SYT3 Human

Synaptotagmin III Human Recombinant
Shipped with Ice Packs
In Stock

Description

Functional Role in Synaptic Transmission

SYT3 accelerates synaptic vesicle resupply during high-frequency neuronal activity, ensuring reliable neurotransmission:

  • Vesicle Replenishment: SYT3 knockout (KO) mice exhibit slower recovery from synaptic depression and reduced readily releasable vesicle pools, indicating its role in rapid vesicle trafficking .

  • Short-Term Plasticity: SYT3 mediates facilitation at synapses with low release probability, enhancing signal fidelity during repetitive stimulation .

  • Calcium Sensitivity: Acts as a high-affinity Ca²⁺ sensor, triggering vesicle replenishment during residual Ca²⁺ elevations post-activity .

Clinical Relevance

SYT3 dysfunction is linked to neurological disorders:

  • Epilepsy and Autism: Mutations in SYT3 are associated with impaired synaptic transmission and neurodevelopmental conditions .

  • Therapeutic Potential: Targeting SYT3 could restore neurotransmitter balance in diseases characterized by synaptic fatigue (e.g., epilepsy) .

Research Tools and Recombinant SYT3

The recombinant SYT3 Human protein (Novatein Biosciences) is widely used for biochemical studies:

ParameterSpecification
Purity>85% (SDS-PAGE)
Buffer20 mM Tris-HCl (pH 8.0), 10% glycerol, 0.4 M urea
Storage-20°C (long-term); 4°C for short-term use
Functional AssaysCa²⁺-dependent lipid binding, vesicle fusion experiments

Key Research Findings

  • Knockout Models: Syt3 KO mice show enhanced short-term depression and slower recovery at synapses, confirming its role in sustaining high-frequency transmission .

  • Vesicle Trafficking: Computational models suggest SYT3 accelerates vesicle docking at active zones, combating synaptic depression .

  • Disease Mechanisms: Impaired SYT3 function disrupts synaptic homeostasis, contributing to excitability imbalances in epilepsy .

Future Directions

Ongoing research aims to:

  1. Develop SYT3-targeted gene therapies for neurodevelopmental disorders .

  2. Elucidate SYT3’s interplay with other synaptotagmins (e.g., SYT1, SYT7) .

  3. Explore SYT3’s postsynaptic roles in receptor internalization and synaptic plasticity .

Product Specs

Introduction
Synaptotagmin-3 (SYT3), a member of the synaptotagmin family, participates in Ca(2+)-dependent exocytosis of secretory vesicles. This involvement is facilitated by Ca(2+) and phospholipid binding to the C2 domain. SYT3 may also act as a Ca(2+) sensor during vesicular trafficking and exocytosis.
Description
Recombinantly produced in E.coli, SYT3 Human is a single, non-glycosylated polypeptide chain. It comprises 540 amino acids (76-590) and has a molecular weight of 57.7 kDa. The protein includes a 25 amino acid His-tag at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
A clear solution that has undergone sterile filtration.
Formulation
The SYT3 solution (1mg/ml) is prepared in a buffer containing 20mM Tris-HCl (pH 8.0), 10% glycerol, and 0.4M Urea.
Stability
For short-term storage (2-4 weeks), the solution should be kept at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. To further ensure stability during long-term storage, consider adding a carrier protein (0.1% HSA or BSA). It is important to avoid subjecting the solution to multiple freeze-thaw cycles.
Purity
SDS-PAGE analysis indicates a purity greater than 85%.
Synonyms
Synaptotagmin III, SYT3, SytIII, synaptotagmin-3.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSEFWKLCW VPWRDKGGSA VGGGPLRKDL GPGVGLAGLV GGGGHHLAAG LGGHPLLGGP HHHAHAAHHP PFAELLEPGS LGGSDTPEPS YLDMDSYPEA AAAAVAAGVK PSQTSPELPS EGGAGSGLLL LPPSGGGLPS AQSHQQVTSL APTTRYPALP RPLTQQTLTS QPDPSSEERP PALPLPLPGG EEKAKLIGQI KPELYQGTGP GGRRSGGGPG SGEAGTGAPC GRISFALRYL YGSDQLVVRI LQALDLPAKD SNGFSDPYVK IYLLPDRKKK FQTKVHRKTL NPVFNETFQF SVPLAELAQR KLHFSVYDFD RFSRHDLIGQ VVLDNLLELA EQPPDRPLWR DIVEGGSEKA DLGELNFSLC YLPTAGRLTV TIIKASNLKA MDLTGFSDPY VKASLISEGR RLKKRKTSIK KNTLNPTYNE ALVFDVAPES VENVGLSIAV VDYDCIGHNE VIGVCRVGPD AADPHGREHW AEMLANPRKP VEHWHQLVEE KTVTSFTKGS KGLSEKENSE.

Q&A

What is Synaptotagmin-3 (SYT3) and what is its primary function in the nervous system?

Synaptotagmin-3 (SYT3) is a gene-encoded protein that functions as a high-affinity Ca²⁺ sensor in the brain. Its primary role is enabling synaptic transmission by facilitating the replenishment of chemical neurotransmitters across synapses (gaps between neurons). SYT3 is critically involved in maintaining reliable synaptic transmission, particularly during periods of sustained neuronal activity. When neurons are highly active, they can deplete their neurotransmitter supply, potentially causing communication breakdown. SYT3 activates a "boost mode" that rapidly replenishes these neurotransmitters, ensuring continued neuronal communication .

How does SYT3 differ structurally and functionally from other synaptotagmin family members?

SYT3 is distinguished from other synaptotagmin family members primarily by its high calcium sensitivity and specific role in vesicle replenishment. While multiple synaptotagmins function as calcium sensors, SYT3 specifically operates as a high-affinity Ca²⁺ sensor that responds to submicromolar calcium concentrations. Functionally, SYT3 drives vesicle replenishment and influences short-term synaptic plasticity. It works in coordination with other synaptotagmins like SYT7, with each fulfilling complementary roles in synaptic enhancement . Structurally, like other synaptotagmins, SYT3 contains C2 domains that bind both calcium and phospholipid membranes, but with different binding kinetics and affinities that enable its specialized function in the vesicle resupply process .

What neurological disorders have been associated with SYT3 gene mutations?

Mutations in the SYT3 gene have been implicated in several neurological disorders, most notably autism spectrum disorder (ASD) and epilepsy. These associations highlight the critical role SYT3 plays in maintaining proper neuronal communication and brain function . The connection between SYT3 dysfunction and these disorders likely stems from its fundamental role in synaptic transmission and neurotransmitter regulation. Imbalances in neurotransmitter release—which can occur when SYT3 function is compromised—are known underlying causes for many neurological disorders. This relationship between SYT3 mutations and neurological conditions underscores the potential for developing targeted therapeutic approaches focused on this protein .

How does SYT3 specifically mediate the calcium-dependent acceleration of synaptic vesicle replenishment?

SYT3 mediates calcium-dependent acceleration of synaptic vesicle replenishment through a sophisticated mechanism involving both calcium sensing and vesicle docking. Upon detecting submicromolar presynaptic Ca²⁺ signals, SYT3 undergoes conformational changes that enable it to bind both calcium ions and phospholipid membranes . This dual binding capacity allows SYT3 to promote the transition of loosely docked vesicles to tightly docked, primed states ready for release.
Biophysical modeling suggests that SYT3 accomplishes this by interacting with assembled SNARE complexes when calcium is present . In the absence of SYT3 (as demonstrated in knockout models), synapses exhibit enhanced short-term depression and significantly slower recovery from depression, with recovery becoming insensitive to presynaptic residual calcium. During sustained neuronal firing, SYT3 both accelerates vesicle replenishment and increases the readily releasable pool size, ensuring continuous neurotransmission even during high-frequency activity .

What are the molecular interactions between SYT3 and SNARE complexes during exocytosis?

The molecular interactions between SYT3 and SNARE complexes represent a critical mechanism in calcium-regulated exocytosis. When calcium levels rise, SYT3 binds to both phospholipid membranes and assembled SNARE complexes, with both interactions contributing to triggering exocytosis . The C2 domains of SYT3 are instrumental in these interactions, undergoing calcium-dependent conformational changes that enable binding to the SNARE complex proteins (syntaxin, SNAP-25, and synaptobrevin/VAMP).
This calcium-induced binding to SNARE complexes helps regulate the fusion of vesicles with the presynaptic membrane. The interaction likely stabilizes partially assembled SNARE complexes or promotes their full assembly, facilitating membrane fusion and subsequent neurotransmitter release . Unlike some other synaptotagmins that primarily trigger immediate release, SYT3's interaction with SNAREs appears more specialized toward sustaining release during periods of high activity by promoting vesicle docking and priming rather than immediate fusion .

How do SYT3 and SYT7 cooperatively influence short-term synaptic plasticity?

SYT3 and SYT7 cooperatively influence short-term synaptic plasticity through complementary but distinct mechanisms. Both are high-affinity calcium sensors, but they appear to target different aspects of the vesicle release and replenishment cycle. Research indicates that SYT3 mediates both vesicle replenishment and facilitation by promoting the transition of loosely docked vesicles to tightly docked, primed states .
Under conditions of low release probability, SYT3 mediates short-term facilitation. Importantly, SYT3 can promote synaptic enhancement in conjunction with SYT7, suggesting they work together in a coordinated fashion rather than redundantly . This cooperation likely involves SYT7 playing a more direct role in asynchronous release and certain forms of facilitation, while SYT3 focuses more on vesicle replenishment and availability. Their combined action ensures both the immediate response to calcium influx and the sustained ability to maintain transmission during prolonged activity, providing neurons with a sophisticated system for short-term plasticity regulation.

What are the most effective methods for studying SYT3 function in vitro versus in vivo?

For studying SYT3 function, complementary in vitro and in vivo approaches yield the most comprehensive insights:
In Vitro Methods:

  • Recombinant Protein Studies: Using purified SYT3 protein (like the E.coli-expressed human recombinant SYT3) to examine binding properties, structural characteristics, and interactions with calcium and phospholipids .

  • Cell Culture Models: Transfection of cultured neurons with wild-type or mutant SYT3 constructs to assess localization and function.

  • shRNA Knockdown: Using shRNA plasmid kits to selectively reduce SYT3 expression in cultured cells to study functional consequences .

  • Electrophysiological Recordings: Patch-clamp recordings from cultured neurons to measure synaptic transmission parameters.
    In Vivo Methods:

  • Knockout Models: Generation of SYT3 knockout mice to study the physiological impact of complete SYT3 absence, as demonstrated in research showing enhanced short-term depression and slower recovery .

  • CRISPR Gene Editing: Using CRISPR/Cas9 systems to create precise modifications to the SYT3 gene, allowing for targeted study of specific domains or mutations .

  • Conditional Knockouts: Temporally or spatially restricted deletion of SYT3 to avoid developmental compensation.

  • In Vivo Electrophysiology: Recording synaptic transmission in intact brain slices or in vivo to assess SYT3 function in native neural circuits.
    The choice between these approaches depends on the specific research question, with in vitro methods offering greater molecular control and in vivo methods providing physiological relevance.

What controls and validation steps are essential when using SYT3 knockout or knockdown models?

When using SYT3 knockout or knockdown models, several critical controls and validation steps are essential to ensure experimental rigor:
For Genetic Modifications:

  • Verification of Target Specificity: Confirm that CRISPR guide RNAs or shRNA constructs specifically target SYT3 without off-target effects .

  • Knockout/Knockdown Validation: Verify SYT3 deletion/reduction at both mRNA (qPCR) and protein levels (Western blot), with antibody specificity validated using knockout tissues as negative controls.

  • Phenotype Rescue Experiments: Reintroduce wild-type SYT3 to confirm that observed phenotypes are directly attributable to SYT3 absence rather than off-target effects or developmental compensation.
    Functional Validation:

  • Electrophysiological Characterization: Compare basic synaptic transmission parameters between knockout/knockdown and control preparations.

  • Calcium Sensitivity Testing: Verify altered responses to varying calcium concentrations in SYT3-deficient synapses.

  • Vesicle Pool Assessment: Quantify readily releasable pool size and replenishment rates, which should be specifically affected by SYT3 manipulation .
    Additional Controls:

  • Evaluation of Other Synaptotagmins: Assess potential compensatory changes in expression of related proteins, particularly SYT7 which shares some functional overlap .

  • Developmental Timeline Analysis: Determine whether phenotypes result from acute loss of function or developmental adaptations.

  • Background Strain Consistency: Ensure genetic background is consistent between experimental and control animals to avoid strain-specific confounds.
    These validation steps ensure that experimental outcomes can be confidently attributed to SYT3 manipulation rather than experimental artifacts.

What are the recommended protocols for analyzing SYT3 interactions with calcium and phospholipid membranes?

Analysis of SYT3 interactions with calcium and phospholipid membranes requires specialized techniques that can detect these dynamic molecular interactions:
Biochemical Interaction Assays:

  • Calcium Binding Assays: Using isothermal titration calorimetry (ITC) or microscale thermophoresis to determine calcium binding affinities of purified SYT3 C2 domains.

  • Liposome Co-sedimentation Assays: Incubating purified SYT3 protein with artificial liposomes containing various phospholipid compositions, then assessing binding through centrifugation and subsequent analysis of pellet versus supernatant fractions .

  • FRET-Based Interaction Assays: Employing fluorescence resonance energy transfer between labeled SYT3 and membrane components to measure dynamic interactions in real-time.
    Structural Analysis:

  • X-ray Crystallography or Cryo-EM: Determining the three-dimensional structure of SYT3 in both calcium-bound and calcium-free states.

  • NMR Spectroscopy: Analyzing conformational changes in SYT3 C2 domains upon calcium binding.
    Functional Correlation:

  • Mutagenesis Studies: Systematically mutating calcium-binding residues in the C2 domains to correlate structural features with functional outcomes.

  • Lipid Composition Variations: Testing SYT3 binding to membranes with different phospholipid compositions to determine specificity.

  • Calcium Concentration Gradients: Examining SYT3 responses across physiologically relevant calcium concentration ranges (nanomolar to micromolar).
    When conducting these analyses, it's critical to use highly purified recombinant SYT3 protein (such as the human recombinant described in the literature) under carefully controlled buffer conditions that mimic physiological environments . Additionally, comparing SYT3 binding properties with those of other synaptotagmin family members provides valuable context for understanding its unique functional characteristics.

How can SYT3 dysfunction contribute to the pathophysiology of autism spectrum disorders and epilepsy?

SYT3 dysfunction can contribute to the pathophysiology of autism spectrum disorders (ASD) and epilepsy through several potential mechanisms:
Synaptic Transmission Dysregulation:

  • SYT3 mutations can impair the calcium-dependent replenishment of synaptic vesicles, leading to accelerated depletion of neurotransmitters during sustained activity . This depletion particularly affects information processing during high-frequency neuronal firing, potentially contributing to altered sensory processing in ASD or triggering seizure activity in epilepsy.
    Excitatory/Inhibitory Imbalance:

  • By regulating neurotransmitter availability, SYT3 helps maintain the balance between excitatory and inhibitory transmission. Dysfunction can lead to imbalances that contribute to both ASD (often associated with altered E/I ratios) and epilepsy (characterized by excessive excitation) .
    Synaptic Plasticity Abnormalities:

  • SYT3 mediates certain forms of short-term synaptic plasticity, including facilitation under conditions of low release probability . Alterations in these plasticity mechanisms can affect learning, memory, and information processing, potentially contributing to cognitive and behavioral symptoms of ASD.
    Developmental Consequences:

  • Beyond its acute role in synaptic transmission, SYT3 dysfunction during critical developmental periods might lead to altered neural circuit formation, contributing to the long-term structural and functional abnormalities observed in neurodevelopmental disorders like ASD .
    Understanding these pathophysiological mechanisms provides potential targets for therapeutic intervention, with approaches potentially focusing on compensating for impaired vesicle replenishment or restoring proper calcium sensitivity at synapses affected by SYT3 mutations.

What are the most promising therapeutic approaches targeting SYT3 for neurological disorders?

Therapeutic approaches targeting SYT3 for neurological disorders are still emerging, but several promising strategies can be identified based on current research:
Gene Therapy Approaches:

How might biomarkers of SYT3 function be developed for clinical applications?

Development of biomarkers for SYT3 function could significantly advance both diagnosis and treatment monitoring for associated neurological disorders:
Genetic Biomarkers:

  • Mutation Screening Panels: Development of comprehensive genetic panels to identify pathogenic mutations in SYT3 and related genes in patients with epilepsy or ASD .

  • Expression Quantification: Measuring SYT3 mRNA levels in accessible tissues as a potential correlate of expression in the nervous system.

  • Epigenetic Markers: Identifying epigenetic modifications that regulate SYT3 expression as potential biomarkers.
    Functional Biomarkers:

  • Neurophysiological Measures: EEG-based assessments of short-term synaptic plasticity that might reflect SYT3 function, particularly during high-frequency stimulation protocols.

  • Neuroimaging Biomarkers: Advanced functional MRI paradigms designed to probe activity-dependent neurotransmission that depends on SYT3 function.

  • Synaptic Vesicle Markers: Development of PET ligands that bind to components of the synaptic vesicle cycle influenced by SYT3.
    Surrogate Biomarkers:

  • Patient-Derived Models: Using induced pluripotent stem cells (iPSCs) from patients to generate neurons for personalized assessment of synaptic function and SYT3 activity.

  • Blood-Based Protein Panels: Identifying plasma protein signatures that correlate with SYT3 function or dysfunction in the CNS.

  • Metabolomic Profiles: Detecting metabolic changes resulting from altered synaptic transmission associated with SYT3 variants.
    Development of these biomarkers would benefit from the existing research tools, including SYT3 knockout models, CRISPR editing systems, and high-quality antibodies for protein detection . The ultimate goal would be creating clinically accessible measures that could facilitate early diagnosis, patient stratification for clinical trials, and monitoring of treatment responses in conditions associated with SYT3 dysfunction.

What are the challenges and solutions in producing high-quality recombinant SYT3 protein for structural and functional studies?

Producing high-quality recombinant SYT3 protein presents several challenges that require specific technical solutions:
Key Challenges:

  • Protein Solubility Issues: The hydrophobic regions and membrane-binding domains of SYT3 can cause aggregation and poor solubility.

  • Maintaining Native Conformation: Ensuring that recombinant SYT3 retains its calcium-binding properties and proper folding.

  • Purification Complexity: Removing contaminants while preserving functional integrity of the protein.

  • Post-Translational Modifications: Bacterial expression systems lack the machinery for human post-translational modifications that might be important for SYT3 function.
    Technical Solutions:

  • Expression System Optimization:

    • Using specialized E. coli strains designed for membrane-associated proteins .

    • Employing eukaryotic expression systems (insect or mammalian cells) for proteins requiring complex folding or modifications.

    • Expressing only the soluble domains (C2A and C2B) separately when full-length protein is problematic.

  • Fusion Tags and Solubility Enhancers:

    • Utilizing solubility-enhancing tags such as SUMO, MBP, or GST.

    • Incorporating a well-designed His-tag system as seen in the recombinant SYT3 described in the literature .

    • Employing removable tags with precise protease cleavage sites.

  • Buffer Optimization:

    • Including stabilizing agents such as glycerol (10% as used in some SYT3 preparations) .

    • Careful pH selection to maintain protein stability.

    • Using mild solubilizing agents like the urea (0.4M) mentioned in some SYT3 formulations when necessary .

  • Purification Strategy:

    • Multi-step purification combining affinity chromatography, ion exchange, and size exclusion.

    • Implementing rigorous quality control including SDS-PAGE to verify purity (>85% as standard) .

    • Mass spectrometry validation of the final product.
      By addressing these challenges systematically, researchers can produce the high-quality SYT3 recombinant protein necessary for detailed structural and functional studies that advance our understanding of this important synaptic protein.

How should researchers design experiments to distinguish between the roles of SYT3 and other synaptotagmin family members?

Designing experiments to distinguish between SYT3 and other synaptotagmin family members requires careful methodological approaches:
Genetic Manipulation Strategies:

  • Single and Combinatorial Knockouts:

    • Generate single knockouts for SYT3 and related synaptotagmins (particularly SYT7) to identify unique phenotypes .

    • Create double or triple knockout combinations to reveal functional redundancy or synergistic effects.

    • Use CRISPR/Cas9 systems for precise genomic editing rather than traditional knockout approaches when subtle modifications are needed .

  • Domain Swap Experiments:

    • Create chimeric proteins containing domains from different synaptotagmins to identify which regions confer specific functional properties.

    • Express these chimeras in knockout backgrounds to assess functional rescue capability.
      Functional Discrimination Approaches:

  • Calcium Sensitivity Profiling:

    • Compare the calcium concentration response curves for different synaptotagmins under identical experimental conditions.

    • For SYT3, focus on its response to submicromolar calcium concentrations that distinguish it from lower-affinity family members .

  • Temporal Resolution Studies:

    • Use high-resolution electrophysiology to distinguish between immediate synchronous release (often mediated by SYT1/2) versus the facilitation and vesicle replenishment functions attributed to SYT3 .

    • Employ optical methods with genetically encoded calcium indicators to correlate calcium signals with specific synaptotagmin actions.

  • Subcellular Localization Analysis:

    • Use super-resolution microscopy with specific antibodies or tagged proteins to determine the precise localization of different synaptotagmins.

    • Correlate localization patterns with functional domains within the presynaptic terminal.
      Biochemical Differentiation:

  • Isoform-Specific Interaction Studies:

    • Identify protein binding partners unique to SYT3 versus other family members.

    • Use co-immunoprecipitation with highly specific antibodies to isolate SYT3 complexes.

    • Perform targeted proteomics to identify the specific protein interaction network for SYT3.

  • Lipid Binding Selectivity:

    • Compare the phospholipid binding profiles of different synaptotagmins under various calcium concentrations.

    • Analyze how these interactions change during different phases of the synaptic vesicle cycle.
      These approaches, when systematically applied, allow researchers to delineate the specific contributions of SYT3 in relation to other synaptotagmin family members in synaptic function and neurological disorders.

What are the key considerations when interpreting contradictory findings about SYT3 function across different experimental models?

When confronting contradictory findings about SYT3 function across different experimental models, researchers should consider several critical factors for proper interpretation:
Model-Specific Variables:

  • Species Differences:

    • Consider evolutionary divergence in SYT3 function between rodent models and human systems.

    • Evaluate whether the human SYT3 protein (as referenced in recombinant studies) may have distinct properties from those in animal models .

  • Cell Type Specificity:

    • Recognize that SYT3 may function differently in various neuron types (excitatory vs. inhibitory, different brain regions).

    • Consider whether contradictions arise from studying different neuronal populations with unique presynaptic properties.

  • Developmental Stage Variations:

    • Assess whether contradictory findings reflect different developmental timepoints, as SYT3's role may change during neural development.

    • Evaluate potential compensatory mechanisms that might mask phenotypes in constitutive knockout models versus acute manipulations.
      Methodological Considerations:

  • Protein Expression Levels:

    • Determine whether overexpression artifacts in some studies might contradict findings from knockout models.

    • Consider the degree of knockdown achieved in shRNA studies versus complete knockout approaches .

  • Experimental Conditions:

    • Examine differences in calcium concentrations, stimulus parameters, and temperature between studies.

    • For in vitro studies, consider variations in buffer composition that might affect protein function, such as the specific formulations used for recombinant SYT3 preparations .

  • Measurement Techniques:

    • Evaluate how different electrophysiological protocols might emphasize different aspects of SYT3 function.

    • Consider resolution limitations of various imaging approaches used to study vesicle dynamics.
      Synthetic Interpretation Approaches:

  • Multi-level Analysis:

    • Integrate findings across molecular, cellular, and systems levels to develop comprehensive models of SYT3 function.

    • Consider how seemingly contradictory findings might reveal context-dependent functions rather than true contradictions.

  • Computational Modeling:

    • Use biophysical modeling approaches to test whether different experimental findings can be reconciled within a single mechanistic framework .

    • Simulate how variations in experimental parameters might lead to apparently conflicting results.

  • Consensus-Building Methodologies:

    • Design experiments specifically to test competing hypotheses arising from contradictory findings.

    • Focus on identifying experimental conditions where contradictions emerge to better understand context-dependent functions.
      By systematically addressing these considerations, researchers can develop nuanced interpretations that reconcile apparently contradictory findings and advance understanding of SYT3's complex roles in neural function.

What emerging technologies will likely advance our understanding of SYT3 function in the next decade?

Several cutting-edge technologies are positioned to transform SYT3 research in the coming decade:
Advanced Imaging Technologies:

  • Super-resolution Nanoscopy: Techniques like STORM, PALM, and STED microscopy will allow visualization of SYT3 within the complex architecture of the presynaptic terminal at nanometer resolution.

  • Live-cell Calcium Imaging: Genetically encoded calcium indicators with improved sensitivity will enable real-time correlation between calcium dynamics and SYT3-mediated vesicle movements.

  • Cryo-electron Tomography: This will provide unprecedented structural detail of SYT3 in its native membrane environment, potentially revealing conformational changes during calcium binding.
    Genetic Engineering Advances:

  • Spatiotemporally Precise Optogenetics: Combining optogenetic control of SYT3 function with high-speed imaging will allow researchers to manipulate and observe protein activity with millisecond precision.

  • Base Editing and Prime Editing: More precise CRISPR technologies will enable introduction of specific SYT3 mutations identified in human disorders without the limitations of traditional knockout approaches .

  • Single-cell Genomics and Proteomics: These approaches will reveal cell-type-specific expression patterns and interaction networks of SYT3 across brain regions and developmental stages.
    Artificial Intelligence Applications:

  • Machine Learning Analysis of Synaptic Function: AI algorithms will detect subtle phenotypes in electrophysiological recordings that might be missed by traditional analysis.

  • Protein Structure Prediction: Tools like AlphaFold will provide increasingly accurate structural models of SYT3 and its interactions with binding partners.

  • Multi-modal Data Integration: AI systems will help synthesize findings across molecular, cellular, and systems levels to build comprehensive models of SYT3 function.
    These technological advances will likely resolve current contradictions in our understanding of SYT3 and provide new insights into its role in both normal brain function and neurological disorders.

What are the most critical unresolved questions about SYT3 function that require further investigation?

Despite significant advances in SYT3 research, several critical questions remain unresolved and warrant focused investigation:
Molecular Mechanism Questions:

  • Precise Calcium Sensing Mechanisms: How exactly does calcium binding to SYT3's C2 domains translate into functional changes in vesicle docking and fusion?

  • SNARE Complex Interactions: What is the detailed molecular choreography of SYT3's interaction with SNARE proteins during different phases of the vesicle cycle ?

  • Regulatory Modifications: How is SYT3 function modulated by post-translational modifications, and what signaling pathways regulate these modifications?
    Physiological Function Questions:

  • Cell-Type Specificity: Does SYT3 play different roles in different types of neurons, and how does this contribute to circuit function?

  • Developmental Roles: Beyond its role in mature synapses, does SYT3 contribute to synapse formation and maturation during development?

  • Homeostatic Mechanisms: How does SYT3 contribute to homeostatic plasticity mechanisms that maintain stable neural network function?
    Disease-Related Questions:

  • Mutation Mechanisms: How do specific SYT3 mutations identified in autism and epilepsy patients alter protein function at the molecular and cellular levels ?

  • Therapeutic Windows: Is there a critical developmental period during which SYT3-targeted therapies would be most effective for neurodevelopmental disorders?

  • Biomarker Potential: Can measurements of SYT3 function or related parameters serve as useful biomarkers for disease progression or treatment response?
    Addressing these questions will require interdisciplinary approaches combining structural biology, electrophysiology, advanced imaging, and clinical research, ultimately advancing both basic neuroscience and therapeutic development for SYT3-related disorders.

How might integration of SYT3 research with broader neuroscience fields enhance our understanding of brain function and dysfunction?

Integration of SYT3 research with broader neuroscience fields promises to yield synergistic insights across multiple levels of brain organization:
Systems Neuroscience Integration:

  • Circuit-Level Function: Understanding how SYT3-mediated short-term plasticity shapes information processing in neural circuits could bridge molecular mechanisms with systems-level computation .

  • Behavioral Correlates: Correlating SYT3 function with specific behavioral processes could reveal how molecular mechanisms of vesicle replenishment translate to cognitive outcomes.

  • Network Stability: SYT3's role in maintaining reliable transmission during sustained activity has implications for understanding network stability and information coding in complex neural systems.
    Translational Neuroscience Connections:

  • Pathophysiological Mechanisms: Integrating SYT3 research with broader studies of autism and epilepsy could reveal common synaptic dysfunction mechanisms across multiple genetic causes .

  • Biomarker Development: Combining SYT3-specific findings with broader neuroimaging and electrophysiological approaches could yield clinically useful biomarkers for synaptic dysfunction.

  • Therapeutic Approaches: Positioning SYT3-targeted interventions within the context of other therapeutic approaches could lead to combination strategies with enhanced efficacy.
    Computational Neuroscience Intersection:

  • Multi-scale Modeling: Incorporating molecular details of SYT3 function into computational models of neural networks could improve predictions of circuit-level dynamics.

  • Predictive Frameworks: Developing computational frameworks that predict how specific SYT3 mutations affect network function could guide personalized treatment approaches.

  • Information Theory Applications: Analyzing how SYT3-dependent vesicle replenishment affects information transfer in neural codes could provide new theoretical insights. By fostering these integrative approaches, SYT3 research can both benefit from and contribute to broader neuroscientific understanding, ultimately advancing our knowledge of fundamental brain mechanisms and therapeutic opportunities for neurological disorders.

Product Science Overview

Discovery and Cloning

Synaptotagmin III was first cloned from a rat brain cDNA library by Mizuta et al. in 1994 . The protein consists of 588 amino acids and shares significant identity with other synaptotagmins, such as synaptotagmin-1 and synaptotagmin-2 . The high degree of conservation in the regulatory domain of protein kinase C among these synaptotagmins suggests a crucial role in cellular signaling .

Expression and Function

Synaptotagmin III mRNA is expressed in the brain, various endocrine tissues, and hormone-secreting clonal cells . It is involved in calcium-dependent exocytosis of secretory vesicles in both endocrine cells and neurons . In vitro studies have shown that recombinant rat SYT3 exhibits calcium-dependent binding to phospholipids and syntaxins, as well as calcium-independent binding to adaptor protein-2 .

Role in Synaptic Plasticity

Recent research has highlighted the role of SYT3 in synaptic plasticity. Awasthi et al. (2019) discovered that SYT3 localizes to postsynaptic endocytic zones and is involved in the removal of AMPA receptors from synaptic plasma membranes in response to stimulation . This process is essential for long-term depression (LTD) and the decay of long-term potentiation (LTP) of synaptic strength . Mice lacking SYT3 exhibited normal learning but showed a lack of forgetting, indicating the importance of SYT3 in memory processes .

Genetic Mapping

The SYT3 gene is located on human chromosome 19q13.33 . In mice, the Syt3 gene is located on chromosome 7, and it is closely linked to the kallikrein gene family . This conserved linkage group includes at least 28 genes, highlighting the evolutionary importance of this region .

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 2025 TheBiotek. All Rights Reserved.