SNCA (synuclein alpha) is a human gene located on chromosome 4q22.1 that encodes the alpha-synuclein protein, a presynaptic neuronal protein critical for synaptic function and vesicle trafficking . It is predominantly expressed in the brain but also found in the heart, skeletal muscle, and pancreas . Dysregulation of SNCA expression or mutations in its gene locus are strongly associated with neurodegenerative disorders, including Parkinson’s disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA) .
Over 30 pathogenic SNCA mutations have been identified, primarily linked to early-onset Parkinson’s disease :
Synaptic vesicle regulation: Modulates dopamine release and synaptic vesicle recycling .
Neuroprotection: Inhibits p53-mediated apoptosis in non-dopaminergic neurons .
Microtubule dynamics: Interacts with tau to stabilize neuronal cytoskeleton .
Aggregation: Misfolded α-synuclein forms oligomers and fibrils, contributing to Lewy body formation .
Mitochondrial dysfunction: Impairs Complex I activity, increasing oxidative stress .
Interneuronal transmission: Propagates aggregates across brain regions via prion-like mechanisms .
Antisense oligonucleotides (ASOs): Target SNCA transcripts with novel 3’ UTRs, reducing aggregation .
Small-molecule inhibitors: Screen identifies compounds modulating SNCA transcription (e.g., kinase inhibitors) .
Enhancer editing: Deletion of intronic enhancer clusters reduces SNCA expression in neurons .
Evolutionary conservation: SNCA’s NAC domain is highly conserved across vertebrates, while the N-terminal region shows lineage-specific destabilizing substitutions to maintain intrinsic disorder .
Tissue expression: Highest in neurons (frontal cortex, hippocampus) and glia; detectable in mitochondria of dopaminergic neurons .
Isoforms: Alternative splicing generates α-synuclein-112, -126, and -140, with truncated forms showing altered aggregation propensity .
Novel transcripts: Long-read RNA-seq identifies unannotated SNCA transcripts encoding truncated proteins (e.g., αSyn-112, αSyn-128) in human brain .
Enhancer clusters: Intronic enhancers act as “super-enhancers” in PD-risk loci, regulated by THAP1 and CTCF .
Structural epistasis: Mutations in the critical region (residues 32–58) disrupt interdomain communication, altering SNCAIP interactions .
MDVFMKGLSK AKEGVVAAAE KTKQGVAEAA GKTKEGVLYV GSKTKEGVVH GVATVAEKTK EQVTNVGGAV VTGVTAVAQK TVEGAGSIAA ATGFVKKDQL GKNEEGAPQE GILEDMPVDP DNEAYEMPSE EGYQDYEPEA.
The SNCA gene encodes alpha-synuclein protein, a major component of Lewy bodies found in Parkinson's disease (PD) . Alpha-synuclein is predominantly expressed in the brain and plays critical roles in synaptic function. The importance of SNCA in neurodegeneration is highlighted by evidence that increased dosage of wild-type human SNCA protein is critical to PD pathogenesis . Single point mutations, duplications, triplications, or rearrangements of the SNCA gene can cause familial forms of PD, with symptom severity correlating with the number of SNCA copies . Additionally, polymorphisms in SNCA gene regulatory regions that enhance SNCA expression are associated with sporadic PD . Currently, SNCA is considered one of the most validated therapeutic targets for PD, with numerous studies demonstrating beneficial impacts of manipulating SNCA levels .
SNCA expression in the human brain is regulated through a complex mechanism involving transcription factors and enhancer regions. Research has identified that THAP1 (THAP domain-containing apoptosis-associated protein 1) and its interaction partner CTCF (CCCTC-binding factor) act as transcription regulators of SNCA . THAP1 controls SNCA intronic enhancers' activities, while CTCF regulates its enhancer-promoter loop formation .
The SNCA intronic enhancers display neurodevelopment-dependent activities and form enhancer clusters similar to "super-enhancers" in the brain . These enhancers are predominantly active in neural tissues, with extremely high expression levels of SNCA observed in the human cerebellum and frontal cortex compared to non-neuronal tissues . This tissue-specific regulation explains why SNCA is predominantly expressed in the brain.
SNCA duplication carriers exhibit significant cognitive deficits, particularly in reward learning, even before the onset of motor symptoms associated with Parkinson's disease . In a study involving seven siblings who were asymptomatic carriers of SNCA duplication, researchers found that these individuals exhibited chance-level performance without any evidence of learning during reward-based tasks . This impairment is similar to findings in young, non-medicated patients with Parkinson's disease and aligns with computational models of cognitive reinforcement learning in parkinsonism .
Importantly, these reward learning deficits were observed in the premotor stage of the disease, indicating that cognitive deficits can precede motor symptoms and are dissociable from them . During follow-up periods, all carriers in the study eventually developed Parkinson's disease and experienced marked cognitive decline . This suggests that SNCA duplication impacts cognitive function early in the disease process, potentially through mechanisms affecting dopamine release in nigrostriatal synapses .
The intronic enhancers of SNCA display tissue-specific activation patterns and play a crucial role in regulating SNCA expression, particularly in the brain. These enhancers form clusters similar to "super-enhancers" specifically in brain tissue and are enriched with PD-associated single-nucleotide polymorphisms (SNPs) . The enhancer activity is neurodevelopment-dependent, with differential activation during neuronal differentiation .
Research has demonstrated that the SNCA intronic enhancers predominantly regulate its expression by controlling RNA polymerase II (Pol II) pause release rather than transcription initiation . When the intronic enhancer clusters of SNCA were deleted in a transgenic rat model, researchers observed that the paused Pol II remained at the SNCA promoter and was unable to proceed to the gene body, leading to a drastic reduction in SNCA expression in the brain . This mechanism explains the brain-specific high expression of SNCA despite its promoter being active in various tissues.
The cell lineage-dependent activation of SNCA enhancers correlates with SNCA expression levels during differentiation: very low levels in induced pluripotent stem cells (iPSCs), moderate expression in iPSC-derived neurons, and extremely high expression in human brain tissues such as cerebellum and frontal cortex .
THAP1 and CTCF are critical regulators of SNCA transcription, operating through distinct but complementary mechanisms. THAP1, a DYT6 gene product, regulates SNCA expression by controlling its promoter and intronic enhancers' activities . Studies have shown that THAP1 binds to the SNCA promoter and its intronic enhancers, particularly enhancer-1 (En-1) and enhancer-2 (En-2) .
In transgenic rat models, co-expression of human THAP1 and human SNCA led to significantly increased expression of both rat SNCA and human SNCA compared to rats expressing only human SNCA . This increase was most pronounced in the cerebellum, where THAP1 expression was highest, confirming THAP1's role in upregulating SNCA expression in vivo .
Meanwhile, CTCF, an interaction partner of THAP1, regulates the enhancer-promoter loop formation of the SNCA gene . This three-dimensional chromatin architecture is essential for bringing distant enhancers into proximity with the SNCA promoter to facilitate transcription. The coordinated action of THAP1 and CTCF ensures proper regulation of SNCA expression in the brain.
SNCA overexpression impairs dopamine release in nigrostriatal synapses, which can occur even before overt neuropathological changes or motor symptoms . Research by Nemani et al. demonstrated that slight overexpression of SNCA in synaptic terminals, similar to the estimated effect of gene duplication, significantly inhibits neurotransmitter release . This inhibition occurs through reduced synaptic vesicle density at the active zone and deficient vesicle clustering .
The impaired dopamine release directly impacts reward learning capabilities. In a study of asymptomatic SNCA duplication carriers, participants showed pronounced impairments in reward learning tasks despite having no clinical signs of Parkinson's disease and no detectable dopamine transporter abnormalities on [123I]β-CIT SPECT imaging . The following table shows the demographic comparison between SNCA duplication carriers and controls:
A clear dosage effect exists between SNCA gene copy number and Parkinson's disease severity . Duplication, triplication, or rearrangements of the wild-type SNCA gene can cause familial PD, with the severity of clinical phenotypes directly correlating with the number of SNCA copies . Patients with SNCA triplication typically experience earlier disease onset, more rapid progression, and more severe cognitive impairment compared to those with duplication .
This gene dosage effect is further supported by animal models where the degree of neurodegeneration correlates with SNCA expression levels. Mouse and rat models overexpressing human wild-type SNCA protein display alpha-synucleinopathies, nigrostriatal degeneration, and even inclusion body formation . The severity of these pathological features increases with higher levels of SNCA expression.
In human studies, carriers of SNCA duplication who initially appeared asymptomatic eventually developed Parkinson's disease during follow-up periods . These individuals also experienced marked cognitive decline, with Mini-Mental State Examination scores dropping from a baseline of 30.0 to 22.4 during follow-up . This progressive deterioration highlights the direct relationship between SNCA copy number, protein levels, and disease manifestation.
Studying SNCA expression in human brain tissue requires a combination of molecular biology techniques and careful tissue handling. Quantitative PCR (qPCR) is commonly used to measure SNCA mRNA levels, while Western blotting is employed to quantify protein expression . For spatial localization, immunohistochemistry can determine the cellular and subcellular distribution of alpha-synuclein protein.
To investigate the chromatin structure and enhancer activity of the SNCA gene, techniques such as chromatin immunoprecipitation (ChIP) followed by sequencing (ChIP-seq) are employed . Histone modifications like H3K27ac and H3K4me1 are analyzed to identify active enhancers . Additionally, chromosome conformation capture technologies (3C, 4C, Hi-C) can determine the three-dimensional interactions between SNCA enhancers and its promoter .
For functional studies of SNCA enhancers, reporter assays in which enhancer regions are cloned upstream of a minimal promoter driving a reporter gene (like luciferase) can measure enhancer activity in different cell types . CRISPR-Cas9-based techniques allow for precise genetic manipulation, including enhancer deletions or modifications, to assess their impact on SNCA expression .
While iPSC-derived neurons offer valuable insights into SNCA regulation, they have significant limitations compared to mature human brain tissue. Research has shown that the transcription machinery of the human SNCA gene and its expression level in the brain differ substantially from those in cultured neurons . iPSC-derived neurons express only moderate levels of SNCA compared to the extremely high levels observed in human brain tissues such as cerebellum and frontal cortex .
This discrepancy is partly due to the developmental stage of iPSC-derived neurons, which may not fully recapitulate the mature state of neurons in the adult brain. The SNCA intronic enhancers show neurodevelopment-dependent activities, with gradual activation during neuronal differentiation . Consequently, iPSC-derived neurons may not accurately reflect the enhancer activities present in adult brain tissue.
Furthermore, the three-dimensional organization of chromatin and enhancer-promoter interactions may differ between iPSC-derived neurons and brain tissue. The cell type heterogeneity present in the brain is also difficult to model using iPSC-derived neurons, which typically represent a more homogeneous population .
These limitations underscore the importance of interpreting results from iPSC-derived neuron studies with caution, particularly when studying the transcription regulation of the human SNCA gene or the effects of PD risk-associated SNPs .
Enhancer-targeting approaches offer promising new avenues for treating SNCA-related diseases by addressing the tissue-specific nature of SNCA expression regulation. The discovery that SNCA intronic enhancers predominantly control its expression in the brain through regulation of RNA polymerase II pause release provides a novel therapeutic target .
Unlike strategies targeting general transcription factors or broad histone modifications, which may cause global gene expression changes and side effects, enhancer-targeting approaches could enable tissue-specific modulation of SNCA expression . For instance, targeting the brain-specific enhancer cluster of SNCA could potentially reduce its expression in the brain while preserving its expression in other tissues where it may serve essential functions.
Additionally, the enhancer-targeting approach may be more precise than targeting the SNCA promoter. Research has shown that while the SNCA promoter is active in various tissues, the intronic enhancers are predominantly active in the brain . This suggests that targeting the promoter alone might not be sufficient to down-regulate SNCA expression in the brain, whereas targeting brain-specific enhancers could achieve this goal more effectively.
As evidence supporting the feasibility of this approach, experiments have demonstrated that knocking out specific enhancers (such as En-1) in dopaminergic neuronal cells moderately represses SNCA expression, while deletion of the same enhancer in cells where it is inactive has no effect on SNCA expression . This cell-type specificity could translate to reduced side effects compared to more global approaches to SNCA suppression.
Alpha-synuclein is involved in several critical biological processes:
Alpha-synuclein is a major component of Lewy bodies, which are pathological hallmarks of several neurodegenerative diseases, collectively known as synucleinopathies. These include: