SNCA Delta-NAC Human

Alpha Synuclein Delta-NAC Human Recombinant
Shipped with Ice Packs
In Stock

Description

Biological Role and Functional Implications

Alpha-synuclein is a natively unfolded protein critical in synaptic vesicle trafficking and dopamine regulation. The NAC region (61–95) is hydrophobic and amyloidogenic, driving aggregation into fibrils linked to Parkinson’s disease (PD) and Lewy body dementia. The Delta-NAC variant lacks this region, altering aggregation kinetics and interactions with other domains:

Key Functional Differences

AspectWild-Type SNCASNCA Delta-NAC
Aggregation PropensityHigh (NAC-driven fibril formation).Reduced (absence of NAC disrupts core).
Intramolecular InteractionsNAC-C-terminal interactions suppress nucleation.Disrupted interactions may expose N-terminal regions.
Chaperone ActivityRetained (C-terminal tail required).Unaffected (C-terminal intact).

Sources:

Research Applications and Findings

SNCA Delta-NAC is utilized to study alpha-synuclein’s aggregation mechanisms and neurotoxicity. Below are key studies and insights:

3.1. Aggregation Dynamics

The NAC region’s role in fibril formation is critical. Truncations or mutations in this region (e.g., S87N) accelerate nucleation by disrupting intramolecular interactions between N-terminal/pre-NAC and C-terminal regions. For example:

  • C-terminal Truncations: Removing residues 104–140 in wild-type SNCA reduces lag time for fibril formation, mimicking murine SNCA behavior. This suggests the C-terminal region suppresses nucleation.

  • S87N Mutation: Substituting serine at position 87 (NAC region) in human SNCA accelerates fibril formation, akin to murine SNCA. This highlights the NAC region’s regulatory role.

Sources:

3.2. Therapeutic Relevance

While SNCA Delta-NAC itself is not a therapeutic agent, its study informs strategies targeting alpha-synuclein aggregation:

  • NAC Inhibition: Compounds disrupting NAC-mediated aggregation (e.g., small molecules) may reduce fibril formation.

  • Glutathione Modulation: N-Acetylcysteine (NAC) supplementation in transgenic mice reduces SNCA overexpression and dopaminergic terminal loss, suggesting antioxidant strategies may mitigate SNCA toxicity.

Sources:

Product Specs

Introduction
Alpha-synuclein, a 140-amino acid neuronal protein, exhibits high heat resistance and exists in an intrinsically disordered state, primarily adopting random coil conformations. This protein has been implicated in the development of Parkinson's disease and other neurodegenerative disorders, potentially playing a role in the regulation of vesicular transport within neurons. Additionally, recent research suggests that alpha-synuclein possesses chaperone-like activity, which is compromised upon deletion of its C-terminal acidic tail (amino acids 96-140).
Description
This product consists of a recombinant human alpha-synuclein protein with a deletion in the NAC region (amino acids 61-95), expressed in E. coli. It is a single, non-glycosylated polypeptide chain composed of 111 amino acids, resulting in a molecular weight of 11.9 kDa. A 6-amino acid linker is incorporated into the protein sequence. Despite its calculated molecular weight, the protein exhibits a higher apparent molecular size on SDS-PAGE. Purification of the recombinant human alpha-synuclein delta-NAC protein is achieved through proprietary chromatographic methods.
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
The SNCA Delta-NAC protein solution is provided at a concentration of 1 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 7.5) and 100 mM NaCl.
Stability
For short-term storage (up to 4 weeks), the product can be kept at 4°C. For extended storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage. Repeated freezing and thawing of the product should be avoided.
Purity
The purity of this product is greater than 95.0%, as determined by SDS-PAGE analysis.
Synonyms
Alpha-synuclein, Non-A beta component of AD amyloid, Non-A4 component of amyloid precursor, NACP, PD1, PARK1, PARK4, MGC110988, a-Synuclein, SNCA.
Source
Escherichia Coli.
Amino Acid Sequence

MDVFMKGLSK AKEGVVAAAE KTKQGVAEAA GKTKEGVLYV GSKTKEGVVH GVATVAEKTK GTEIWMKKDQ LGKNEEGAPQ EGILEDMPVD PDNEAYEMPS EEGYQDYEPE A.

Q&A

What is the SNCA gene and what role does the NAC domain play in alpha-synuclein protein function?

The SNCA gene encodes alpha-synuclein (α-Syn), a protein predominantly found in presynaptic terminals that regulates neurotransmitter release and synaptic function. Structurally, α-Syn comprises three regions:

  • The N-terminal region, important for membrane binding

  • The non-amyloid-beta component (NAC) region (amino acids 61-95), crucial for aggregation

  • The C-terminal region, which has neuroprotective properties

The NAC domain is highly hydrophobic and plays a critical role in the protein's ability to form aggregates, which are characteristic of synucleinopathies like Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. This region is particularly significant in both the initial misfolding and subsequent fibril formation of α-Syn.

How does N-acetylcysteine (NAC) supplementation affect SNCA pathology in neurodegenerative diseases?

N-acetylcysteine (NAC) is an acetylated cysteine compound that functions as a glutathione precursor with antioxidant and anti-inflammatory properties. In relation to SNCA pathology, NAC has several documented effects:

  • NAC increases glutathione (GSH) levels in the brain, which can be depleted in neurodegenerative diseases

  • It reduces oxidative damage that contributes to α-Syn misfolding and aggregation

  • In transgenic mice overexpressing α-synuclein, NAC treatment has shown:

    • Increased striatal tyrosine hydroxylase positive terminal density

    • Reduced α-synuclein immunolabeling in brain tissue

    • Higher GSH levels in the substantia nigra

Research has demonstrated that oral NAC supplementation attenuated the loss of dopaminergic terminals associated with SNCA overexpression, even though the effect on brain glutathione levels was transient . This suggests NAC may have long-lasting neuroprotective effects despite relatively short-term impacts on GSH levels.

What methodologies are used to detect alpha-synuclein pathology in human tissue samples?

Detection of α-synuclein pathology in human tissue employs multiple complementary approaches:

  • Neuroimaging techniques:

    • Novel PET tracers like F0502B (a benzothiazole-ethenyl-phenol derivative) that selectively bind to α-Syn fibrils with high affinity (Kd of 5.75 nM)

    • Differential binding patterns that distinguish α-Syn aggregates from Aβ plaques and tau tangles

  • Histopathological methods:

    • Immunohistochemistry using phosphorylation-specific antibodies (p-α-Syn S129)

    • Thioflavin-S staining for beta-sheet structures in protein aggregates

    • Immunofluorescence with conformation-specific antibodies (such as 5G4)

  • Peripheral tissue sampling:

    • Skin biopsies to detect α-Syn inclusions, which have shown 60% positivity in epidermal and pilosebaceous unit cells in Parkinson's disease patients

    • Differential staining patterns between Parkinson's disease and atypical parkinsonism patients

These methods allow researchers to identify and characterize α-Syn pathology across different tissue types, enabling earlier detection and more precise monitoring of disease progression.

How does deletion of the NAC domain affect alpha-synuclein aggregation properties and neurotoxicity in human cellular models?

Deletion of the NAC domain (Δ61-95) from alpha-synuclein dramatically alters its aggregation properties and neurotoxicity in human cellular models:

  • Aggregation kinetics:

    • Wild-type α-Syn shows typical sigmoid aggregation curves

    • Δ-NAC variants exhibit greatly reduced fibril formation

    • The lag phase for aggregation is extended in Δ-NAC variants

  • Structural properties:

    • Δ-NAC variants form fewer β-sheet-rich structures

    • They demonstrate reduced Thioflavin-S (ThS) binding compared to wild-type α-Syn

    • Electron microscopy reveals fewer fibrillar structures and more amorphous aggregates

  • Cellular effects:

    • Reduced formation of intracellular inclusions

    • Decreased activation of ER stress responses

    • Attenuated mitochondrial dysfunction

    • Lower levels of oxidative stress markers

  • Neurotoxicity assessment:

    • Reduced cell death in neuronal cultures

    • Decreased inflammatory responses from microglia

    • Altered calcium homeostasis

    • Different patterns of synaptic dysfunction compared to full-length α-Syn

These findings confirm the NAC domain is crucial for pathological aggregation of α-Syn, and its deletion significantly reduces the protein's neurotoxic potential, suggesting potential therapeutic strategies targeting this region.

What are the interactions between N-acetylcysteine treatment and SNCA expression in patient-derived iPSC models?

Patient-derived induced pluripotent stem cell (iPSC) models reveal complex interactions between N-acetylcysteine (NAC) treatment and SNCA expression:

  • Effect on SNCA expression and processing:

    • NAC treatment does not directly alter SNCA mRNA levels

    • Post-translational modifications of α-Syn are affected, particularly phosphorylation at S129

    • Reduction in higher molecular weight α-Syn species formation

  • Oxidative stress parameters:

    • NAC treatment reduces reactive oxygen species (ROS) levels in iPSC-derived neurons

    • Increases GSH/GSSG ratio, reflecting improved redox status

    • Restores mitochondrial membrane potential

    • Decreases lipid peroxidation markers

  • Cellular phenotypes:

    • Improves neurite outgrowth and complexity

    • Enhances dopamine release in dopaminergic neurons

    • Protects against α-Syn-induced cell death

Studies of iPSCs derived from patients with SNCA triplication demonstrate that neurons differentiated from these cells show elevated SNCA expression (1.44-7.17 times higher than controls) and a twofold increase in α-Syn protein . These neurons exhibit increased expression of oxidative stress and protein aggregation-related genes (1.5-4 fold higher) and greater susceptibility to oxidative stress-induced cell death. NAC treatment can partially rescue this phenotype, with significantly reduced caspase-3 positive cells under hydrogen peroxide challenge .

How do imaging techniques differentiate between different conformational states of alpha-synuclein in human brain tissue?

Advanced imaging techniques can differentiate between conformational states of alpha-synuclein in human brain tissue:

  • PET imaging approaches:

    • Novel tracers like F0502B bind specifically to α-Syn fibrils

    • Selective binding is confirmed through counter-screening against Aβ and Tau aggregates

    • In vivo validation shows specific binding to α-Syn deposits in animal models

    • Brain/plasma ratio analysis confirms good brain penetration with B/P ratios increasing from 0.106 at 1 minute to 13.37 at 1 hour

  • Histological differentiation techniques:

    • Combined immunostaining for p-S129 α-Syn with ThS staining

    • Co-localization studies using confocal microscopy

    • Differential staining patterns between various aggregated forms

  • Experimental validation approaches:

    • Testing across multiple models:

      • AAV-α-Syn A53T-injected mouse models

      • SNCA transgenic mice treated with rotenone

      • Comparison with Aβ plaques in 5xFAD mice and tau tangles in P301S mice

    • Binding kinetics determination showing high affinity (Kd of 5.75 nM) for α-Syn aggregates

These techniques allow researchers to distinguish between different α-Syn conformations in situ, providing crucial tools for studying disease progression and evaluating potential therapeutics.

What are the optimal protocols for studying NAC effects on SNCA aggregation in human neuron models?

Optimized protocols for studying NAC effects on SNCA aggregation in human neuron models require careful methodological considerations:

  • Cell model selection and preparation:

    • Patient-derived iPSCs with SNCA mutations or multiplications provide ideal models

    • Differentiation into dopaminergic neurons following validated protocols

    • Confirmation of neuronal identity through tyrosine hydroxylase (TH) immunostaining

    • Quantification of baseline SNCA expression using qRT-PCR and western blotting

  • NAC treatment parameters:

    • Determination of optimal concentration range (typically 0.1-10 mM)

    • Time-course analysis from acute (hours) to chronic (weeks) treatment

    • Assessment of GSH levels to confirm NAC uptake and conversion

    • Control treatments with other antioxidants to distinguish mechanism-specific effects

  • Aggregation analysis methods:

    • Thioflavin-S/T fluorescence assays for β-sheet-rich aggregates

    • Immunostaining for phosphorylated α-Syn (p-S129)

    • Biochemical fractionation to isolate soluble and insoluble protein species

    • Electron microscopy for direct visualization of aggregate morphology

  • Functional outcome measurements:

    • Mitochondrial function assessment (membrane potential, respiratory capacity)

    • Cell viability under baseline and stressed conditions

    • Neurite morphology and synaptic marker expression

    • Electrophysiological properties and neurotransmitter release

  • Data analysis considerations:

    • Dose-response relationship modeling

    • Statistical analysis accounting for inter-clone variability

    • Correlation between GSH levels and functional outcomes

    • Multivariate analysis of aggregation parameters and cellular phenotypes

These protocols enable systematic investigation of NAC's effects on SNCA aggregation and associated neuronal dysfunction in human cellular models.

What methods are most effective for quantifying the interactions between the NAC domain and N-acetylcysteine in experimental systems?

Quantifying interactions between the NAC domain and N-acetylcysteine requires sophisticated methodological approaches:

  • Biophysical interaction analysis:

    TechniqueApplicationAdvantagesLimitations
    Isothermal Titration CalorimetryDirect measurement of binding thermodynamicsLabel-free, quantitativeRequires substantial protein amounts
    Surface Plasmon ResonanceReal-time binding kineticsHigh sensitivity, minimal sampleSurface immobilization may affect interactions
    Microscale ThermophoresisSolution-phase binding analysisLow sample consumptionRequires fluorescent labeling
    NMR SpectroscopyAtomic-level interaction mappingDetailed structural informationSize limitations, expensive
  • Biochemical approaches:

    • Site-directed mutagenesis of key NAC domain residues

    • Chemical cross-linking followed by mass spectrometry

    • Hydrogen-deuterium exchange mass spectrometry

    • Fluorescence-based interaction assays (FRET, fluorescence quenching)

  • Cellular visualization methods:

    • Proximity ligation assays in fixed cells

    • FRET-based reporters in live cells

    • Split fluorescent/luminescent protein complementation

    • Super-resolution microscopy with dual labeling

  • Computational modeling:

    • Molecular dynamics simulations of NAC-NAC interactions

    • Docking studies to identify potential binding sites

    • Quantum mechanical calculations for electron distribution effects

    • Machine learning approaches to predict interaction sites from experimental data

  • Functional validation:

    • Aggregation kinetics with and without NAC treatment

    • Protease protection assays to identify structural changes

    • Circular dichroism to detect secondary structure alterations

    • Cellular phenotype rescue experiments

These methods provide complementary information about the molecular interactions that underlie the potential therapeutic effects of N-acetylcysteine on alpha-synuclein aggregation.

How can researchers effectively design experiments to distinguish between NAC's antioxidant effects and its direct effects on alpha-synuclein?

Designing experiments to distinguish between NAC's antioxidant effects and direct effects on alpha-synuclein requires careful controls and mechanistic isolation:

  • Pathway-specific interventions:

    • GSH synthesis inhibition (e.g., buthionine sulfoximine) to block antioxidant effects

    • Structural NAC analogs lacking thiol groups to eliminate direct antioxidant activity

    • Site-directed mutagenesis of cysteine residues in α-Syn

    • Targeted antioxidants with different mechanisms (e.g., mitochondria-specific vs. cytosolic)

  • Oxidative stress decoupling:

    • Oxygen-reduced experimental conditions

    • Genetic models with constitutively active Nrf2 to elevate antioxidant responses

    • Time-course analysis separating immediate (antioxidant) from delayed (protein interaction) effects

    • Redox-insensitive α-Syn mutants

  • Direct binding assessments:

    • In vitro aggregation assays with purified components

    • Analysis of NAC effects on pre-formed fibrils

    • Competition assays with known NAC domain binding partners

    • Mass spectrometry to detect NAC-α-Syn adducts

  • Cellular compartmentalization studies:

    • Subcellular fractionation to localize effects

    • Organelle-targeted NAC derivatives

    • Correlation of local GSH/GSSG ratios with α-Syn aggregation

    • Visualization of redox-sensitive fluorescent proteins alongside α-Syn

  • Mechanistic biomarkers:

    • Oxidative modification profiling of α-Syn

    • Monitoring NF-κB signaling alongside α-Syn pathology

    • Assessment of proteasomal and autophagic flux

    • Analysis of protein disulfide status

These experimental approaches can help delineate whether NAC's beneficial effects on SNCA pathology stem primarily from its well-established antioxidant properties or involve direct interactions with the protein itself.

How might research on SNCA Delta-NAC variants inform therapeutic strategies for synucleinopathies?

Research on SNCA Delta-NAC variants offers several promising avenues for therapeutic development:

  • Peptide-based strategies:

    • Development of peptides that bind to the NAC domain to prevent aggregation

    • Design of Delta-NAC mimetic peptides that could act as dominant negatives

    • Cell-penetrating peptides targeting intracellular α-Syn

    • Stabilized peptides that maintain optimal conformation for NAC domain binding

  • Small molecule approaches:

    • Compounds designed to specifically bind the NAC region

    • Structure-based drug design informed by NAC domain interactions

    • Screening compounds that stabilize non-aggregation-prone conformations

    • Development of imaging agents that could double as therapeutics

  • Gene therapy possibilities:

    • Expression of modified α-Syn lacking the NAC domain

    • CRISPR-based approaches to modify endogenous SNCA

    • Antisense oligonucleotides targeting NAC domain expression

    • Viral delivery of aggregation-resistant α-Syn variants

  • Combination therapies:

    • NAC supplementation coupled with NAC domain-targeting agents

    • Enhancing clearance mechanisms alongside aggregation prevention

    • Targeting multiple domains of α-Syn simultaneously

    • Addressing both neuronal and glial aspects of pathology

What are the implications of studying SNCA-NAC interactions for personalized medicine approaches to neurodegenerative diseases?

The study of SNCA-NAC interactions has significant implications for developing personalized medicine approaches:

  • Patient stratification factors:

    • Genetic variants affecting the NAC domain or α-Syn expression

    • Oxidative stress biomarkers that might predict NAC efficacy

    • Proteomic profiles indicating different α-Syn conformational states

    • Imaging characteristics revealing specific aggregate patterns

  • Tailored therapeutic approaches:

    • Individualized NAC dosing based on oxidative stress profiles

    • Combined therapies targeting patient-specific pathways

    • Monitoring GSH levels to optimize treatment regimens

    • Adjusting therapy based on SNCA expression levels

  • Biomarker development:

    • Identification of α-Syn conformational state markers in biofluids

    • Development of imaging agents for specific α-Syn forms

    • Peripheral tissue markers (skin biopsies) for disease monitoring

    • Digital biomarkers that correlate with molecular pathology

  • Clinical implementation considerations:

    • Development of companion diagnostics for NAC therapy

    • Monitoring protocols based on individual disease progression patterns

    • Integration with other personalized approaches

    • Longitudinal assessment strategies

Research on patient-derived iPSCs from individuals with SNCA triplication has already demonstrated variability in response to oxidative stress and potential therapeutic interventions , highlighting the need for personalized approaches that consider individual genetic and molecular profiles.

How do findings from SNCA Delta-NAC research translate between different experimental models and human patients?

Translational challenges in SNCA Delta-NAC research require careful consideration of model-specific differences:

  • Comparative model characteristics:

    Model SystemAdvantagesLimitationsTranslational Value
    Patient-derived iPSCsHuman genetic background, disease mutationsLack aging, limited complexityHigh for cellular mechanisms
    Transgenic miceIn vivo system, behavioral assessmentSpecies differences in SNCAModerate for systemic effects
    AAV-injected modelsRapid pathology developmentArtificial overexpressionGood for proof-of-concept
    Rotenone-treated modelsCombines genetic and environmental factorsNon-specific toxicityHigh for environmental interactions
    Human post-mortem tissueAuthentic human pathologyEnd-stage disease onlyGold standard reference
  • Species-specific considerations:

    • Differences in SNCA isoform expression between humans and rodents

    • Variation in NAC domain interactions with cellular components

    • Species-specific post-translational modifications

    • Differences in clearance mechanism efficiencies

  • Methodological translation strategies:

    • Multi-model validation of findings

    • Parallel studies in human and animal systems

    • Development of humanized animal models

    • Comparative analyses of binding kinetics across species

  • Clinical translation approaches:

    • Focus on conserved mechanisms across models

    • Validation in human post-mortem tissue

    • Development of translatable biomarkers

    • Consideration of aging and comorbidity factors absent in most models

What emerging technologies show promise for targeting SNCA Delta-NAC interactions with therapeutic potential?

Several cutting-edge technologies show promise for therapeutic targeting of SNCA Delta-NAC interactions:

  • Advanced imaging and structural biology:

    • Cryo-EM determination of human α-Syn fibril structures with atomic resolution

    • Development of PET tracers with specificity for different α-Syn conformations

    • Rational design of molecules targeting the NAC domain based on structural insights

    • Live-cell imaging of NAC domain accessibility in different conformational states

  • Novel therapeutic modalities:

    • Antisense oligonucleotides targeting NAC domain expression

    • CRISPR-based approaches for precise SNCA modification

    • Intrabodies specifically designed against the NAC domain

    • Conformationally-restricted peptides mimicking protective SNCA states

  • Improved delivery systems:

    • Brain-penetrant nanoparticles for NAC and related compounds

    • Extracellular vesicles for delivery of therapeutic proteins

    • Focused ultrasound to enhance BBB permeability for targeted delivery

    • Cell-penetrating peptides conjugated to therapeutic agents

  • Advanced cellular models:

    • Brain organoids incorporating multiple cell types

    • Microfluidic systems modeling regional vulnerability

    • Patient-derived 3D models with accelerated aging

    • Multiparametric phenotypic screening platforms

These technologies could overcome current limitations in targeting specific domains of alpha-synuclein and potentially lead to more effective and precise interventions for synucleinopathies.

How can multi-omics approaches enhance our understanding of NAC-mediated protection against SNCA pathology?

Multi-omics approaches offer powerful new insights into NAC-mediated protection against SNCA pathology:

  • Integrated systems biology approaches:

    • Transcriptomics to identify NAC-responsive gene networks

    • Proteomics to map changes in α-Syn post-translational modifications

    • Metabolomics to track GSH metabolism and oxidative stress markers

    • Lipidomics to examine membrane composition changes affecting α-Syn binding

  • Single-cell technologies:

    • Single-cell RNA-seq to identify cell-type-specific responses to NAC

    • Spatial transcriptomics to map regional effects in brain tissue

    • CyTOF for high-dimensional protein analysis at single-cell resolution

    • Spatial proteomics to localize NAC effects within subcellular compartments

  • Network biology applications:

    • Protein-protein interaction networks centered on SNCA

    • Pathway analysis of NAC effects beyond antioxidant mechanisms

    • Temporal network changes during disease progression and treatment

    • Identification of key network nodes as intervention points

  • Translational applications:

    • Development of biomarker signatures for NAC responsiveness

    • Identification of synergistic drug targets from network analysis

    • Patient stratification based on molecular profiles

    • Optimization of NAC dosing and timing based on systems modeling

These approaches can reveal how NAC's effects extend beyond simple antioxidant activity to influence complex cellular networks involved in α-Syn homeostasis, potentially identifying previously unrecognized therapeutic targets and biomarkers.

What methodological advances are needed to better study the interaction between the NAC domain of alpha-synuclein and N-acetylcysteine in human brain tissue?

Advancing our understanding of interactions between the NAC domain and N-acetylcysteine in human brain tissue requires methodological innovations:

  • Improved tissue preservation and processing:

    • Rapid fixation protocols that preserve protein-small molecule interactions

    • Tissue clearing techniques compatible with small molecule detection

    • Cryogenic sample preparation to maintain native states

    • Microdissection approaches for region-specific analysis

  • Advanced microscopy methods:

    • Super-resolution techniques reaching 10-20 nm resolution

    • Label-free chemical imaging (CARS, SRS) for small molecule detection

    • Correlative light and electron microscopy for multilevel analysis

    • Expansion microscopy to physically magnify interaction sites

  • Molecular detection innovations:

    • In situ proximity ligation assays adapted for small molecules

    • MALDI imaging mass spectrometry with improved spatial resolution

    • Conformation-specific probes for different α-Syn states

    • Click chemistry approaches for NAC derivative localization

  • Human tissue model development:

    • Advanced organoids incorporating aged cellular phenotypes

    • Humanized mouse models with patient-derived SNCA

    • Post-mortem tissue slice culture systems

    • Microfluidic devices incorporating human tissue samples

  • Computational modeling advances:

    • Improved force fields for protein-small molecule interactions

    • Machine learning approaches to predict binding from limited data

    • Integration of imaging data with molecular simulations

    • Network models incorporating spatial information

These methodological advances would help bridge the gap between in vitro studies and human disease, providing more accurate insights into how NAC might interact with the NAC domain of alpha-synuclein in the complex environment of the human brain.

Product Science Overview

Background of Alpha Synuclein Delta-NAC (Human Recombinant)

Alpha-Synuclein (α-Synuclein) is a presynaptic protein predominantly found in neural tissue. It is a major component of Lewy bodies, which are protein aggregates that are a hallmark of Parkinson’s Disease (PD) and other neurodegenerative disorders . The protein is composed of 140 amino acids and is known for its role in synaptic vesicle regulation and neurotransmitter release .

Delta-NAC Variant: The Delta-NAC variant of alpha-synuclein is a deletion mutant that lacks the Non-Amyloid Component (NAC) region, specifically amino acids 61-95 . This region is crucial for the aggregation properties of alpha-synuclein, and its absence in the Delta-NAC variant makes it a valuable tool for studying the mechanisms of protein aggregation and its implications in neurodegenerative diseases .

Recombinant Production: The human recombinant Alpha Synuclein Delta-NAC is produced in Escherichia coli (E. coli) and purified to ensure high purity and consistency . The recombinant protein is typically expressed as a single, non-glycosylated polypeptide chain of 111 amino acids, with an additional 6 amino acids added as a linker . The molecular mass of this recombinant protein is approximately 11.9 kDa .

Applications: The recombinant Alpha Synuclein Delta-NAC is used in various research applications, including:

  • Aggregation Studies: Understanding the aggregation properties of alpha-synuclein and its role in neurodegenerative diseases .
  • Seeding Experiments: Investigating the seeding and propagation of alpha-synuclein aggregates .
  • Molecular Standards: Serving as a standard in biochemical assays and experiments .

Storage and Stability: The recombinant protein is typically stored in a buffer containing 20mM Tris-HCl (pH 7.5) and 100mM NaCl . For long-term storage, it is recommended to keep the protein at -20°C and avoid multiple freeze-thaw cycles to maintain its stability .

Conclusion: The human recombinant Alpha Synuclein Delta-NAC is a crucial tool for researchers studying the pathogenesis of Parkinson’s Disease and related neurodegenerative disorders. By providing a highly purified and consistent protein, it enables detailed investigations into the molecular mechanisms underlying protein aggregation and its impact on neuronal function.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.