MSGDDDIPEG LEAINLKMNA TTDDSLESTR RMLALCEESK EAGIKTLVML DDQGEQLERCEGALDTINQD MKEAEDHLKG MEKCCGLCVL PWNKTDDFEK TEFAKAWKKD DDGGVISDQPRITVGDSSMG PQGGYITKIT NDAREDEMDE NVQQVSTMVG NLRNMAIDMS TEVSNQNRQL DRIHDKAQSN EVRVESANKR AKNLITK.
SNAP25 (Synaptosomal-associated protein 25kDa) functions as a presynaptic plasma membrane protein essential for synaptic vesicle fusion and neurotransmitter release in C. elegans. This t-SNARE protein forms part of the core SNARE complex alongside synaptobrevin (v-SNARE) and syntaxin (t-SNARE), which is crucial for docking and holding synaptic vesicles at the presynaptic membrane. The assembled complex consists of a bundle of four helices, with SNAP25 contributing two helices, syntaxin providing one, and synaptobrevin supplying one. This molecular arrangement enables efficient membrane fusion, directly mediating the release of neurotransmitters into the synaptic cleft .
The C. elegans SNAP25 protein is a single, non-glycosylated polypeptide chain containing 207 amino acids with a molecular mass of approximately 23 kDa when expressed in its full form. This differs somewhat from the recombinant version produced in E. coli for research purposes, which can appear as 13.3 kDa (127 aa) as confirmed by MALDI-TOF analysis. It's worth noting that the molecular weight can appear higher when analyzed by SDS-PAGE due to the protein's structural properties .
The amino acid sequence of C. elegans SNAP25 includes several functional domains that are conserved across species, although there are specific structural differences compared to mammalian versions. The full sequence includes: MSGDDDIPEG LEAINLKMNA TTDDSLESTR RMLALCEESK EAGIKTLVML DDQGEQLERC EGALDTINQD MKEAEDHLKG MEKCCGLCVL PWNKTDDFEK TEFAKAWKKD DDGGVISDQP RITVGDSSMG PQGGYITKIT NDAREDEMDE NVQQVSTMVG NLRNMAIDMS TEVSNQNRQL DRIHDKAQSN EVRVESANKR AKNLITK .
When conducting literature searches on SNAP25 in C. elegans, researchers should be aware of the multiple synonyms used to refer to this protein. These include: Super-Protein, SUP, RIC4, SEC9, SNAP, RIC-4, SNAP25, SNAP-25, Synaptosomal-associated protein 25, Synaptosomal-associated 25 kDa protein, FLJ23079, bA416N4.2, dJ1068F16.2, and resistance to inhibitors of cholinesterase 4 homolog. The diversity of nomenclature reflects the protein's identification through different experimental approaches and genetic screens across various research groups .
The most established method for recombinant expression of C. elegans SNAP25 involves amplification of the gene by PCR from C. elegans genomic material or cDNA, followed by cloning into an E. coli expression vector with an appropriate tag (commonly His-tag). For purification, conventional chromatography techniques are employed to achieve high purity (>95% as determined by SDS-PAGE). The typical yield produces recombinant protein at a concentration of approximately 1 mg/ml, which can be verified by BCA assay .
For optimal storage and stability, the purified protein should be maintained in an appropriate buffer such as 20 mM Tris-HCl (pH 7.5) containing 0.1 M NaCl, or alternatively in 20mM Tris-HCl pH7.5 with 50mM NaCl, 5mM DTT, 1mM EDTA, and 10% Glycerol. Short-term storage at +4°C is suitable for 1-2 weeks, while for long-term preservation, the protein should be aliquoted and stored at -20°C to avoid repeated freeze-thaw cycles which can compromise protein integrity .
When designing experiments to assess SNAP25 function in C. elegans neurotransmission, consider implementing electrophysiological recordings similar to those used in knockout studies of related proteins. These approaches can distinguish between calcium-dependent and calcium-independent neurotransmitter release mechanisms. Based on comparative studies in other model systems, measurement protocols should include:
Recording of spontaneous release events (calcium-independent)
Evoked release through field stimulation (calcium-dependent)
Hypertonic solution-induced release (to probe the readily releasable pool)
Endocytosis measurements using FM-dye uptake and release
This multimodal approach allows researchers to differentiate between various aspects of vesicle trafficking and fusion. Previous research in related systems has demonstrated that SNAP25 deficiency significantly impacts calcium-dependent evoked release while leaving approximately 10-12% of calcium-independent release intact. Furthermore, SNAP25-deficient synapses, unlike synaptobrevin-2 knockout models, show virtual insensitivity to calcium-dependent stimulation and lack facilitation during high-frequency stimulation .
When performing immunohistochemical localization of SNAP25 in C. elegans neurons, researchers should consider several technical factors to obtain reliable results. First, tissue fixation protocols must preserve the presynaptic plasma membrane structure where SNAP25 is primarily localized. A combination of paraformaldehyde fixation followed by permeabilization with appropriate detergents (0.1-0.5% Triton X-100) typically yields good results.
For antibody selection, consider using antibodies raised against recombinant C. elegans SNAP25 rather than antibodies developed for mammalian systems, as epitope recognition may differ. When validating antibody specificity, include appropriate controls such as SNAP25 mutant or knockout strains if available. Co-localization studies with other presynaptic markers such as syntaxin can help confirm the specificity of SNAP25 labeling.
The detection system should be optimized for the C. elegans nervous system's small size, potentially employing confocal or super-resolution microscopy to accurately resolve the presynaptic localization pattern. Finally, quantification of immunofluorescence should follow standardized protocols to ensure reproducibility across experiments and allow for comparison between different neuronal populations or experimental conditions .
SNAP25 in C. elegans appears to play a particularly critical role in calcium-secretion coupling compared to other SNARE proteins. Comparative studies between SNAP25 and synaptobrevin-2 knockouts in related systems have revealed distinct functional differences. While both proteins are components of the SNARE complex essential for vesicle fusion, SNAP25 deficiency causes a more severe phenotype regarding calcium-dependent release mechanisms.
Furthermore, unlike synaptobrevin-2-deficient synapses, SNAP25-deficient synapses do not exhibit facilitation of release during high-frequency stimulation. This evidence suggests that SNAP25 plays a more significant and specific role in coupling calcium influx to the exocytotic machinery, potentially through direct or indirect interactions with calcium sensors or regulatory proteins involved in the fusion process .
To investigate neuron cell type-specific SNAP25 expression patterns in C. elegans, researchers can employ several complementary approaches:
Promoter Analysis: Identify the regulatory sequences controlling SNAP25 expression by isolating the promoter region of the C. elegans SNAP25 gene. This approach can reveal neuron-specific regulatory elements through deletion and mutagenesis analyses .
Reporter Gene Constructs: Generate transgenic C. elegans lines expressing fluorescent proteins (GFP, mCherry) under the control of the SNAP25 promoter or as translational fusions with the SNAP25 protein. This allows visualization of expression patterns in live animals and precise cellular localization.
Single-Cell RNA Sequencing: Apply scRNA-seq techniques to identify differential expression of SNAP25 across different neuronal populations, providing a comprehensive map of expression levels in various cell types.
Immunohistochemistry: Use antibodies against SNAP25 in conjunction with known neuronal subtype markers to characterize expression patterns at the protein level across different stages of development.
Conditional Expression Systems: Employ tissue-specific or temporally controlled promoters to express or silence SNAP25 in specific neuronal subtypes, allowing for the assessment of functional consequences in defined neural circuits.
These approaches collectively can reveal whether SNAP25 expression is uniform across all neurons or shows preferential expression in specific subtypes, potentially correlating with particular synaptic properties or neurotransmitter systems .
Mutations in SNAP25 significantly impact synaptic vesicle dynamics and neurotransmitter release in C. elegans through several mechanisms. Based on comparative studies, we can infer that SNAP25 mutations would affect:
Calcium-Dependent Exocytosis: SNAP25 mutations likely severely compromise calcium-evoked neurotransmitter release, as the protein appears critical for coupling calcium influx to vesicle fusion. Unlike mutations in other SNARE proteins such as synaptobrevin, SNAP25 deficiency results in synapses that are virtually unresponsive to calcium-dependent stimulation .
Readily Releasable Pool (RRP) Utilization: While approximately 10-12% of the RRP might remain accessible for release through calcium-independent mechanisms (such as hypertonic sucrose stimulation), the efficiency of this release would be significantly reduced compared to wild-type synapses .
Short-Term Plasticity: SNAP25-deficient synapses would likely lack the facilitation normally observed during high-frequency stimulation, suggesting a role for SNAP25 in activity-dependent modulation of release probability .
Endocytosis: Interestingly, SNAP25 mutations would reduce but not eliminate endocytosis during evoked stimulation. Unlike synaptobrevin, SNAP25 does not appear to directly function in endocytosis, as synaptic vesicle turnover probed by FM-dye uptake and release during hypertonic stimulation remains relatively unaffected by SNAP25 absence .
These effects collectively suggest that SNAP25 mutations would have profound consequences for synaptic transmission, particularly affecting evoked release while sparing some forms of spontaneous and calcium-independent release mechanisms.
SNAP25 function shows remarkable conservation between C. elegans and other model organisms, though with some notable species-specific adaptations. The core role of SNAP25 as a t-SNARE component essential for synaptic vesicle fusion is preserved across evolution, from nematodes to mammals. In all systems studied, SNAP25 contributes two helices to the four-helix bundle that forms the SNARE complex, working alongside syntaxin and synaptobrevin to drive membrane fusion .
Post-translational modifications also show evolutionary divergence. In mammals, SNAP25 undergoes palmitoylation at cysteine residues, which is critical for membrane association. While this modification appears conserved in C. elegans SNAP25, the specific pattern and regulation of palmitoylation may differ, potentially influencing the protein's membrane targeting and function in nematode neurons .
Research on SNAP25 in C. elegans offers valuable insights for understanding human neurological disorders through several mechanisms:
Synaptic Dysfunction Models: C. elegans SNAP25 studies provide simplified models to understand fundamental aspects of synaptic transmission defects that underlie many neurological disorders. The well-defined nervous system of C. elegans allows researchers to observe how SNAP25 dysfunction affects specific neural circuits and behaviors.
Genetic Interaction Networks: The genetic tractability of C. elegans facilitates comprehensive screens for genetic modifiers of SNAP25 function, potentially revealing novel interacting partners relevant to human disease. These studies can identify suppressor or enhancer mutations that may represent therapeutic targets for SNAP25-associated disorders.
Drug Screening Platforms: C. elegans SNAP25 mutants can serve as platforms for high-throughput screening of compounds that restore synaptic function, potentially identifying leads for therapeutic development in human conditions where SNAP25 function is compromised.
Neurodevelopmental Insights: By studying SNAP25 function throughout C. elegans development, researchers can better understand how synaptic proteins contribute to circuit formation and maturation, processes often disrupted in neurodevelopmental disorders.
Human SNAP25 variants have been implicated in conditions including attention deficit hyperactivity disorder (ADHD), schizophrenia, and certain epilepsy syndromes. The simplified genetic background and experimental accessibility of C. elegans make it an excellent model to characterize the functional consequences of these variants in a controlled system .
Translating findings from C. elegans SNAP25 studies to mammalian systems presents several methodological challenges that researchers must address:
Structural and Functional Divergence: Despite conservation of core functions, C. elegans SNAP25 differs from mammalian homologs in certain structural features and regulatory mechanisms. Researchers must carefully validate whether specific findings about protein interactions, post-translational modifications, or regulatory pathways are conserved across species.
Neural Circuit Complexity: The C. elegans nervous system contains only 302 neurons with approximately 7,000 synapses, compared to billions of neurons and trillions of synapses in mammalian brains. This difference in complexity means that network-level effects of SNAP25 manipulation may manifest differently between systems.
Isoform Diversity: Mammals express multiple SNAP25 isoforms (SNAP25a and SNAP25b) with developmental and regional specificity, as well as related proteins like SNAP23. C. elegans has fewer isoforms, potentially limiting the direct applicability of findings to the more complex mammalian expression patterns.
Experimental Techniques: Methods optimized for C. elegans, such as whole-animal genetics and optical imaging in transparent animals, may require significant adaptation for mammalian systems. Conversely, techniques readily applied in mammalian neurons, such as electrophysiological recordings with high temporal resolution, can be more challenging in the smaller C. elegans neurons.
Pharmacological Differences: Drug effects observed in C. elegans may differ in mammals due to variations in protein structure, metabolism, or compensatory mechanisms, necessitating careful pharmacological validation across model systems.
Addressing these challenges requires complementary approaches across multiple model systems, with findings in C. elegans serving as a foundation for targeted investigations in more complex mammalian preparations .
To effectively study SNAP25 interactions with other SNARE proteins in C. elegans, researchers can employ several complementary approaches:
In vitro Binding Assays: Using recombinant purified C. elegans SNAP25 (as described in product information), perform pull-down assays, surface plasmon resonance, or isothermal titration calorimetry to quantitatively characterize binding affinities and kinetics with syntaxin and synaptobrevin. These assays can be conducted under varying conditions to assess factors influencing complex formation .
Co-immunoprecipitation: Develop antibodies against C. elegans SNAP25 or use epitope-tagged versions for immunoprecipitation from worm lysates, followed by western blotting or mass spectrometry to identify interacting partners. This approach captures physiologically relevant interactions in their native context.
Yeast Two-Hybrid Screening: Employ SNAP25 as bait to screen for interacting proteins from C. elegans cDNA libraries, potentially identifying novel binding partners beyond the canonical SNARE proteins.
FRET/FLIM Imaging: Generate transgenic worms expressing fluorescently-tagged SNAP25 and other SNARE proteins to visualize interactions in vivo using Förster Resonance Energy Transfer (FRET) or Fluorescence Lifetime Imaging Microscopy (FLIM).
Proximity Labeling: Apply BioID or APEX2 proximity labeling techniques by fusing these enzymes to SNAP25, allowing identification of proteins in close proximity under physiological conditions.
Genetic Interaction Studies: Perform systematic genetic interaction analyses using combinations of mutations or RNAi against SNAP25 and other synaptic proteins to identify functional relationships that may reflect physical interactions.
By combining these approaches, researchers can build a comprehensive understanding of SNAP25's interaction network in C. elegans, including binding dynamics, regulatory mechanisms, and context-dependent changes in complex formation .
When designing and interpreting SNAP25 knockout or knockdown experiments in C. elegans, researchers should consider several critical factors:
Experimental Design Considerations:
Targeting Strategy: For complete knockout, CRISPR-Cas9 gene editing offers precise deletion of the entire SNAP25 coding sequence. For conditional or partial knockdown, RNAi or tissue-specific expression of dominant-negative constructs may be more appropriate. Each approach has different specificity and efficiency profiles.
Control Selections: Include multiple control groups: wild-type animals, heterozygous mutants, and animals with mutations in related genes (e.g., other SNARE components) to distinguish SNAP25-specific effects from general synaptic dysfunction.
Developmental Timing: Since complete SNAP25 knockout might cause developmental lethality or severe defects, consider temporally-controlled disruption using heat-shock promoters or drug-inducible systems to bypass early developmental requirements.
Compensation Assessment: Measure expression levels of other SNARE family members to detect potential compensatory upregulation in response to SNAP25 reduction.
Phenotypic Analysis Framework:
Electrophysiological Measurements: Based on findings from related models, assess both spontaneous and evoked neurotransmission, distinguishing between calcium-dependent and calcium-independent release mechanisms. Expect severe impairment of evoked release with partial preservation of spontaneous release .
Behavioral Assays: Quantify locomotion, pharyngeal pumping, and more complex behaviors that depend on synaptic transmission. Correlate behavioral defects with electrophysiological findings to establish structure-function relationships.
Vesicle Trafficking Visualization: Employ fluorescently-tagged synaptic vesicle proteins and FM dyes to monitor vesicle docking, fusion, and recycling in live animals.
Ultrastructural Analysis: Use electron microscopy to examine synaptic morphology, vesicle distribution, and active zone architecture in SNAP25-deficient synapses.
Interpretation Challenges:
Distinguish Direct vs. Indirect Effects: Severe synaptic dysfunction may cause secondary cellular stress responses or developmental abnormalities; careful temporal analysis can help separate primary from secondary effects.
Consider Non-Synaptic SNAP25 Functions: While primarily studied at synapses, SNAP25 may have additional roles in membrane trafficking or other cellular processes that could contribute to observed phenotypes.
Reconcile Partial vs. Complete Loss: Different levels of SNAP25 reduction may produce qualitatively different phenotypes rather than simply quantitatively different outcomes, complicating dose-response interpretations .
For optimal storage and handling of recombinant C. elegans SNAP25 protein, researchers should follow these evidence-based protocols to maintain protein integrity and functionality:
Storage Conditions:
Storage Duration | Recommended Temperature | Buffer Composition | Additional Stabilizers |
---|---|---|---|
Short-term (1-2 weeks) | +4°C | 20 mM Tris-HCl (pH 7.5), 0.1 M NaCl | None required |
Long-term (months) | -20°C or -70°C | 20 mM Tris-HCl (pH 7.5), 50 mM NaCl, 5 mM DTT, 1 mM EDTA, 10% Glycerol | 0.1% HSA or BSA recommended as carrier protein |
Handling Recommendations:
Aliquoting: Divide the purified protein into small single-use aliquots before freezing to avoid repeated freeze-thaw cycles, which can lead to protein denaturation and loss of activity.
Thawing Protocol: Thaw frozen aliquots rapidly by placing at room temperature for minimal time, then immediately transfer to ice. Avoid vortexing, which can cause protein denaturation, and instead mix gently by pipetting or inversion.
Working Concentration: Recombinant SNAP25 is typically supplied at 1 mg/ml concentration (as determined by BCA assay). For most experimental applications, dilution to working concentrations of 10-100 μg/ml in appropriate assay buffers is recommended.
Avoiding Contamination: Use sterile technique when handling the protein solution, as bacterial contamination can lead to protein degradation through protease activity. Consider adding protease inhibitors for extended work sessions.
Buffer Compatibility: When using SNAP25 in experimental assays, verify compatibility with assay buffers. Significant changes in pH, ionic strength, or the addition of detergents may affect protein structure and function.
Quality Control: Before using in critical experiments, verify protein integrity by SDS-PAGE (expected purity >95%) and functionality through appropriate binding assays with known SNARE partners.
By following these guidelines, researchers can maximize the stability and experimental utility of recombinant C. elegans SNAP25 protein, ensuring reproducible results in structural, biochemical, and functional studies .
Synaptosomal-associated protein 25kDa (SNAP-25) is a crucial component of the SNARE (Soluble NSF Attachment Protein Receptor) complex, which plays a vital role in the exocytotic release of neurotransmitters during synaptic transmission. This protein is highly conserved across species, including the model organism Caenorhabditis elegans (C. elegans). The recombinant form of SNAP-25 from C. elegans has been extensively studied to understand its structure, function, and potential applications in neuroscience research.
SNAP-25 is a presynaptic protein that is essential for the proper functioning of synaptic vesicle exocytosis. It forms a coiled-coil assembly with syntaxin-1 and synaptobrevin, facilitating the apposition of synaptic vesicles to the presynaptic membrane and permitting their calcium-triggered fusion . This process is critical for the release of neurotransmitters into the synaptic cleft, enabling neuronal communication.
The protein exists in two isoforms, SNAP-25a and SNAP-25b, which are differentially expressed during development. SNAP-25a is predominantly expressed during the embryonic stage, while SNAP-25b becomes the major isoform during postnatal life .
Recombinant SNAP-25 from C. elegans is produced in Escherichia coli (E. coli) as a single, non-glycosylated polypeptide chain containing 207 amino acids and having a molecular mass of approximately 23kDa . The gene encoding SNAP-25 is amplified by PCR from C. elegans and cloned into an E. coli expression vector. The protein is then purified using conventional chromatography techniques .
The recombinant form of SNAP-25 from C. elegans is widely used in laboratory procedures to study the protein’s role in synaptic transmission and its interactions with other cellular components. It is particularly valuable in research focused on understanding the molecular mechanisms underlying synaptic vesicle exocytosis and the regulation of neurotransmitter release.
Alterations in SNAP-25 have been associated with various neurological and psychiatric disorders, including Attention Deficit Hyperactivity Disorder (ADHD), schizophrenia, and bipolar disorder . Understanding the function and regulation of SNAP-25 can provide insights into the pathophysiology of these conditions and potentially lead to the development of targeted therapies.