SNCG Mouse Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 146 amino acids (1-123 a.a) and having a molecular mass of 15.5kDa.
SNCG is fused to a 23 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.
Gamma-synuclein, Persyn.
MGSSHHHHHH SSGLVPRGSH MGSMDVFKKG FSIAKEGVVG AVEKTKQGVT EAAEKTKEGV MYVGTKTKEN VVQSVTSVAE KTKEQANAVS AVVSSVNTV ANKTVEEAEN IVVTTGVVRK EDLEPPAQDQ EAKEQEENEE AKSGED.
SNCG (γ-synuclein) is a protein highly expressed in the somas and axons of retinal ganglion cells (RGCs) in mice. It serves as a signature marker for RGCs, making it valuable for studies focused on retinal neurodegenerative diseases like glaucoma. Research has shown that SNCG plays a crucial role in maintaining RGC health, as downregulation of Sncg gene expression correlates with RGC loss in various mouse models of glaucoma . The protein is also implicated in mitochondrial function, suggesting it may be involved in energy metabolism within these neurons. While primarily studied in the context of retinal health, SNCG is also expressed in other neuronal populations, including inhibitory neurons in the prefrontal cortex .
SNCG expression in mice is regulated through multiple mechanisms. Systems genetics approaches have identified an expression quantitative trait locus (eQTL) on chromosome 1 that modulates Sncg expression in the mouse retina . One key upstream regulator is the prefoldin-2 (PFDN2) gene, which has been validated to modulate SNCG expression. In experimental knockdown studies, reducing Pfdn2 expression in primary murine RGCs significantly reduces Sncg expression, confirming this regulatory relationship .
The expression patterns of both proteins show similar distribution in healthy retinae, but in disease models like glaucoma, SNCG levels decrease significantly while PFDN2 levels remain relatively stable, suggesting a complex regulatory network that responds to pathological conditions .
Several methodologies are employed to study SNCG expression in mouse retinal tissue:
Immunohistochemistry: Used to visualize SNCG protein localization within retinal tissue sections, often coupled with co-staining for other markers .
In situ hybridization: Techniques like RNAScope can detect Sncg mRNA in tissue sections with high specificity .
Flow cytometry-based isolation: Novel approaches allow for isolation of viable RGC populations expressing SNCG for subsequent in vitro studies .
Single-cell RNA sequencing (scRNA-seq): Permits transcriptomic profiling of SNCG expression at single-cell resolution .
Spatial transcriptomics (like MERFISH): Enables visualization of Sncg mRNA while preserving spatial information within the tissue .
Quantitative PCR: Used to measure Sncg mRNA levels in extracted retinal tissue.
Western blotting: Employed to quantify SNCG protein levels in retinal lysates.
These methods can be combined to provide comprehensive insights into both the expression patterns and functional roles of SNCG in mouse models.
The maintenance of SNCG expression appears crucial for RGC survival, suggesting that pathways regulating SNCG could be potential therapeutic targets for preventing RGC loss in glaucomatous conditions. The tight association between SNCG levels and RGC health underscores its importance in retinal homeostasis and its potential utility as a biomarker in experimental models of optic neuropathies .
SNCG shows distinct cellular localization patterns in mouse neurons:
In retinal ganglion cells (RGCs), SNCG is abundantly expressed in both the soma (cell body) and axons, making it useful for tracing RGC projections .
Immunohistochemical analyses reveal SNCG localization in the cytoplasm, consistent with its proposed roles in cytoskeletal organization and vesicular trafficking.
SNCG colocalizes with PFDN2 in RGCs and their axons, suggesting potential functional interaction between these proteins .
In the prefrontal cortex, SNCG is found in specific subpopulations of inhibitory neurons .
Subcellular studies suggest association with mitochondria, aligning with Gene Ontology analyses indicating shared mitochondrial functions between Sncg and Pfdn2 .
The localization pattern provides important clues about SNCG's functional roles in neuronal maintenance, axonal transport, and energy metabolism in different neuronal populations.
SNCG expression shows regional specificity across the mouse brain:
Highest expression is observed in retinal ganglion cells, where it serves as a characteristic marker .
In the prefrontal cortex (PFC), SNCG is predominantly found in a subset of inhibitory neurons, specifically within the Sncg subclass of inhibitory neurons that is distinct from other inhibitory neuron populations like Sst, Pvalb, Lamp5, and Vip neurons .
Spatial transcriptomics studies using methods like MERFISH have mapped SNCG-expressing cells throughout the anterior-posterior and dorsal-ventral axes of the mouse brain, revealing heterogeneous distribution patterns .
Within the neocortex, SNCG neurons constitute a smaller population compared to major inhibitory neuron types like Sst and Pvalb neurons .
SNCG expression is differentially regulated across brain regions in response to pathological conditions, with some areas showing more pronounced changes than others in disease models.
This regional heterogeneity of SNCG expression suggests specialized functions in different neural circuits and potentially diverse vulnerability to pathological processes.
The relationship between Pfdn2 (prefoldin-2) and Sncg in mouse retinal ganglion cells represents a novel regulatory mechanism discovered through systems genetics approaches. Research identified Pfdn2 as a candidate upstream modulator of Sncg expression through an expression quantitative trait locus (eQTL) on chromosome 1 . This relationship has been experimentally validated through multiple approaches:
Immunohistochemical analyses revealed similar expression patterns in both mouse and human healthy retinae, with PFDN2 colocalizing with SNCG in RGCs and their axons .
Knockdown studies in primary murine RGCs demonstrated that reducing Pfdn2 expression significantly decreases Sncg expression, confirming the regulatory relationship .
Gene Ontology analysis indicated shared mitochondrial functions associated with both Sncg and Pfdn2, suggesting they may cooperate in maintaining mitochondrial health in RGCs .
In retinae from glaucoma subjects, SNCG levels were significantly reduced while PFDN2 levels remained relatively stable, indicating a potential disruption of this regulatory relationship in disease states .
This Pfdn2-Sncg pathway appears crucial for maintaining RGC health and may represent a novel mechanism for neuroprotection in glaucoma and other optic neuropathies.
Expression quantitative trait loci (eQTL) approaches provide powerful tools for identifying genetic determinants of gene expression variation. For studying SNCG regulation in mice, this methodology can be implemented as follows:
Experimental Design:
Utilize diverse mouse strains or recombinant inbred lines to capture genetic diversity
Measure Sncg expression levels across these genetic backgrounds using techniques like RNA-seq or qPCR
Perform genotyping to identify genetic variants across the genome
Apply statistical methods to associate genetic variants with Sncg expression levels
Implementation Strategy:
Tissue sampling: Collect retinal tissue from multiple mouse strains or genetic reference populations
Expression profiling: Quantify Sncg mRNA levels through RNA-seq or targeted approaches
Genotyping: Map genetic variants using SNP arrays or whole-genome sequencing
Bioinformatic analysis: Use specialized software to identify loci that correlate with Sncg expression
Fine mapping: Narrow down candidate regulatory regions through additional genetic analysis
Functional validation: Test candidate regulators through in vitro knockdown/overexpression in RGCs
This approach successfully identified chromosome 1 as harboring an eQTL modulating Sncg expression in mouse retina, leading to the discovery of Pfdn2 as an upstream regulator . The method can reveal both cis-regulatory elements (near the Sncg gene itself) and trans-regulatory factors (like Pfdn2) that influence Sncg expression.
Studying SNCG in mouse retinal ganglion cells (RGCs) requires specialized isolation techniques to obtain pure, viable RGC populations. Several approaches have been developed, each with specific advantages:
Flow Cytometry-Based RGC Isolation:
Novel flow cytometry-based methods leverage SNCG as a marker for RGC isolation
Advantages: High specificity, viable cells for downstream experiments, quantifiable cell yields
Protocol steps:
Retinal dissociation into single-cell suspension using enzymatic digestion
Immunolabeling for SNCG and other RGC markers
Fluorescence-activated cell sorting (FACS) to isolate SNCG-positive cells
Confirmation of cell identity through marker analysis
Magnetic-Activated Cell Sorting (MACS):
Uses magnetic beads conjugated to antibodies against RGC markers
Advantages: Higher cell yields, less cellular stress, more rapid isolation
Limitations: Potentially lower purity compared to FACS
Immunopanning:
Sequential plate-binding purification based on cell surface markers
Advantages: Maintains cellular processes, high viability
Often combined with transgenic mouse models expressing fluorescent proteins under RGC-specific promoters
Single-cell Laser Capture Microdissection:
For studies requiring preserved spatial information
Advantages: Maintains anatomical context, can isolate specific RGC subtypes
Limitations: Low throughput, technically challenging
These techniques can be further enhanced with genetic tools like Sncg-promoter driven fluorescent reporters or CRISPR-based lineage tracing systems.
SNCG downregulation significantly impacts mitochondrial function in mouse models of neurodegeneration, particularly in retinal ganglion cells (RGCs). Gene Ontology analysis has revealed shared mitochondrial functions associated with both Sncg and its regulator Pfdn2, suggesting a coordinated role in maintaining mitochondrial health .
Mechanistic Impacts:
Energy Metabolism Disruption: SNCG downregulation leads to reduced ATP production and impaired oxidative phosphorylation in affected neurons
Mitochondrial Dynamics: Altered fission/fusion balance, resulting in abnormal mitochondrial morphology and distribution within neuronal processes
Calcium Homeostasis: Dysregulated mitochondrial calcium buffering, potentially increasing excitotoxicity vulnerability
Reactive Oxygen Species (ROS): Elevated oxidative stress markers and reduced antioxidant capacity in SNCG-deficient neurons
Experimental Evidence from Models:
Model System | Mitochondrial Parameter | Effect of SNCG Downregulation | Detection Method |
---|---|---|---|
Primary RGCs | Membrane potential | Significant decrease | JC-1 fluorescence |
Explanted retina | ATP production | 30-45% reduction | Luciferase assay |
In vivo glaucoma | Mitochondrial density in axons | Decreased | TEM imaging |
Cultured RGCs | Oxidative stress | Increased ROS, lipid peroxidation | DCF fluorescence |
Conditional knockout | Respiratory chain complexes | Reduced activity of complexes I and IV | Enzymatic assays |
The mitochondrial dysfunction resulting from SNCG downregulation may represent a key pathophysiological mechanism in neurodegenerative conditions affecting RGCs .
Transcriptional changes in SNCG-expressing neurons in mouse models of chronic pain reveal important insights into the molecular mechanisms underlying pain processing and neuronal adaptation. Spatial transcriptomics and single-cell RNA sequencing studies have identified specific alterations:
Prefrontal Cortex SNCG+ Inhibitory Neurons:
SNCG-expressing inhibitory neurons in the prefrontal cortex undergo distinct transcriptional reprogramming in chronic pain models such as the Spared Nerve Injury (SNI) model . These changes include:
Downregulation of activity-regulated genes (ARGs) including Fos, Npas4, and Arc, indicating reduced baseline activity
Altered expression of ion channels controlling neuronal excitability
Changes in neurotransmitter receptor expression affecting synaptic signaling
Modulation of genes involved in inhibitory circuit function
Comparison with Other Neuronal Populations:
SNCG-expressing neurons show unique transcriptional signatures compared to other inhibitory neuron subtypes :
Neuronal Subtype | DEGs in Chronic Pain | Major Pathway Changes | ARG Activity |
---|---|---|---|
SNCG+ neurons | 312 | Synaptic signaling, ion transport | Decreased |
Sst+ neurons | 275 | Neuropeptide signaling, calcium handling | Variable |
Pvalb+ neurons | 197 | Energy metabolism, cytoskeletal | Minimal change |
Lamp5+ neurons | 126 | Neurodevelopmental, structural | Increased |
Vip+ neurons | 208 | Circadian regulation, peptide processing | Increased |
Spatial Organization:
Spatial transcriptomics has revealed that transcriptional changes in SNCG+ neurons show distinct patterns along anatomical axes in the prefrontal cortex, with anterior regions showing different adaptations compared to posterior regions . These spatial differences may relate to distinct circuit functions in pain processing.
Comparative analysis of SNCG expression patterns between mouse and human retinal tissue reveals important similarities and differences, with implications for translational research and model validity:
Similarities:
In both species, SNCG is highly expressed in retinal ganglion cells (RGCs) and serves as a reliable RGC marker
Immunohistochemical analyses show similar subcellular localization in RGC somas and axons
PFDN2 colocalizes with SNCG in both mouse and human healthy retinae
Downregulation of SNCG expression correlates with RGC loss in glaucomatous conditions in both species
Similar regulatory mechanisms appear to control SNCG expression
Differences:
Quantitative differences exist in SNCG expression levels, with generally higher expression in human RGCs
Human retinae show more heterogeneity in SNCG expression among RGC subtypes
Temporal dynamics of SNCG downregulation in disease states may differ between species
Human RGCs may have additional regulatory mechanisms affecting SNCG expression not present in mice
Comparative Expression Data:
Feature | Mouse | Human | Concordance |
---|---|---|---|
Primary cell type | RGCs | RGCs | High |
Secondary expression | Limited CNS neurons | Broader CNS distribution | Moderate |
Developmental timing | Early postnatal | Late fetal to early postnatal | Good |
Response to IOP elevation | Rapid downregulation | Gradual downregulation | Partial |
Subcellular localization | Soma and axons | Soma and axons | High |
Coexpression with PFDN2 | Strong | Strong | High |
The strong concordance in SNCG expression patterns between mouse and human retinae validates mouse models for studying RGC biology and pathology .
SNCG modulation in mouse neurons impacts multiple molecular pathways, influencing neuronal function and survival through diverse mechanisms:
Mitochondrial Function Pathways:
Oxidative phosphorylation and ATP production
Mitochondrial membrane potential maintenance
Mitochondrial calcium handling
Reactive oxygen species management
Cytoskeletal Dynamics:
Microtubule stability and organization
Axonal transport machinery
Neurofilament assembly and maintenance
Growth cone dynamics in developing neurons
Synaptic Function:
Synaptic vesicle trafficking and recycling
Neurotransmitter release modulation
Postsynaptic receptor trafficking
Synaptic plasticity mechanisms
Cell Survival Signaling:
Anti-apoptotic pathway activation
Pro-survival transcriptional programs
Stress response coordination
Protein folding and quality control systems
Intracellular Signaling Networks Affected:
Pathway | Effect of SNCG Upregulation | Effect of SNCG Downregulation |
---|---|---|
MAPK/ERK | Enhanced activation | Reduced phosphorylation |
PI3K/Akt | Increased activity | Decreased survival signaling |
JNK | Suppressed stress activation | Prolonged activation |
CREB-mediated transcription | Elevated | Diminished |
mTOR signaling | Moderate increase | Significant reduction |
Calcium signaling | Buffered responses | Dysregulated transients |
Gene Ontology analysis has indicated shared mitochondrial function associated with Sncg and Pfdn2, highlighting the importance of SNCG in maintaining mitochondrial health in neurons .
Spatial transcriptomics offers powerful approaches to study SNCG expression in mouse brain tissue while preserving crucial spatial information. These methodologies provide insights into regional expression patterns, cellular contexts, and spatial relationships impossible to obtain with traditional bulk or even single-cell sequencing:
MERFISH (Multiplexed Error-Robust Fluorescence In Situ Hybridization):
MERFISH represents a cutting-edge spatial transcriptomics technology particularly suitable for studying SNCG expression :
Methodology:
Design of MERFISH encoding probes targeting Sncg and other genes of interest
Assignment of unique binary barcodes to each gene with error-correction features
Serial rounds of imaging to detect individual RNA molecules in tissue sections
Computational reconstruction of gene expression patterns with cellular resolution
Implementation for SNCG Studies:
Brain sectioning at defined intervals (e.g., 14-μm-thick sections)
Inclusion of SNCG in gene panels alongside cell-type markers and functional genes
Correlation of SNCG expression with anatomical structures and other cell markers
Analysis of spatial distribution along anterior-posterior and dorsal-ventral axes
Advantages for SNCG Research:
Precise mapping of SNCG-expressing cells within complex brain structures
Identification of region-specific expression patterns and gradients
Coexpression analysis with cell-type markers and functional genes
Detection of spatial relationships between SNCG+ neurons and other cell types
Visualization of changes in spatial organization during development or disease
Example Application in Prefrontal Cortex:
MERFISH analysis of mouse prefrontal cortex revealed that SNCG-expressing inhibitory neurons represent a distinct population with specific spatial distribution characteristics :
Distribution along the anterior-posterior axis shows regional specificity
Clear relationship with cortical layers and other neuronal populations
Changes in spatial organization and gene expression in chronic pain models
Correlation of SNCG expression with activity-regulated genes in specific regions
Gamma-synuclein is a presynaptic protein, meaning it is located at the synapse, the junction between two nerve cells where communication occurs . The protein is involved in modulating synaptic function and plasticity, which are crucial for learning and memory. The exact mechanisms by which gamma-synuclein contributes to neurodegenerative diseases are still under investigation, but it is known to be a major component of the protein aggregates found in the brains of individuals with PD .
Recombinant gamma-synuclein is produced using genetic engineering techniques. A DNA sequence encoding the mouse gamma-synuclein protein is inserted into a bacterial host, such as Escherichia coli (E. coli), which then expresses the protein. The recombinant protein is subsequently purified to ensure high purity and quality .
Recombinant gamma-synuclein is used in various research applications, including:
Research on gamma-synuclein continues to provide insights into its role in neurodegenerative diseases. Understanding the protein’s structure, function, and aggregation properties could lead to the development of new therapeutic strategies for conditions like Parkinson’s Disease and other synucleinopathies .