stable secondary structure. The C-terminal region (residues 96–140) is an acidic, proline-rich with high negative , promoting and inhibiting unwanted intermolecular interactions. Biophysical studies using (NMR) spectroscopy reveal an extended, coil-like conformation with narrow linewidths and limited dispersion in 2D ¹H-¹⁵N HSQC spectra, confirming the absence of persistent secondary structure elements and indicating transient helical propensities in only about 10% of residues, primarily in the . (CD) spectroscopy further supports this disorder, estimating less than 2% α-helical content and approximately 70% in solution, with characteristic minima near 200 nm rather than the helical signatures at 208 and 222 nm. A key aspect of the native structure debate concerns whether alpha-synuclein predominantly exists as a monomer or a higher-order oligomer in vivo. Seminal evidence from native gel electrophoresis, analytical ultracentrifugation, and electron microscopy indicates that it forms a dynamic, α-helically folded tetramer (~58 kDa) under physiological conditions in neurons and brain tissue, which resists aggregation by maintaining a compact, non-amyloidogenic conformation. This tetrameric form, stabilized without requiring lipids, shows CD spectra with helical minima at 208 and 222 nm and lacks thioflavin T reactivity even after prolonged incubation, contrasting with the aggregation-prone monomeric state observed in denaturing conditions. Subsequent studies have reinforced this view, suggesting that shifts toward monomers may underlie pathological aggregation in synucleinopathies.

Conformational Dynamics and Lipid Interactions

Alpha-synuclein, in its intrinsically disordered native state, undergoes significant conformational changes upon interaction with lipid membranes, particularly those mimicking synaptic vesicles. The N-terminal region, rich in amphipathic sequences, preferentially binds to anionic phospholipids such as 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoglycerol (POPG), inducing the formation of α-helices that insert into the at a shallow angle. This binding is facilitated by electrostatic interactions between positively charged and residues in the and the negatively charged lipid headgroups, leading to a structured conformation that anchors the protein to the membrane surface. The protein exhibits two primary binding modes: a U-shaped peripheral conformation, where the N- and C-terminal helices wrap around the membrane in a non-penetrating manner, and an extended helical mode that allows deeper insertion, potentially spanning the bilayer in a transmembrane-like fashion under certain conditions. These modes are influenced by lipid composition and membrane properties, with the U-shaped form predominant on planar bilayers and the extended form favored on highly curved surfaces. Alpha-synuclein displays higher binding affinity for curved membranes, such as those of small synaptic vesicles (around 25-40 nm diameter), compared to flat bilayers, enabling it to sense and respond to membrane curvature through increased hydrophobic exposure and lipid packing defects. This curvature sensitivity is crucial for its localization at synaptic sites, where it modulates vesicle dynamics. The α-helical structures are stabilized by intra- and inter-helical hydrogen bonds, which enhance the overall stability of the membrane-bound state.48926-1/fulltext) The interaction dynamics involve rapid exchange between membrane-bound and unbound states, occurring on timescales of milliseconds to seconds, allowing alpha-synuclein to dynamically associate and dissociate without long-term entrapment. This fast kinetics is driven by the protein's low barrier and is modulated by lipid headgroup charge and bilayer fluidity, ensuring responsiveness to changing cellular environments. Experimental evidence from () has demonstrated that membrane-bound alpha-synuclein forms parallel-oriented dimers, with the N-terminal helices aligning side-by-side on the surface to facilitate multimerization. Cryo-electron (cryo-EM) studies further support this, revealing extended helical dimers in close proximity to curved bilayers, confirming the structural basis for these interactions.

Tissue Expression

Central Nervous System Expression

Alpha-synuclein, encoded by the SNCA gene, exhibits high expression within the , primarily in neurons across various regions. It is most abundant in the , , , , and , where the protein concentrates in presynaptic terminals of both excitatory and inhibitory synapses. Immunohistochemical studies demonstrate a characteristic punctate localization in these presynaptic compartments, reflecting association with synaptic vesicles and membranes. SNCA mRNA transcripts are prominently detected in dopaminergic neurons of the substantia nigra, with protein levels enriched in their presynaptic terminals. In these neurons, alpha-synuclein constitutes a significant portion of the cytosolic , comprising up to 1% of total soluble protein in extracts. This neuronal predominance underscores its specialized localization, with minimal contribution from other cellular compartments in healthy tissue. Expression of alpha-synuclein follows a developmental timeline marked by upregulation during neuronal . In NTera2 teratocarcinoma cells induced to differentiate into neurons, SNCA mRNA and protein levels increase substantially over 4–6 weeks, paralleling synaptic maturation. Similarly, in primary hippocampal cultures, alpha-synuclein protein emerges around 6 days in vitro, peaks at 1 week, and stabilizes in adulthood with exclusive presynaptic enrichment by 3 weeks. This pattern aligns with broader brain development, where expression escalates postnatally to adult peaks in mature neural circuits. Regulation of SNCA expression involves transcription factors such as Nurr1, which binds to its promoter to drive transcription in neurons. Nurr1-mediated ensures sustained levels during neuronal , particularly in the . In glial cells, including and , alpha-synuclein expression remains low or undetectable, distinguishing it from neuronal abundance.

Peripheral and Non-Neuronal Expression

Alpha-synuclein is expressed in the (ENS), where it is particularly abundant in the of the . Studies have identified high levels of alpha-synuclein mRNA and protein in enteric neurons, with localization primarily in the and processes of these cells. This expression pattern in the ENS has been linked to its potential involvement in the gut-brain axis, though the protein's distribution varies across different regions of the gut. Beyond the ENS, alpha-synuclein is detectable in various peripheral tissues, including platelets, red blood cells, adrenal glands, and the heart. Platelets contain significant amounts of alpha-synuclein, where it associates with vesicular structures and is released upon . Similarly, red blood cells contain alpha-synuclein, with the protein comprising a notable portion of cytosolic content in erythrocytes. In the adrenal glands and heart, alpha-synuclein is found in neuroendocrine and cardiac tissues, respectively, at levels sufficient for detection in tissue homogenates. In non-neuronal cells, alpha-synuclein expression is generally low in and , contrasting with its predominance in neurons of the . However, elevated expression has been observed in cells, where the protein is highly abundant and localized to the and . Quantitative assessments reveal that alpha-synuclein protein is detectable in cerebrospinal fluid (CSF), blood, and saliva, with tissue-specific variations in concentration and notable assay variability due to factors like platelet contamination. Levels in neural tissues remain the highest, while peripheral fluids show measurable amounts, such as approximately 1-10 ng/mL in plasma and ~1-2 ng/mL in CSF (as of 2023). In blood cells, total alpha-synuclein concentrations can reach up to 100-fold higher than in saliva or CSF, reflecting its enrichment in hematopoietic elements like platelets and erythrocytes.

Physiological Functions

Synaptic and Vesicular Roles

Alpha-synuclein is predominantly localized to presynaptic terminals, where it plays essential roles in maintaining and facilitating release. It interacts directly with to regulate their trafficking and clustering, thereby influencing the readily releasable pool of vesicles. Specifically, alpha-synuclein binds to membranes of , promoting their assembly and preventing excessive fusion, which ensures efficient vesicle recycling during repeated synaptic activity. A key function of alpha-synuclein involves chaperoning the assembly of the SNARE complex, critical for . It directly binds to VAMP2 (also known as synaptobrevin), a v-SNARE protein on , and facilitates the formation of the SNARE complex with syntaxin-1 and SNAP-25 on the plasma membrane. This interaction enhances SNARE complex stability and accelerates vesicle priming, thereby supporting sustained neurotransmitter release. and studies demonstrate that alpha-synuclein acts as a nonclassical chaperone, lowering the energy barrier for SNARE zippering without being part of the final complex. Alpha-synuclein also modulates neurotransmission by influencing vesicle mobilization and reuptake mechanisms. In dopaminergic terminals, it inhibits the (), reducing reuptake and prolonging extracellular availability, which affects short-term such as paired-pulse facilitation. Knockout mice lacking alpha-synuclein exhibit altered vesicle mobilization, with impaired replenishment of the readily releasable pool and reduced evoked , particularly under high-frequency stimulation, highlighting its role in maintaining synaptic reserve. These models show normal basal but deficits in short-term , underscoring alpha-synuclein's regulatory influence on dynamics. Furthermore, alpha-synuclein cooperates with cysteine-string protein alpha (CSPα), a chaperone, to maintain vesicle integrity and prevent neurodegeneration. This interaction stabilizes CSPα on synaptic vesicles, supporting proper SNARE function and vesicle maintenance during aging. In CSPα knockout mice, transgenic expression of alpha-synuclein partially rescues synaptic defects by enhancing SNARE complex assembly, indicating their synergistic roles in presynaptic maintenance.

Proneurogenic and Regulatory Functions

Alpha-synuclein contributes to by modulating the proliferation and of neural stem cells in the adult brain. In the , studies using alpha/beta-synuclein double models show that absence of these synucleins leads to increased neuronal , indicating a role in fine-tuning neurogenic processes. Overexpression of wild-type alpha-synuclein in cultured cortical neurons from alpha-synuclein mice enhances main length by approximately 39% (from 108.5 μm to 150.4 μm) and increases collateral branching by about 6.5-fold compared to controls, suggesting a physiological function in promoting neurite outgrowth and axonal arborization during neuronal development. Conversely, ablation of alpha-synuclein impairs synaptic function and overall neuronal maturation, as seen in models exhibiting synaptic abnormalities and reduced density, underscoring its regulatory necessity for proper . Nuclear localization of alpha-synuclein enables its involvement in transcriptional regulation, where it interacts with DNA and histones to influence gene expression critical for neuronal differentiation. Alpha-synuclein forms complexes that promote protein arginine methyltransferase 5 (PRMT5)-mediated symmetric dimethylation of histone H4 at arginine 3 (H4R3me2s), facilitating epigenetic modifications that control neuronal gene transcription and development. This nuclear activity modulates profiles of genes involved in epigenetic alterations, supporting the stability and differentiation of neural cells under physiological conditions. At physiological levels, alpha-synuclein exerts neuroprotective effects through mechanisms and mitochondrial stabilization. It functions as an by inhibiting in neuronal membranes, preventing oxidative damage from reactive species and thereby protecting cellular integrity. Upregulation of alpha-synuclein in neurons exposed to chronic low-level correlates with reduced and enhanced cell survival, highlighting its protective role against oxidative insults. Additionally, physiological expression stabilizes mitochondrial adaptor proteins like , supporting mitochondrial quality control and transport essential for neuronal health.

Autoproteolytic Activity

Alpha-synuclein exhibits intrinsic autoproteolytic activity, enabling self-cleavage without the involvement of external proteases. This process occurs primarily at the bond between Val71 and Thr72 within the central amyloidogenic domain (residues 61-93). studies demonstrate that alpha-synuclein undergoes self-cleavage when incubated at 37°C in neutral (sodium-phosphate, pH 7.5) for extended periods, such as 14-25 days, under conditions that mimic cellular stress and promote conformational changes. Evidence from ion mobility (IMS-MS), tandem MS sequencing, and confirms the generation of proteolytic fragments, with controls using protease inhibitors verifying the absence of contaminating enzymatic activity. The mechanism likely involves transient structured intermediates in the intrinsically disordered protein, where the N-terminal region may facilitate initial unfolding to expose the cleavage site. Key fragments produced include the C-terminal piece αSyn(72-140), with a mass of 7274 Da, alongside N-terminally truncated variants such as αSyn(7-140) and αSyn(40-140). These truncations, particularly αSyn(72-140), exhibit accelerated oligomerization and aggregation compared to full-length alpha-synuclein, suggesting a role in generating aggregation-prone species during protein processing. Post-translational modifications, such as potential N-terminal truncations, may initiate or enhance this activity by altering the protein's conformational dynamics. Although primarily observed in vitro, this autoproteolytic processing contributes to alpha-synuclein's turnover under stress, with implications for amplified fragment generation in aging-related conditions where protein is compromised.

Pathological Mechanisms

Aggregation and Fibril Formation

Alpha-synuclein aggregation proceeds through a series of conformational transitions from its intrinsically disordered monomeric state to soluble oligomers and insoluble , a process central to the of synucleinopathies. These are rich in β-sheet secondary structure, contrasting with the protein's physiological α-helical conformations upon lipid . Oligomeric intermediates, often transient and heterogeneous, serve as precursors to formation and are considered highly toxic . The molecular mechanism of aggregation is best described by the , in which primary —the slow, rate-limiting association of monomers into stable seeds—initiates the process, followed by rapid as monomers add to ends. A key feature for alpha-synuclein is secondary , where existing surfaces catalyze the breakage or templating of new oligomeric nuclei from solution-phase monomers, leading to autocatalytic amplification and of aggregates. This secondary process dominates under physiological conditions, as evidenced by kinetic studies showing its strong dependence on concentration and surface properties. Structurally, alpha-synuclein adopt a cross-β , with β-strands arranged in an in-register parallel fashion to form the core. A representative high-resolution reveals a Greek key topology spanning residues approximately 42–98, featuring four β-strands connected by loops and stabilized by hydrophobic interactions (PDB: 2N0A). are polymorphic, exhibiting strain-specific variations in protofilament packing, twist, and core extent; for instance, some polymorphs consist of two protofilaments forming a 10 diameter , while others show twisted or flat morphologies that influence seeding efficiency. Several triggers accelerate aggregation by altering the protein's conformational landscape or kinetics. Post-translational modifications, particularly phosphorylation at serine 129 (pSer129), promote fibril formation by enhancing β-sheet propensity and reducing solubility, with nearly 90% of alpha-synuclein in pathological inclusions being modified at this site. Metal ions such as Cu²⁺ and Fe³⁺ bind to the C-terminal region, inducing partial folding and that facilitate misfolding. Acidic pH environments, common in stressed cellular compartments, dramatically increase secondary rates by protonating residues and exposing hydrophobic surfaces. The non-amyloid-β component () region (residues 61–95) serves as the primary hydrophobic core driving intermolecular interactions essential for oligomerization and fibrillogenesis. In vitro aggregation kinetics typically follow a sigmoidal trajectory: an initial lag phase dominated by primary (often hours to days), an elongation phase where grow rapidly, and a final plateau as monomers are depleted. These phases are quantitatively assessed using Thioflavin T (ThT) fluorescence assays, where ThT binds specifically to cross-β structures, providing real-time monitoring of assembly with fluorescence intensity correlating to aggregate mass.

Propagation and Seeding

Alpha-synuclein exhibits prion-like propagation, wherein misfolded aggregates are released from affected cells and taken up by neighboring cells, thereby amplifying across neural networks. This process involves the extracellular release of alpha-synuclein primarily through exosomes, small membrane-bound vesicles derived from multivesicular bodies within the endosomal pathway. Studies in neuronal cell lines have demonstrated that monomeric and oligomeric forms of alpha-synuclein are packaged into exosomes in a calcium-dependent manner, facilitating their under physiological and pathological conditions. Additionally, alpha-synuclein can be released via non-exosomal mechanisms, such as direct leakage from damaged cells or bulk flow, though exosomal packaging enhances stability and uptake efficiency. Once released, alpha-synuclein aggregates are internalized by recipient cells through , including receptor-mediated pathways involving sulfate proteoglycans, or via direct cytoplasmic transfer through tunneling nanotubes (TNTs), actin-based structures connecting cells. TNTs enable the transport of fibrillar alpha-synuclein within lysosomal vesicles, promoting efficient neuron-to-neuron and neuron-to-glia transmission. Glial cells, particularly and , actively participate in this propagation; for instance, can engulf and redistribute alpha-synuclein aggregates via TNTs, exacerbating spread to neurons. This intercellular transmission supports the observed patterned progression of pathology in synucleinopathies, as exemplified by in , where inclusions ascend from the and dorsal motor nucleus of the vagus to higher and cortical regions, potentially via vagal and sympathetic pathways. Central to propagation is the seeding mechanism, where internalized act as templates to induce misfolding of endogenous soluble alpha-synuclein monomers, leading to aggregate formation. This templated conformational change was first demonstrated in cultured cells, where exogenous preformed triggered the assembly of intracellular Lewy body-like inclusions resembling those in . Seeding efficiency varies with fibril structure, giving rise to strain-specific : distinct alpha-synuclein conformers, differing in core architecture or post-translational modifications like at serine 129, propagate unique pathologies with varying and regional . evidence from mouse models underscores this; intracerebral injection of synthetic or brain-derived alpha-synuclein into wild-type mice induces progressive, prion-like spread of phosphorylated inclusions from the injection site to anatomically connected regions, mimicking over months. These models confirm that and occur independently of transgenic overexpression, relying on endogenous alpha-synuclein.

Mitochondrial and Cellular Toxicity

Alpha-synuclein (α-syn) interacts directly with mitochondria, primarily through binding to the translocase of the outer mitochondrial membrane (TOM) complex, particularly TOM20, which facilitates its import into the organelle. This interaction disrupts the TOM20-TOM22 co-receptor association, impairing the import of nuclear-encoded mitochondrial proteins and leading to mitochondrial dysfunction. Once imported, α-syn accumulates within the mitochondria, where it binds to inner membrane components and inhibits the activity of , reducing ATP production and bioenergetic capacity. The mitochondrial toxicity of α-syn manifests through multiple pathways, including excessive production of (ROS), which arises from complex I inhibition and subsequent leakage. This exacerbates cellular damage, while α-syn also perturbs mitochondrial dynamics by inhibiting fusion proteins such as mitofusin and promoting fission via Drp1 hyperactivation, resulting in fragmented mitochondria. Additionally, α-syn dysregulates calcium by altering (ER)-mitochondria contact sites, leading to aberrant calcium influx into mitochondria, , and further ROS generation. Beyond mitochondria, α-syn induces broader cellular toxicity, including ER stress through activation of the unfolded protein response and accumulation of misfolded proteins. It also impairs lysosomal function by disrupting and enzyme activity, such as , which hinders the degradation of accumulated material. Furthermore, α-syn inhibits , particularly macroautophagy and , by sequestering key regulators like TFEB and blocking autophagosome-lysosome fusion, leading to defective clearance of damaged organelles. These effects collectively compromise cellular homeostasis and contribute to neuronal vulnerability. Recent studies, including those from 2025, have highlighted α-syn's role in mitophagy failure, where phosphorylated forms of the protein induce mitochondrial damage and block /Parkin-mediated mitophagic flux, preventing the removal of dysfunctional mitochondria. In alpha-synuclein () models, such as those in mice, cells exhibit enhanced mitochondrial resilience to stressors, with preserved complex I activity and reduced ROS under pathological conditions, underscoring α-syn's pathological necessity for toxicity. These findings from studies demonstrate that loss of α-syn mitigates mitochondrial fragmentation and bioenergetic deficits observed in models.

Clinical Significance

Role in Synucleinopathies

Alpha-synuclein is a central pathological protein in synucleinopathies, a group of neurodegenerative disorders characterized by the accumulation of its misfolded aggregates in the central and peripheral nervous systems. These diseases include (PD), (DLB), and (MSA), where alpha-synuclein forms intraneuronal or intraglial inclusions that contribute to progressive neuronal dysfunction and loss. In , alpha-synuclein aggregates primarily as Lewy bodies and Lewy neurites within of the , leading to degeneration and the hallmark motor symptoms of bradykinesia, rigidity, and resting . These inclusions were first identified as containing alpha-synuclein in postmortem brain tissue from idiopathic PD cases, establishing it as the primary structural component of Lewy pathology. The loss of nigral correlates with the severity of motor impairment, though alpha-synuclein pathology often extends beyond the to other brain regions in advanced stages. Dementia with Lewy bodies features widespread cortical alpha-synuclein inclusions, including Lewy bodies in neocortical and limbic areas, which underlie cognitive decline, visual hallucinations, and fluctuating attention alongside parkinsonian features. Unlike , where pathology is more subcortical, DLB shows a predominance of cortical involvement, with alpha-synuclein aggregates disrupting synaptic function and contributing to early dementia. Multiple system atrophy is distinguished by alpha-synuclein-positive glial cytoplasmic inclusions (GCIs), primarily in of the , , and , leading to multisystem neurodegeneration and prominent autonomic failure such as and . These GCIs, also known as Papp-Lantos bodies, represent a non-neuronal form of alpha-synuclein aggregation unique to , correlating with the loss of neurons in affected regions and the development of , , or . Synucleinopathies can arise sporadically or through genetic mechanisms; in familial cases, multiplications of the SNCA gene encoding alpha-synuclein lead to its overexpression and early-onset with rapid progression, as seen in pedigrees with duplications or triplications. Sporadic forms, comprising the majority, follow a predictable progression of alpha-synuclein according to the Braak , which posits a caudal-to-rostral spread starting in the dorsal motor nucleus of the vagus and ascending to the and . Recent studies from 2024 and 2025 have strengthened links between alpha-synuclein pathology and prodromal conditions like (PAF) and isolated behavior disorder (iRBD), where phosphorylated alpha-synuclein deposits in peripheral nerves and structures presage conversion to overt synucleinopathies such as or . In iRBD, alpha-synuclein aggregates in nuclei correlate with dream-enacting behaviors and predict phenoconversion to synucleinopathies with over 90% lifetime risk.

Biomarkers and Diagnostics

Seed amplification assays (SAAs), particularly (RT-QuIC), have emerged as highly sensitive methods for detecting misfolded alpha-synuclein in (CSF) and skin biopsies. In CSF, RT-QuIC identifies pathological alpha-synuclein seeding activity with sensitivities of 84-95% and specificities exceeding 90% in (PD) patients compared to healthy controls. Similarly, skin RT-QuIC on punch biopsies demonstrates diagnostic reliability, achieving 90-100% sensitivity and 95-100% specificity for synucleinopathies including PD and (DLB). These assays amplify minute quantities of misfolded alpha-synuclein, enabling early detection in at-risk individuals such as those with isolated REM sleep behavior disorder. Fluid-based biomarkers, including phosphorylated forms like pSer129 alpha-synuclein, provide additional insights into alpha-synuclein pathology. In CSF, pSer129 alpha-synuclein levels are often elevated in early , serving as a marker of aggregation, though total alpha-synuclein concentrations may decrease due to into aggregates. neuronal-derived alpha-synuclein, particularly in exosomes, correlates with disease progression and cognitive decline, with elevated levels distinguishing from controls in longitudinal studies. Seed-dependent assays in plasma further enhance detection of propagative forms, though they exhibit variability from peripheral sources. Positron emission tomography (PET) imaging with tracers targeting alpha-synuclein aggregates represents a non-invasive approach. The tracer [18F]ACI-12589 binds specifically to pathological aggregates, showing uptake in and cortical regions of PD, DLB, and (MSA) patients, with signal retention up to 120 minutes post-injection. This tracer differentiates MSA from other synucleinopathies and non-alpha-synuclein disorders, aiding . Recent advances include blood-based achieving over 90% sensitivity for in 2025 studies, using enhanced serum protocols to detect activity non-invasively. biopsies for phosphorylated alpha-synuclein have also improved, with detection rates of 92.7% in confirmed cases and near-100% specificity against controls. Despite these developments, challenges persist in specificity, particularly distinguishing alpha-synuclein from co-aggregates like amyloid-beta or in , where may yield false positives due to cross-. Standardization across assays and validation in diverse populations remain critical for clinical adoption.

Protein Interactions

Lipid Membrane Interactions

Alpha-synuclein exhibits a strong affinity for lipid membranes containing anionic phospholipids, such as (PS) and (PI), due to electrostatic interactions between the protein's positively charged N-terminal residues and the negatively charged headgroups. This preference is particularly pronounced in liquid-disordered domains, where alpha-synuclein binds with higher affinity compared to neutral or zwitterionic lipids like . modulates these interactions by altering packing and fluidity; elevated levels typically reduce binding efficiency by promoting a more ordered environment that hinders insertion. Binding to lipid vesicles promotes the assembly of alpha-synuclein into higher-order multimers on the surface, driven by adsorption mechanisms with an energetic coupling of approximately -8 kJ/mol per binding step. The stoichiometry of this multimeric binding depends on the vesicle size, composition, and protein-to- ratio, often involving 8 or more monomers per small unilamellar vesicle, which collectively deform the into curved structures like ellipsoids or invaginations. These multimers influence by preferentially associating with fluid-phase , thereby enhancing local and potentially rigidifying or fluidizing regions based on the degree of protein crowding. In pathological conditions, alpha-synuclein aggregates compromise lipid membrane integrity by forming pores or perforations in bilayers, resulting in the leakage of cellular contents including calcium ions, enzymes, and other solutes. This disruptive activity is exacerbated in endolysosomal compartments, where aggregate-induced damage accelerates fibril release into the , promoting further seeding and . Molecular dynamics simulations illustrate that alpha-synuclein penetrates membranes via insertion of its amphipathic N-terminal helices into the acyl chain region, with the extent of burial reaching up to 10-15 Å and stabilizing the bound conformation through hydrophobic and electrostatic forces. Complementing these insights, cryo-electron microscopy structures resolved in 2022 reveal - co-assemblies where specific anionic phospholipids like 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphate (POPA) coordinate with surfaces, influencing the polymorphic and aggregation of alpha-synuclein.

Key Protein Partners

Alpha-synuclein (α-synuclein) engages in numerous protein-protein interactions that modulate its function and aggregation propensity in both healthy and diseased states. These interactions are critical for understanding its role in synaptic regulation and neurodegeneration, with key partners including components of the , molecular chaperones, and disease-associated proteins. In physiological contexts, α-synuclein interacts with synaptobrevin-2 (VAMP2), a core , to facilitate and clustering. This binding enhances SNARE complex assembly and promotes efficient release, as demonstrated in co-immunoprecipitation studies from neuronal models. Additionally, α-synuclein associates with , a ; monomeric forms can inhibit tau fibrillization , potentially supporting under normal conditions. These interactions underscore α-synuclein's role in maintaining synaptic integrity and cytoskeletal dynamics. Molecular chaperones such as and parkin serve as quality control partners for α-synuclein in physiological settings. binds to α-synuclein monomers and oligomers, inhibiting formation and promoting refolding or degradation to prevent misfolding. Parkin, an , interacts with α-synuclein to facilitate its ubiquitination and proteasomal clearance, ensuring in neurons. These chaperone-mediated interactions are essential for cellular and are disrupted in aging or stress conditions. In pathological contexts, α-synuclein forms aberrant complexes with proteins like and beta-amyloid (Aβ), exacerbating synucleinopathies. , a implicated in , phosphorylates α-synuclein at serine 129, enhancing its aggregation and , as evidenced by in vitro kinase assays and brain tissue analyses from affected individuals. Similarly, α-synuclein interacts with Aβ in mixed pathologies such as and , where the proteins co-aggregate and amplify fibril formation and neuronal damage through synergistic seeding mechanisms. Methods for identifying these partners include yeast two-hybrid screening, co-immunoprecipitation (co-IP), and advanced approaches. Yeast two-hybrid and co-IP have classically revealed direct binders like VAMP2 and , while recent affinity purification-mass spectrometry (AP-MS) studies from 2020–2025 have mapped over 100 interactors, highlighting networks in synaptic and mitochondrial compartments. These functional impacts often involve modulation of post-translational modifications; for instance, LRRK2-driven stabilizes toxic oligomers, whereas parkin-mediated ubiquitination targets them for , influencing overall proteotoxic burden.

References

  1. [1]
    Alpha-synuclein in Parkinson's disease and other synucleinopathies
    Mar 1, 2023 · Alpha-synuclein structure and physiological function. A-syn is a small (14 KDa) acidic protein expressed in neurons of the central and ...Alpha-Synuclein Structure... · Dementia With Lewy Bodies · Biomarkers
  2. [2]
    Alpha-synuclein structure and Parkinson's disease - PubMed Central
    Jul 22, 2019 · A current consensus is that αS functions to promote membrane curvature, thereby contributing to synaptic trafficking and vesicle budding [38, 39] ...α-Synuclein: Native... · Fibril Structure And Early... · Polymorphic Amyloids - Rods...
  3. [3]
    Alpha-synuclein structure, functions, and interactions - PMC
    May 9, 2016 · As a molecular biomarker for the detection of PD in its earlier stages, alpha-synuclein (α-syn), which is a key component of Lewy bodies, in ...Alpha-Synuclein... · Alpha-Synuclein Functions · Alpha-Synuclein-Protein...
  4. [4]
    6622 - Gene ResultSNCA synuclein alpha [ (human)] - NCBI
    Sep 14, 2025 · The G51D SNCA mutation generates a slowly progressive alpha-synuclein strain in early-onset Parkinson's disease. Cryo-EM structure of amyloid ...
  5. [5]
    Entry - *163890 - SYNUCLEIN, ALPHA; SNCA - OMIM
    ▻ Mapping. Hartz (2010) mapped the SNCA gene to chromosome 4q22.1 based on an alignment of the SNCA sequence (GenBank L36675) with the genomic sequence (GRCh37) ...
  6. [6]
    Alpha-synuclein structure and Parkinson's disease – lessons and ...
    Jul 22, 2019 · Mutations typically result in reduced phospholipid binding, as for example in G51D, A30P [68, 69] and A53E [70] variants. In contrast E46K and ...α-Synuclein: Native... · Fibril Structure And Early... · Polymorphic Amyloids - Rods...
  7. [7]
    Effect of allelic variation at the NACP–Rep1 repeat upstream of the α ...
    In this study we explore the role of the region upstream of the SNCA gene in regulation of α-synuclein expression and further show the contribution of the ...Effect Of Allelic Variation... · Luciferase Reporter... · Acknowledgements
  8. [8]
    SNCA - Alpha-synuclein - Homo sapiens (Human) | UniProtKB
    P37840-1. This isoform has been chosen as the canonical sequence. All positional information in this entry refers to it. This is also the sequence that ...
  9. [9]
    The Roles of Post-translational Modifications on α-Synuclein in the ...
    In this review, we will summarize physiological and pathological roles of α-synuclein, emphasizing the involvement of PTMs. Structure of α-Synuclein. In ...Abstract · Aggregation of α-Synuclein · Toxicity of α-Synuclein · Post-Translational...
  10. [10]
    How important is the N-terminal acetylation of alpha-synuclein for its ...
    Nov 16, 2022 · N-α-acetylation is a frequently occurring post-translational modification in eukaryotic proteins. It has manifold physiological consequences ...
  11. [11]
    The role of Ser129 phosphorylation of α-synuclein in ... - PubMed
    Approximately 90% of α-syn deposited in Lewy bodies is phosphorylated at serine 129 (Ser129). In contrast, only 4% or less of total α-syn is phosphorylated at ...
  12. [12]
    Preferentially increased nitration of α-synuclein at tyrosine-39 ... - NIH
    We found a nine-fold increase (relative to controls) in levels of 3NT at Tyr-39 of α-synuclein in an inducible transgenic cellular model of Parkinson's disease.
  13. [13]
    Ubiquitination of alpha-synuclein - PubMed
    We investigate ubiquitination of alpha-synuclein in vitro and in vivo and report the ubiquitination sites and the effects of familial PD-linked mutations.
  14. [14]
    The Parkinson Disease gene SNCA: Evolutionary and structural ...
    Apr 15, 2016 · This study aims to decipher evolutionary and structural insights of SNCA by revealing its sequence and structural evolutionary patterns among sarcopterygians.<|control11|><|separator|>
  15. [15]
    Evidence of Native α-Synuclein Conformers in the Human Brain - PMC
    α-Synuclein is a soluble protein consisting of 140 amino acids with a predicted molecular mass of 14,460.16 Da and an isoelectric point of 4.67. α-Synuclein ...
  16. [16]
    Biophysical characterization of α-synuclein and its controversial ...
    Extensive biophysical investigations have provided evidence that isolated α-synuclein is an intrinsically disordered protein (IDP) in vitro. Subsequently ...<|control11|><|separator|>
  17. [17]
    Biophysical characterization of α-synuclein and its controversial ...
    Analysis of the CD spectrum of free α-synuclein led to estimates of less than 2% α-helical and nearly 70% random coil content in solution.
  18. [18]
    α-Synuclein occurs physiologically as a helically folded tetramer that ...
    Our experiments provide multiple, independent lines of evidence that endogenous cellular αSyn exists in large part as an α-helically folded, ∼58 kDa tetramer ...
  19. [19]
    α-synuclein Binding & Conformation: Single-Molecule Fluorescence
    We studied the coupled binding and folding of α-synuclein, an intrinsically disordered protein linked with Parkinson's disease.
  20. [20]
    The N-Terminus of the Intrinsically Disordered Protein α-Synuclein ...
    Amino-acid sequence of α-synuclein. The N-terminal region is blue, the aggregation-prone (NAC) domain is in green, and the C-terminal region is in red. The ...
  21. [21]
    Structure of membrane-bound α-synuclein from site-directed ... - PNAS
    Dec 16, 2008 · Collectively, our data indicate that α-synuclein forms an extended helical structure when bound to phospholipid bilayer membranes but that it ...
  22. [22]
    Multiple tight phospholipid-binding modes of α-synuclein revealed ...
    In dopaminergic neurons, a-synuclein (αS) partitions between a disordered cytosolic state and a lipid-bound state. Binding of αS to membrane phospholipids ...
  23. [23]
    The Behavior of α-Synuclein in Neurons
    55. Rapid exchange of synuclein between bound and unbound states may provide a mechanism to ensure that stable cellular structures remain dynamic and ...
  24. [24]
    α-Synuclein assembles into higher-order multimers upon ... - PNAS
    We demonstrate that α-synuclein multimerizes during membrane binding and that the membrane-bound, multimeric form of α-synuclein mediates SNARE complex ...
  25. [25]
    Pathological and physiological functional cross-talks of α-synuclein ...
    In the brain, αSyn is preferentially expressed in the neocortex, hippocampus, substantia nigra, thalamus, and cerebellum, where it binds presynaptic vesicles ...
  26. [26]
    The Synaptic Function of α-Synuclein - PMC - PubMed Central - NIH
    In parallel, α-synuclein was identified as the non-amyloid-β component (NAC) found in amyloid plaques of Alzheimer's disease patients [2].
  27. [27]
    α-Synuclein at the Presynaptic Axon Terminal as a Double-Edged ...
    Mar 27, 2022 · In the presynaptic axon terminals, α-syn behaves as a soluble cytosolic protein, with about 80% of the protein reportedly bound to vesicles [7].
  28. [28]
    Novel Therapeutic Horizons: SNCA Targeting in Parkinson's Disease
    Aug 6, 2024 · Additionally, studies have shown higher levels of SNCA mRNA in the midbrain tissues and substantia nigra dopaminergic neurons of sporadic PD ...
  29. [29]
    The contribution of alpha synuclein to neuronal survival and function
    α‐syn is expressed in high concentrations within neural tissues where it primarily localizes to the presynaptic terminal (Iwai et al. 1995) (Fig. 1a). Some ...
  30. [30]
    Expression of α-synuclein is regulated in a neuronal cell type ...
    Oct 25, 2018 · However, α-synuclein is expressed only in excitatory synapses of the affected regions, such as the cerebral cortex, hippocampus, thalamus, and ...
  31. [31]
    Alpha-synuclein expression is up-regulated in NTera2 cells during ...
    Alpha-synuclein expression is up-regulated in NTera2 cells during neuronal differentiation but unaffected by exposure to cytokines and neurotrophic factors.Missing: development | Show results with:development
  32. [32]
    Synucleins Are Developmentally Expressed, and ... - PubMed Central
    Our data suggest that α-synuclein may interact with and regulate specific pools of synaptic vesicles, thereby modulating synaptic functions in the normal brain.
  33. [33]
    Advancing Stem Cell Models of Alpha-Synuclein Gene Regulation ...
    Apr 9, 2018 · SNCA-tri overexpresses alpha-synuclein and expression increases during in vitro neuronal differentiation. SNCA-tri neurons fail to develop ...
  34. [34]
    Nurr1 transcriptionally regulates the expression of alpha-synuclein.
    Nurr1 was shown to be involved in the regulation of alpha-synuclein, as decreased expression of Nurr1, which has been found in Parkinson's disease patients with ...Missing: SNCA | Show results with:SNCA
  35. [35]
    The role of Nurr1 in the development of dopaminergic neurons and ...
    Nurr1, a transcription factor belonging to the orphan nuclear receptor superfamily, is critical in the development and maintenance of the dopaminergic system.
  36. [36]
    Distinct ultrastructural phenotypes of glial and neuronal alpha ...
    Since aSyn is abundantly expressed in neurons13-15 and has only low expression levels in oligodendrocytes,16-19 the source and abundance of accumulated aSyn in ...Fibrillar Glial Cytoplasmic... · Neuronal Cytoplasmic... · Alpha-Synuclein Positive...
  37. [37]
    Alpha-Synuclein Expression in the Oligodendrocyte Lineage
    Notably, we observed a significant decrease in ΑSYN during oligodendrocyte maturation. Additionally, we show the presence of transcripts in PDGFRΑ/CD140a+ cells ...
  38. [38]
    The Baseline Structure of the Enteric Nervous System and Its Role in ...
    Native α-synuclein is considered to be abundant in the myenteric plexus of young adult rats. ... alpha-synuclein display alterations in colonic myenteric ...
  39. [39]
    Expression pattern and localization of alpha-synuclein in the human ...
    The aim of the study was to determine the gene expression pattern and localization of α-syn/p-α-syn in the human enteric nervous system (ENS).
  40. [40]
    Alpha-Synuclein Accumulation and Its Phosphorylation in the ...
    Nov 23, 2020 · The deposition of pS129α-Syn was observed only in the submucosa and the myenteric plexus and was detected in the cytoplasm of neurons. We used ...
  41. [41]
    SNCA protein expression summary - The Human Protein Atlas
    Synuclein alpha. Gene name i. Official ... Cell type enhanced (Erythroid cells, Melanocytes, Excitatory neurons, Oligodendrocytes, Basal respiratory cells).
  42. [42]
    Initial assessment of α-synuclein structure in platelets - PMC
    α-synuclein is also found in the hematopoietic system where it is expressed in erythroid precursors and megakaryocytes in bone marrow, as well as erythrocytes ...
  43. [43]
    Physiology, Synuclein - StatPearls - NCBI Bookshelf
    Alpha-synuclein is predominantly found in the nervous system, composing 1% of total cytosolic protein. Alpha-synuclein is also abundant in erythrocytes and ...
  44. [44]
    Cell Biology and Pathophysiology of α-Synuclein - PMC
    αa-Synuclein was identified in the electric organ of Torpedo californica using an antibody to purified cholinergic vesicles (Fig. 1) (Maroteaux et al. 1988). In ...
  45. [45]
    Alpha-Synuclein in Peripheral Tissues as a Possible Marker for ...
    It has been described that human melanoma cell lines have a high expression of aSyn, with this protein being undetectable in non-melanoma skin cancer cell lines ...
  46. [46]
    Salivary total α-synuclein, oligomeric α-synuclein and SNCA ...
    Jun 23, 2016 · The levels of α-synuclein in serum and plasma are up to 10,000-fold higher than in CSF as given above; Red blood cells (RBCs) contain high ...
  47. [47]
    Total α-synuclein levels in human blood cells, CSF, and saliva ...
    Total α-Syn levels in whole blood cells (WBC), cerebrospinal fluid (CSF), and saliva were determined by the lipid-ELISA method. Keywords: Biomarkers; ELISA; ...Missing: tissues | Show results with:tissues
  48. [48]
    α-Synuclein in synaptic function and dysfunction - PubMed - NIH
    Dec 23, 2022 · Here, we present an overview of the effects of α-synuclein on SNARE-complex assembly, neurotransmitter release, and synaptic vesicle pool ...
  49. [49]
    Structural basis of synaptic vesicle assembly promoted by α-synuclein
    Sep 19, 2016 · This observation can be explained by the higher affinity of αS for significantly curved membrane surfaces, which increases the ...Missing: sensing | Show results with:sensing
  50. [50]
    α-Synuclein Promotes SNARE-Complex Assembly in Vivo and in Vitro
    α-synuclein directly interacts with the SNARE protein synaptobrevin and functions as a catalyst for SNARE-complex assembly.
  51. [51]
    Alpha-synuclein promotes SNARE-complex assembly in vivo and in ...
    α-synuclein directly bound to the SNARE-protein synaptobrevin-2/vesicle-associated membrane protein 2 (VAMP2) and promoted SNARE-complex assembly.
  52. [52]
    Dynamic control of the dopamine transporter in neurotransmission ...
    Mar 5, 2021 · ... short-term plasticity in ... A53T-alpha-synuclein overexpression impairs dopamine signaling and striatal synaptic plasticity in old mice.
  53. [53]
    Double-knockout mice for alpha- and beta-synucleins - PubMed
    Oct 12, 2004 · To investigate the functions of synucleins systematically, we have now generated single- and double-knockout (KO) mice that lack alpha- and/or ...
  54. [54]
    The Physiological Role of α-synuclein and Its ... - PubMed - NIH
    ... synuclein triple knockout mice now shows that synuclein accelerates dilation of the exocytic fusion pore. This form of regulation affects primarily the release ...
  55. [55]
    Role of α-Synuclein in Adult Neurogenesis and Neuronal Maturation ...
    Nov 21, 2012 · We investigated how α-synuclein levels modulate adult neurogenesis and the development of dendritic arborization and spines in the dentate gyrus.
  56. [56]
    A role for α-Synuclein in axon growth and its implications in ...
    Mar 30, 2020 · Our findings in cultured cortical neurons indicate a role for α-Syn in elongation of the main axon and its collaterals, resulting in enhanced axonal ...
  57. [57]
    Exploring α-Syn's Functions Through Ablation Models: Physiological ...
    May 19, 2025 · α-Syn's ablation induces cellular and physiological changes, including synaptic abnormalities, altered leukocyte development, inflammation, neurodegeneration, ...
  58. [58]
    Alpha-synuclein is upregulated in neurones in response to chronic ...
    These data indicate that increased alpha-synuclein content is associated with neuroprotection from relatively low levels of oxidative stress. Publication ...
  59. [59]
    Alpha-Synuclein Effects on Mitochondrial Quality Control in ...
    Dec 22, 2024 · (i) The overexpression of αS may stabilize Miro proteins, which are required for the formation of mitochondrial-derived vesicles (MDVs). (ii) αS ...
  60. [60]
    Autoproteolytic Fragments are Intermediates in the Oligomerization
    The autoproteolytic degradation of αSyn and the significance of the fragment αSyn(72-140) were ascertained by a number of additional mass spectrometric and gel ...Missing: activity | Show results with:activity
  61. [61]
  62. [62]
    Cell-Produced α-Synuclein Is Secreted in a Calcium-Dependent ...
    May 19, 2010 · Our results show for the first time that cell-produced α-synuclein is secreted via an exosomal, calcium-dependent mechanism.
  63. [63]
    Lysosomal dysfunction increases exosome-mediated alpha ...
    This study clearly demonstrates the importance of exosomes in both the release of alpha synuclein and its transmission between cells.
  64. [64]
    Tunneling nanotubes spread fibrillar α‐synuclein by intercellular ...
    Aug 22, 2016 · Here we show that α‐synuclein fibrils efficiently transfer from donor to acceptor cells through tunneling nanotubes (TNTs) inside lysosomal vesicles.
  65. [65]
    Cofilin 1 promotes the pathogenicity and transmission of ... - Nature
    Jan 10, 2022 · ... templated conformational change. The concept of “strain” has been proposed in recent years, which suggest that different strains of α ...
  66. [66]
    Human Astrocytes Transfer Aggregated Alpha-Synuclein via ...
    Dec 6, 2017 · We show that human astrocytes actively transfer aggregated α-synuclein (α-SYN) to healthy astrocytes via direct contact and tunneling nanotubes (TNTs), rather ...<|control11|><|separator|>
  67. [67]
    The prion hypothesis in Parkinson's disease: Braak to the future
    May 8, 2013 · Alpha synuclein can be released into the extracellular space via (1) leakage from injured cells with compromised membrane integrity.
  68. [68]
    Exogenous α-synuclein fibrils seed the formation of Lewy body-like ...
    Nov 24, 2009 · We show here that intracellular α-Syn aggregation can be triggered by the introduction of exogenously produced recombinant α-Syn fibrils into cultured cells.
  69. [69]
    α-synuclein strains that cause distinct pathologies differentially ...
    Jul 22, 2020 · Here, we generated two types of α-synuclein fibrils from identical monomer and investigated their seeding and propagation ability in mice and ...
  70. [70]
    α-Synuclein binds to TOM20 and inhibits mitochondrial ... - PubMed
    Jun 8, 2016 · This binding prevented the interaction of TOM20 with its co-receptor, TOM22, and impaired mitochondrial protein import. Consequently, there ...<|separator|>
  71. [71]
    Alpha-synuclein interacts with regulators of ATP homeostasis in ...
    Aug 16, 2025 · Mitochondrial accumulation of αSyn impairs the function of complex 1 and reduces mitochondrial activity. Additionally, oligomeric species of ...
  72. [72]
    α-Synuclein pathology disrupts mitochondrial function in ...
    Oct 8, 2024 · Alpha-synuclein mitochondrial interaction leads to irreversible translocation and complex I impairment. Arch Biochem Biophys. 2018;651:1–12 ...Missing: review | Show results with:review
  73. [73]
    Role of mitochondria in α-synuclein mediated neuronal toxicity - PMC
    The build-up of alpha-synuclein aggregates on the mitochondrial surface blocks Complex I activity and generation of reactive oxygen species (ROS), resulting in ...
  74. [74]
    α-Synuclein pathology and mitochondrial dysfunction: Toxic partners ...
    Jun 1, 2025 · In this review, we explore the contributions of α-synuclein pathology and impaired mitochondrial homeostasis in the etiology of PD.Missing: neuroprotection | Show results with:neuroprotection
  75. [75]
    Serine-129 phosphorylated α-synuclein drives mitochondrial ...
    Mar 30, 2025 · Our findings demonstrate that p-α-syn disrupts ER-mitochondria calcium homeostasis, providing new insights into its role in mitochondrial ...
  76. [76]
    Alpha synuclein modulates mitochondrial Ca 2+ uptake from ER ...
    Sep 23, 2023 · Our results indicate that a-syn can participate as a tethering protein to modulate Ca 2+ flux between ER and mitochondria, with potential physiological ...
  77. [77]
    Article ER Stress and Autophagic Perturbations Lead to Elevated ...
    Mar 8, 2016 · ER stress, autophagic/lysosomal perturbations, and elevated extracellular α-synuclein likely represent critical early cellular phenotypes of PD.
  78. [78]
    Lysosomal dysfunction in α-synuclein pathology - PubMed Central
    Sep 3, 2024 · Converging data suggest that lysosomal dysfunction is a critical event in the etiology of Parkinson's disease (PD).
  79. [79]
    Autophagy in Synucleinopathy: The Overwhelmed and Defective ...
    Here, we aimed to discuss the active participation of autophagy impairment in alpha-synuclein accumulation and propagation, as well as alpha-synuclein- ...
  80. [80]
    [PDF] Alpha-synuclein overexpression triggers divergent cellular ... - bioRxiv
    Aug 27, 2025 · The physiological role of monomeric α-syn in mitochondrial bioenergetics is supported by knockout studies where loss of α-syn resulted in ...
  81. [81]
    Alpha-synuclein biology in Lewy body diseases - PMC
    Oct 27, 2014 · Primary α-synucleinopathies include Parkinson's disease (PD), dementia with Lewy bodies and multiple system atrophy.
  82. [82]
    α-Synuclein in Lewy bodies - Nature
    Aug 28, 1997 · Here we describe strong staining of Lewy bodies from idiopathic Parkinson's disease with antibodies for α-synuclein, a presynaptic protein of unknown function.
  83. [83]
    α-Synuclein in filamentous inclusions of Lewy bodies from ... - PNAS
    We show here that Lewy bodies and Lewy neurites from Parkinson's disease and dementia with Lewy bodies are stained strongly by antibodies directed against amino ...
  84. [84]
    Staging of brain pathology related to sporadic Parkinson's disease
    Essential for neuropathological diagnosis are alpha-synuclein-immunopositive Lewy neurites and Lewy bodies. The pathological process targets specific induction ...
  85. [85]
    Glial cytoplasmic inclusions in white matter oligodendrocytes of ...
    In MSA white matter, alpha-synuclein-positive GCIs were restricted to oligodendrocytes, and alpha-synuclein was localized to the filaments in GCIs by ...
  86. [86]
    Insights into the pathogenesis of multiple system atrophy
    Feb 17, 2020 · Neurodegeneration in MSA brains is preceded by the emergence of glial cytoplasmic inclusions (GCIs), which are insoluble α-synuclein accumulations within ...
  87. [87]
    Skin Biopsy Detection of Phosphorylated α-Synuclein in Patients ...
    Mar 20, 2024 · ... disease (PD), multiple system atrophy (MSA), dementia with Lewy bodies (DLB), and pure autonomic failure (PAF)?. Finding In this cross ...
  88. [88]
    a synopsis of isolated REM sleep behavior disorder as early ...
    Feb 11, 2025 · Common autonomic problems ... Dermal phospho-alpha-synuclein deposits confirm REM sleep behaviour disorder as prodromal Parkinson's disease.Missing: pure | Show results with:pure
  89. [89]
    Development of α-Synuclein Real-Time Quaking-Induced ... - Frontiers
    The sensitivity and specificity of α-syn RT-QuIC using CSF for PD and DLB diagnoses have been sufficiently reliable in various studies (Table 1). Detection of ...
  90. [90]
    Diagnostic value of skin RT-QuIC in Parkinson's disease - Nature
    Nov 15, 2021 · The α-synuclein RT-QuIC assay of minimally invasive skin punch biopsy is a reliable and reproducible biomarker for Parkinson's disease.
  91. [91]
    Misfolded α-Synuclein Assessment in the Skin and CSF by RT-QuIC ...
    May 2, 2023 · This study provides Class III evidence that RT-QuIC-detected AS in the skin and CSF distinguishes patients with IRBD from controls.
  92. [92]
    Cerebrospinal Fluid Phosphorylated Alpha‐Synuclein in Newly ...
    Jun 2, 2025 · Alpha‐synuclein (α‐syn), phosphorylated at serine 129 (pS129‐α‐syn), is a potential biomarker for Parkinson's disease (PD)Missing: pSer129 | Show results with:pSer129
  93. [93]
    A longitudinal study on α-synuclein in plasma neuronal exosomes ...
    Our results suggested that α-syn in plasma neuronal exosomes may serve as a biomarker to aid early diagnosis of PD and also as a prognostic marker for PD ...
  94. [94]
    A novel approach to detecting plasma synuclein aggregates for ...
    Jul 29, 2025 · Alpha-synuclein (αSyn) aggregates are pathognomonic of Parkinson's disease (PD) and play a critical role in its pathogenesis.
  95. [95]
    The α-synuclein PET tracer [18F] ACI-12589 distinguishes multiple ...
    Oct 27, 2023 · Our results indicate that α-synuclein pathology in MSA can be identified using [ 18 F]ACI-12589 PET imaging, potentially improving the diagnostic work-up of ...
  96. [96]
    a-synuclein PET Imaging: From Clinical Utility in Multiple System ...
    Jun 3, 2025 · A major breakthrough came in 2023, when, for the first time, the tracer [18F]ACI-12589 finally demonstrated that (i) a-synuclein pathology can ...
  97. [97]
    Enhanced serum-based seed amplification assay for detecting ...
    ... Open access; Published: 22 May 2025. Enhanced serum-based seed amplification assay for detecting propagative α-synuclein seeds in Parkinson's disease.
  98. [98]
    α-Synuclein seed amplification assay positivity beyond ... - The Lancet
    This review focuses on CSF-based α-synuclein SAAs and their application in detecting co-pathology across non-synucleinopathies, including Alzheimer's disease, ...
  99. [99]
    Current Status of α-Synuclein Biomarkers and the Need for α ...
    Aug 18, 2025 · Several α-synuclein biomarkers have been developed to aid diagnosis and research, such as cerebrospinal fluid (CSF) and blood-based measurements ...
  100. [100]
    Biophysics of α-Synuclein Membrane Interactions - PMC
    Among the most common membrane molecules in vivo include phospholipids, cholesterol ... α-Synuclein selectively binds to anionic phospholipids embedded in liquid- ...
  101. [101]
    Interplay of α-synuclein with Lipid Membranes - ACS Publications
    Mar 19, 2024 · We discuss several aspects of α-synuclein interactions with lipid membranes including cooperative adsorption, membrane remodeling and α-synuclein amyloid ...
  102. [102]
    Cholesterol facilitates interactions between α‐synuclein oligomers ...
    Aug 20, 2015 · It is not immediately evident how cholesterol facilitates the interaction between αSN FN-oligomers and vesicles. One effect may be found in the ...Missing: PS PI
  103. [103]
    α-Synuclein assembles into higher-order multimers upon membrane ...
    We demonstrate that α-synuclein multimerizes during membrane binding and that the membrane-bound, multimeric form of α-synuclein mediates SNARE complex ...Missing: stoichiometry fluidity
  104. [104]
    Impaired endo-lysosomal membrane integrity accelerates the ...
    Aug 9, 2017 · Alpha-synuclein induces lysosomal rupture ... Proaggregant nuclear factor(s) trigger rapid formation of alpha-synuclein aggregates in apoptotic ...Missing: perforation | Show results with:perforation
  105. [105]
    Membrane Interactions of α-Synuclein Revealed by Multiscale ...
    Mar 15, 2021 · α-Synuclein (αS) is a presynaptic protein that binds to cell membranes and is linked to Parkinson's disease (PD). Binding of αS to membranes ...Introduction · Results · Discussion and Conclusions · Supporting Information
  106. [106]
    Lipidic folding pathway of α-Synuclein via a toxic oligomer - Nature
    Jan 17, 2025 · The structures of filamentous intermediates and mature fibrils are now efficiently determined by single-particle cryo-electron microscopy.
  107. [107]
    Protein Partners of α‐Synuclein in Health and Disease - PMC - NIH
    Apr 25, 2016 · This review will focus on normal and disease associated protein partners of AS defined as monomer and oligomer binding ligands for AS in different compartments.
  108. [108]
  109. [109]
    Molecular mechanism for the synchronized electrostatic ... - Nature
    Aug 6, 2022 · We report here that αS phase-separates into liquid condensates by electrostatic complex coacervation with positively charged polypeptides such as Tau.
  110. [110]
  111. [111]
    Interaction between Parkin and α-Synuclein in PARK2-Mediated ...
    In this review article, we provide an overview of post-mortem neuropathological examinations of PARK2 patients and present the current knowledge of a ...
  112. [112]
    Lrrk2 phosphorylates alpha synuclein at serine 129 - PubMed
    Sep 11, 2009 · Here we show that full-length Lrrk2 or fragments containing its kinase domain have a significant capacity to phosphorylate recombinant alpha synuclein (Asyn) ...
  113. [113]
    Examining the mechanisms that link β-amyloid and α-synuclein ...
    Apr 30, 2012 · This review examines the interactions between Aβ and α-syn and proposes potential mechanistic links between Aβ accumulation and α-syn pathogenesis.
  114. [114]
    Proximity proteomics reveals unique and shared pathological ...
    Mar 23, 2025 · In conclusion, synucleinopathies have divergent and convergent αsyn-aggregate interactions, indicating unique and shared pathogenic mechanisms.
  115. [115]
    LRRK2 and α-Synuclein: Distinct or Synergistic Players in ...
    LRRK2 is a component of granular alpha-synuclein pathology in the brainstem of Parkinson's disease. Neuropathol. Appl. Neurobiol. 34 272–283. 10.1111/j.1365 ...