Mitotic dysregulation: Overexpression disrupts BUB1B-mediated mitotic checkpoints, causing chromosomal instability .
Metastasis promotion: Enhances invasive potential by destabilizing intermediate filaments (e.g., keratins) .
Therapy resistance: Chaperones ER-α36 variant, enabling estrogen-independent growth and tamoxifen resistance .
Cancer-Specific Associations
Table 1: SNCG overexpression in malignancies
Reduces MHC-II, CD80/86 expression on DCs, impairing T-cell priming .
Promotes immunosuppressive Treg differentiation (↑Foxp3+ cells by 40%) while suppressing Th1/Th17 responses .
Correlates with altered immune infiltrates:
Table 2: Multivariate survival analysis
Cohort | n | HR (95% CI) | p-value |
---|---|---|---|
TCGA-GC | 375 | 1.87 (1.2–2.9) | 0.006 |
ECC | 96 | 3.1 (1.6–6.0) | 0.001 |
ICC | 51 | 2.9 (1.3–6.5) | 0.009 |
SNCG (gamma-synuclein) belongs to the synuclein family of proteins that includes alpha-synuclein (SNCA) and beta-synuclein (SNCB). Unlike its neuronal-specific family members that are primarily associated with neurodegenerative disorders, SNCG was originally cloned from infiltrating breast carcinoma cells and has not been closely linked to neurological diseases . The protein is characterized by a specific structure featuring 5-6 repeat (KTKEGV) consensus sequences in the N-terminal domain, with a distinctive acidic C-terminal domain that likely contributes to its specialized functions .
In normal biology, SNCG expression is highly regulated and appears tissue-specific, though our understanding of its physiological role remains incomplete. The methodological approach to studying its normal function typically involves comparing its expression patterns across various non-cancerous human tissues using immunohistochemistry, RNA-seq, and proteomics.
When investigating SNCG expression, researchers should employ multiple complementary techniques:
RNA detection methods:
RT-PCR: Used effectively to detect SNCG mRNA in various cell lines as demonstrated in LNCaP, DU145, PC3, and LNCaP-AI prostate cancer cells
RNAscope: For in situ visualization of SNCG transcripts in tissue sections
RNA-seq/single-cell RNA-seq: Enables comprehensive transcriptomic profiling and can identify SNCG expression patterns at single-cell resolution
Protein detection methods:
Western blotting: Successfully applied to detect SNCG protein levels in prostate cancer cell lines with varying androgen sensitivity
Immunohistochemistry: Critical for assessing SNCG expression in human tissue samples and correlating with clinical parameters
Flow cytometry: Useful for quantifying SNCG in specific cell populations
For validation, always include appropriate positive controls (such as LNCaP cells, which express high levels of SNCG) and negative controls (such as DU145 and PC3, which show low or undetectable SNCG expression) .
Substantial evidence connects SNCG to multiple human cancers, particularly in advanced stages:
Documented SNCG overexpression in human cancers:
In prostate cancer specifically, SNCG functions as a novel androgen receptor (AR) coactivator, promoting cellular growth and proliferation by activating AR transcription in an androgen-dependent manner . Mechanistic studies using siRNA-mediated silencing of SNCG in LNCaP cells demonstrated that SNCG inhibition contributes to several anti-cancer effects:
Inhibition of cellular proliferation
Induction of cell-cycle arrest at the G1 phase
Suppression of cellular migration and invasion in vitro
Immunohistochemical analysis revealed that SNCG expression correlates with peripheral and lymph node invasion in prostate cancer, while being almost undetectable in benign or androgen-independent prostate lesions .
SNCG exhibits significant interaction with hormone signaling, particularly in prostate cancer:
Androgen dependency: SNCG expression is regulated by androgen status in human prostate cancer cells, with anti-androgen treatment largely blocking DHT-induced SNCG expression
AR coactivator function: Mechanistic studies have established SNCG as a novel androgen receptor coactivator that:
Role in hormone sensitivity: SNCG overexpression in androgen-independent LNCaP-AI cells restored androgen responsiveness, as evidenced by:
Methodologically, researchers investigating these interactions should employ:
Hormone treatment and deprivation experiments
Luciferase reporter assays for AR activity
Co-immunoprecipitation to detect direct protein-protein interactions
Cell proliferation assays under various hormonal conditions
In vivo xenograft models with hormone manipulation
When investigating SNCG's role in metastasis, researchers should employ a multi-faceted approach:
In vitro methods:
Migration assays: Wound healing and Boyden chamber assays demonstrate that silencing SNCG by siRNA in LNCaP cells suppresses cellular migration
Invasion assays: Matrigel-coated transwell assays show SNCG's involvement in invasive capability
3D culture systems: Organoid cultures provide more physiologically relevant models for studying metastatic processes
In vivo methods:
Orthotopic xenograft models: Allows assessment of primary tumor growth and local invasion
Experimental metastasis models: Tail vein injection to study colonization capacity
Spontaneous metastasis models: To evaluate the complete metastatic cascade
Molecular approaches:
Pathway analysis: Evidence suggests SNCG influences metastasis through several mechanisms:
Researchers should also consider using in situ technologies to visualize SNCG expression at the leading edge of tumors and in circulation to better understand its role throughout the metastatic process.
SNCG has significant immunomodulatory effects, particularly on dendritic cells (DCs), which are critical for initiating immune responses. Research has revealed:
Inhibition of DC maturation: SNCG prevents the functional maturation of bone marrow-derived dendritic cells (BMDCs)
Cytokine modulation: SNCG-treated mature DCs (mDCs) show:
T-cell response alteration: DCs exposed to SNCG:
Immune evasion mechanism: SNCG-treated DCs induce Th2 immunity while decreasing inflammatory cytokines, potentially contributing to inhibition of anti-tumor immunity
These findings suggest SNCG may be part of tumor-induced immunosuppression mechanisms. To study these interactions, researchers should:
Isolate and culture primary DCs with recombinant SNCG
Assess DC maturation markers (CD80, CD86, MHCII) by flow cytometry
Measure cytokine profiles using multiplex assays or ELISA
Conduct DC-T cell co-culture experiments to evaluate T-cell differentiation
Perform in vivo experiments with SNCG-conditioned DCs to assess tumor growth
Several cutting-edge technologies are advancing our understanding of SNCG biology:
Single-cell transcriptomics: This technology allows researchers to:
3D organoid models: Three-dimensional culture systems enable:
Bioengineered platforms: Advanced materials can enhance experimental consistency:
CRISPR-Cas9 genome editing: Enables precise manipulation of SNCG expression and modification of specific domains to elucidate structure-function relationships
Proteomics and interactomics: Mass spectrometry-based approaches can identify the SNCG interactome in different cellular contexts
To leverage these technologies, researchers should consider collaborative approaches with specialized laboratories and ensure appropriate controls are included to account for technical variability.
The SNCG research field faces several contradictions and challenges that researchers should systematically address:
Context-dependent expression patterns:
Methodological approach to resolve contradictions:
Standardize detection methods across studies
Clearly define cell and tissue types being examined
Consider androgen/hormone status when comparing results
Use multiple complementary techniques (protein and RNA detection)
Document clinical characteristics comprehensively
Molecular context considerations:
Assess AR status when studying SNCG in prostate models
Evaluate hormone receptor status in breast cancer studies
Consider immune cell infiltration and tumor microenvironment
Experimental design recommendations:
Include time-course experiments to capture dynamic changes
Use isogenic cell lines to control genetic background
Employ in vivo models that recapitulate human disease progression
Consider patient-derived xenografts or organoids to better reflect human disease
SNCG shows significant potential as both a biomarker and therapeutic target:
Biomarker applications:
Prognostic indicator: High SNCG expression correlates with peripheral and lymph node invasion in prostate cancer, suggesting utility as a biomarker for predicting cancer progression and metastasis
Treatment response prediction: SNCG expression status might predict response to androgen deprivation therapy in prostate cancer, given its androgen-dependent expression pattern
Liquid biopsy development: Detection of SNCG in circulation could potentially serve as a non-invasive biomarker
Therapeutic targeting strategies:
Direct SNCG inhibition: RNA interference approaches demonstrated that silencing SNCG inhibits cellular proliferation, induces cell-cycle arrest, and suppresses migration and invasion in vitro
Disruption of protein-protein interactions: Targeting the SNCG-AR interaction could potentially inhibit androgen-dependent tumor growth
Immunomodulatory approaches: Counteracting SNCG's immunosuppressive effects on dendritic cells could potentially enhance anti-tumor immune responses
Combination therapies: SNCG-targeting approaches might sensitize tumors to hormone therapy or immune checkpoint inhibitors
Research focusing on these applications should include:
Validation in large, well-characterized patient cohorts
Comparison with standard clinical biomarkers
Development of clinically applicable detection methods
Preclinical studies of targeting strategies in relevant models
Gamma-Synuclein is a presynaptic protein that plays a crucial role in the peripheral nervous system and retina . It is involved in maintaining the integrity of the neurofilament network and modulating axonal architecture during development and in adulthood . The protein consists of 127 amino acids and has a molecular mass of approximately 13,331 Da .
Gamma-Synuclein is implicated in several neurodegenerative diseases, including Parkinson’s Disease (PD) and other synucleinopathies . It is a major component of pathological aggregates found in these disorders. The protein’s role in disease pathogenesis is linked to its ability to form insoluble fibrils that contribute to neuronal dysfunction and cell death .
Recombinant human gamma-synuclein is produced using Escherichia coli (E. coli) expression systems. The recombinant protein is purified to ensure high purity and quality, making it suitable for various research applications, including aggregation studies, seeding experiments, and molecular standards . The protein is typically provided in a lyophilized form and can be reconstituted in buffer solutions for experimental use .
Recombinant gamma-synuclein is widely used in research to study its role in neurodegenerative diseases. It is utilized in experiments to understand the mechanisms of protein aggregation, the formation of pathological inclusions, and the development of potential therapeutic strategies . Additionally, gamma-synuclein is used as a molecular standard in various biochemical assays .