VAMP2 antibodies are immunoreagents designed to detect and quantify VAMP2, a 13 kDa synaptic vesicle membrane protein encoded by the VAMP2 gene. These antibodies enable researchers to study VAMP2's role in:
Neurotransmitter release: VAMP2 forms a stable complex with syntaxin-1 and SNAP-25 to drive synaptic vesicle fusion .
Membrane trafficking: Critical for insulin-induced GLUT4 translocation in adipocytes .
Disease mechanisms: Linked to neurodevelopmental disorders, epilepsy, and neurodegenerative conditions .
VAMP2 antibodies are available as monoclonal or polyclonal variants, validated for:
Western blot (WB): Detects VAMP2 in brain lysates (e.g., 13 kDa band in human cerebellum) .
Immunofluorescence (IF): Localizes VAMP2 to synaptic vesicles in neuronal cells .
Flow cytometry: Quantifies intracellular VAMP2 expression in cell lines like SH-SY5Y .
Immunoprecipitation (IP): Isolates VAMP2 complexes with syntaxin-4/SNAP23 .
Neurotransmitter Release: Mice lacking functional VAMP2 exhibit <10% synaptic transmission efficiency, highlighting its necessity in exocytosis .
Phase Separation: VAMP2 promotes α-synuclein condensate formation, implicating it in Parkinson’s disease pathology .
Insulin Signaling: VAMP2 is required for GLUT4 vesicle incorporation into adipocyte membranes .
Neurodevelopmental Disorders: Heterozygous VAMP2 mutations cause hypotonia, autism, and hyperkinetic movements .
Alzheimer’s Disease: Reduced VAMP2 levels correlate with impaired synaptic function .
Epilepsy: VAMP2 knockdown in mice suppresses glutamate release, conferring resistance to seizures .
Supplier | Clone/Product Code | Type | Applications | Species Reactivity | Citations |
---|---|---|---|---|---|
Abcam | EPR12790 (ab181869) | Monoclonal | WB, IF, Flow Cyt | Human, Mouse, Rat | 15+ |
Thermo Fisher | PA1-766 | Polyclonal | WB, IF, IP | Rat, Mouse | N/A |
R&D Systems | 541405 (MAB5136) | Monoclonal | WB, ELISA | Human, Mouse | N/A |
Sample Preparation: Use RIPA buffer for brain tissue lysates; methanol fixation preserves epitopes in IF .
Controls: Include siRNA-treated cells or VAMP2 knockout models to confirm specificity .
Artifact Mitigation: Avoid repeated freeze-thaw cycles; store antibodies at -20°C .
VAMP2, also known as synaptobrevin-2, is a critical vesicle-associated membrane protein involved in membrane fusion events. This 116-amino acid protein (12.7 kDa in humans) is localized to cytoplasmic vesicles and cell membranes, functioning as a core component of the SNARE complex that mediates vesicle fusion . VAMP2 plays essential roles in insulin signaling pathways and exocytosis, making it a significant target for neuroscience, diabetes, and vesicular trafficking research . In Alzheimer's disease, levels of all VAMP isoforms appear significantly lowered, highlighting its importance in neurological research . VAMP2 is also implicated in insulin-sensitive glucose transporter type 4 (GLUT4) regulation, making it relevant for metabolic research .
VAMP2 antibodies specifically target the vesicle-associated membrane protein 2, distinguishing it from antibodies against other SNARE proteins like SNAP-25 and syntaxin. While all SNARE protein antibodies are used to study vesicular fusion, VAMP2 antibodies specifically recognize the v-SNARE component located on vesicle membranes rather than t-SNAREs on target membranes . VAMP2 antibodies typically recognize epitopes in the N-terminal region (such as residues 1-18 in the PA1-766 antibody), contrasting with syntaxin antibodies that often target the H3 domain or SNAP-25 antibodies targeting the N or C terminal regions . This specificity allows researchers to distinguish between different SNARE-mediated fusion events in complex cellular systems.
VAMP2 antibodies are versatile research tools employed across multiple experimental platforms. The most common applications include Western blotting for protein detection and quantification, immunohistochemistry for tissue localization, and immunofluorescence for subcellular distribution studies . They are also valuable in immunoprecipitation experiments to study protein-protein interactions involving VAMP2 . Additionally, these antibodies can be used in functional studies examining exocytosis mechanisms, as seen in research on cAMP-stimulated renin release . Flow cytometry applications are also reported, particularly with fluorochrome-conjugated versions . In knockout or knockdown validation studies, these antibodies serve as essential tools to confirm protein reduction or elimination .
When selecting a VAMP2 antibody, consider multiple critical factors to ensure experimental success. First, match the antibody's species reactivity to your experimental model—available antibodies show reactivity to human, mouse, rat, pig, and bovine VAMP2 with varying affinities . For detecting denatured proteins in Western blots, polyclonal antibodies like DF6381 may offer better sensitivity, while monoclonal antibodies provide higher specificity for immunolocalization studies .
Consider the antibody's epitope location: N-terminal-directed antibodies (like PA1-766 targeting residues 1-18) may not detect VAMP2 fragments after proteolytic cleavage by tetanus toxin at position ~76 . For colocalization studies, select antibodies raised in different host species to avoid cross-reactivity when using multiple primary antibodies. Review published validation data, including Western blot bands (VAMP2 typically appears at 12-19 kDa), immunofluorescence patterns, and knockout controls to ensure antibody specificity .
For optimal Western blot detection of VAMP2, follow these methodological considerations. Begin with proper sample preparation: tissues rich in VAMP2 (brain, nervous system, skeletal muscle) should be lysed in buffers containing appropriate protease inhibitors to prevent degradation of this small protein . Use 12-15% polyacrylamide gels to properly resolve this low molecular weight protein (12.7-19 kDa range) .
Transfer conditions should be optimized for small proteins—typically using lower methanol concentrations (10%) and shorter transfer times to prevent the protein from passing through the membrane. When blocking, use 3-5% BSA rather than milk for phospho-specific VAMP2 detection. Primary antibody dilutions typically range from 1:500 to 1:2000, but always follow manufacturer recommendations for specific antibodies .
For visualization, both chemiluminescence and fluorescent secondary antibodies are suitable, though the latter offers better quantification precision. Always include appropriate controls: positive controls (brain tissue extracts), negative controls (tissues with low VAMP2 expression), and loading controls (preferably similar-sized housekeeping proteins) . For challenging samples, consider enriching vesicular fractions before Western blotting to increase detection sensitivity.
Robust controls are critical for reliable immunohistochemistry (IHC) or immunofluorescence (IF) experiments with VAMP2 antibodies. Always include a positive tissue control known to express VAMP2 (brain tissue sections work well) to verify staining protocols . Negative controls should include both primary antibody omission and ideally tissues from VAMP2 knockout models to confirm specificity .
For absorbed controls, pre-incubate the antibody with its immunizing peptide (such as the PEP-101 peptide for PA1-766) to demonstrate binding specificity . When conducting double immunolabeling, include single-antibody controls to ensure no cross-reactivity or bleed-through between fluorescence channels. For subcellular localization studies, co-stain with established markers of subcellular compartments (synaptic vesicle proteins, membrane markers) to confirm the expected VAMP2 distribution pattern .
When studying tissues where VAMP2 may be cleaved by endogenous or experimental proteases, consider using antibodies targeting different epitopes to distinguish intact versus cleaved forms . For quantitative immunofluorescence, include calibration standards and ensure identical acquisition parameters across all experimental samples.
For investigating SNARE-mediated vesicle fusion dynamics, VAMP2 antibodies can be employed in several sophisticated experimental approaches. Live-cell imaging combined with VAMP2 antibody fragments or fluorescent protein-tagged VAMP2 constructs allows visualization of vesicle trafficking and fusion events in real-time . To study the kinetics of SNARE complex formation, use proximity ligation assays with VAMP2 antibodies paired with antibodies against other SNARE proteins.
For functional studies, combine VAMP2 immunodetection with exocytosis assays such as FM1-43 fluorescence monitoring, which measures membrane surface area changes during fusion events . Research by Mendez and colleagues demonstrated that tetanus toxin cleavage of VAMP2 reduced FM1-43 fluorescence increase by approximately 62% following forskolin/IBMX stimulation, indicating substantial blockade of exocytosis .
Super-resolution microscopy (STORM, PALM) with VAMP2 antibodies provides nanoscale visualization of vesicle docking and fusion sites. For temporal studies of VAMP2 dynamics, consider pulse-chase experiments with antibodies targeting different VAMP2 epitopes or post-translational modifications. Correlative light and electron microscopy approaches combining VAMP2 immunolabeling with ultrastructural analysis can provide comprehensive insights into fusion dynamics across different cellular compartments.
Validating VAMP2 knockdown or knockout requires rigorous antibody-based confirmation methods. Western blot analysis using validated VAMP2 antibodies is the primary validation tool, with successful knockdown typically demonstrated by a 50-70% reduction in band intensity, as seen in studies using shRNA-mediated silencing . Quantitative immunofluorescence microscopy serves as a complementary approach, allowing visualization of reduced VAMP2 signal in specific cellular compartments.
For more complex experimental systems, consider multiplex immunodetection to simultaneously assess VAMP2 reduction alongside changes in other SNARE proteins that might show compensatory regulation . When validating CRISPR/Cas9 knockout models, use antibodies targeting different VAMP2 epitopes to ensure complete protein elimination rather than truncation or mutation that might still be recognized by some antibodies.
Importantly, functional validation must accompany immunodetection. Research demonstrates that a 54% reduction in VAMP2 protein via shRNA resulted in a 67% impairment of cAMP-stimulated renin release, providing functional validation of the knockdown . For controls, include scrambled shRNA treatment groups, which typically show normal VAMP2 levels (as demonstrated by comparable band intensity on Western blots) and preserved functional responses such as cAMP-stimulated exocytosis .
Unexpected molecular weight bands in VAMP2 antibody detection require careful interpretation based on several biological and technical considerations. While canonical VAMP2 typically appears at 12.7-19 kDa, higher molecular weight bands may represent:
Post-translational modifications such as ubiquitination (at K52, K59) or phosphorylation (at S61, S75, T79)
SNARE complexes resistant to SDS denaturation, appearing at approximately 70-80 kDa
Dimerization or oligomerization of VAMP2 molecules
Cross-reactivity with other VAMP family members (VAMP1, VAMP3) due to sequence homology
Lower molecular weight bands often indicate:
Proteolytic cleavage products, particularly following tetanus toxin treatment which cleaves at position ~76
Alternatively spliced isoforms
Degradation during sample preparation
To properly interpret these patterns, run appropriate controls including: recombinant VAMP2 protein standards, competitive blocking with immunizing peptides, and samples from VAMP2 knockout models . Different antibodies targeting distinct epitopes may produce different banding patterns—N-terminal antibodies won't detect C-terminal fragments after proteolytic cleavage by tetanus toxin . Always consider sample preparation conditions, as membrane proteins like VAMP2 can aggregate during boiling, producing higher molecular weight species.
VAMP2 antibodies have become instrumental in elucidating the molecular pathology of various neurological disorders. In Alzheimer's disease research, these antibodies have revealed significantly lowered levels of all VAMP isoforms, suggesting compromised synaptic vesicle trafficking contributes to cognitive decline . Immunohistochemical studies using VAMP2 antibodies allow researchers to visualize alterations in synaptic density and distribution patterns in brain tissue samples from patients and animal models.
For neurodegenerative disease research, quantitative Western blotting with VAMP2 antibodies enables measurement of synaptic loss progression, while immunoprecipitation studies help identify abnormal protein interactions or post-translational modifications affecting VAMP2 function . In epilepsy models, these antibodies help examine how hyperexcitation affects SNARE protein distribution and function at synapses.
VAMP2 antibodies also support research into synaptic dysfunction in autism spectrum disorders and schizophrenia by providing tools to analyze changes in vesicular release machinery. Importantly, comparing results across multiple models and methods strengthens confidence in findings. For example, reduced VAMP2 immunoreactivity should be correlated with functional electrophysiological changes in neurotransmitter release to establish causative relationships between molecular changes and disease phenotypes.
VAMP2 antibodies provide powerful tools for investigating insulin-regulated glucose transport mechanisms. Research has established VAMP2 as a resident protein of insulin-sensitive glucose transporter type 4 (GLUT4) compartments, where it facilitates GLUT4 vesicle incorporation into the cell surface in response to insulin stimulation . Immunofluorescence studies using VAMP2 antibodies allow visualization of GLUT4-containing vesicle trafficking in adipocytes and muscle cells following insulin treatment.
To study temporal dynamics, researchers can perform time-course experiments with VAMP2/GLUT4 co-localization studies following insulin stimulation. Immuno-electron microscopy with VAMP2 antibodies provides ultrastructural details of vesicle docking and fusion events at the plasma membrane. For functional analyses, combine VAMP2 immunodetection with glucose uptake assays to correlate protein localization with transport activity.
VAMP2 knockdown studies validated by antibody detection have demonstrated reduced insulin-stimulated glucose uptake, confirming its functional role . In disease models such as insulin resistance or type 2 diabetes, quantitative immunoblotting with VAMP2 antibodies can reveal deficiencies in SNARE protein expression or regulation. When designing such studies, consider that multiple VAMP isoforms may play compensatory roles, necessitating careful experimental controls and comprehensive immunodetection of multiple SNARE proteins to fully characterize the system.
VAMP2 antibodies have provided critical insights into the molecular mechanisms of renin release and hypertension pathophysiology. Research using VAMP2 antibodies has demonstrated that this protein mediates cAMP-stimulated renin release from juxtaglomerular (JG) cells, a process fundamental to blood pressure regulation . Immunofluorescence studies have helped localize VAMP2 to renin-containing granules, establishing its role in the exocytotic machinery of JG cells.
Functional studies combining immunodetection with physiological measurements have revealed that VAMP2 knockdown (verified by Western blotting showing ~54% protein reduction) impairs cAMP-stimulated renin release by approximately 67%, demonstrating its physiological significance . Importantly, this effect appears specific to VAMP2, as similar knockdown of VAMP3 (~48% reduction) did not affect renin release, highlighting the value of isoform-specific antibodies in distinguishing functional roles .
For investigating the regulatory mechanisms, researchers have employed VAMP2 antibodies in combination with tetanus toxin treatment, which cleaves VAMP2 at position ~76, releasing the N-terminal portion to the cytosol for degradation . The subsequent reduction in forskolin-stimulated FM1-43 fluorescence (a marker of exocytosis) by 62% demonstrates that VAMP2 is essential for exocytotic renin release . These methodological approaches exemplify how VAMP2 antibodies contribute to mechanistic understanding of renin secretion and potential therapeutic targets for hypertension management.
Studying post-translational modifications (PTMs) of VAMP2 requires specialized antibody approaches and careful experimental design. VAMP2 undergoes several PTMs including ubiquitination (at K52, K59), phosphorylation (at S61, S75, T80), and methylation (at R66) . For phosphorylation studies, use phospho-specific antibodies that recognize VAMP2 only when modified at specific residues. These studies should include appropriate controls: lambda phosphatase treatment to remove phosphorylation, positive controls using pharmacological agents that enhance specific phosphorylation events, and parallel detection with pan-VAMP2 antibodies to normalize for total protein levels.
For ubiquitination studies, combine VAMP2 immunoprecipitation with ubiquitin-specific antibody detection, or vice versa. Consider using proteasome inhibitors to enhance detection of ubiquitinated species. Methylation studies may require specialized antibodies against methylated VAMP2 epitopes or techniques combining immunoprecipitation with mass spectrometry.
To correlate PTMs with functional changes, design experiments combining PTM-specific antibody detection with functional assays such as FM1-43 measurement of exocytosis . Advanced approaches include using site-directed mutagenesis to generate PTM-mimetic or PTM-deficient VAMP2 variants, followed by antibody-based validation and functional testing. For temporal dynamics of PTMs, consider pulse-chase experiments with sequential immunoprecipitation steps to track modification patterns over time or following specific stimuli.
Emerging technologies are significantly expanding the capabilities of VAMP2 antibody applications in cutting-edge research. Single-molecule imaging techniques using fluorescently-labeled VAMP2 antibody fragments allow tracking of individual vesicle fusion events with unprecedented temporal and spatial resolution. Expansion microscopy combined with VAMP2 immunolabeling physically enlarges specimens, revealing nanoscale details of SNARE protein organization beyond conventional microscopy limits.
CRISPR-based knock-in strategies now enable endogenous tagging of VAMP2, allowing antibody detection of physiologically relevant protein levels and localizations without overexpression artifacts. Multiplexed antibody-based imaging technologies, such as cyclic immunofluorescence or mass cytometry, permit simultaneous visualization of VAMP2 alongside dozens of other proteins within the same sample.
Microfluidic systems combined with VAMP2 antibody detection enable real-time monitoring of vesicle fusion events in response to precisely controlled stimuli. For clinical applications, ultrasensitive detection methods like single-molecule arrays (Simoa) with VAMP2 antibodies may allow quantification of trace amounts of VAMP2 in biological fluids as potential biomarkers. Intrabodies—antibody fragments expressed within living cells—are being developed to track VAMP2 dynamics in real-time without fixation artifacts. These technological advances collectively provide researchers with unprecedented tools to study VAMP2 biology with greater precision, sensitivity, and physiological relevance.
When faced with contradictory results from different VAMP2 antibodies, researchers should implement a systematic analytical approach. First, examine the epitope specificity—antibodies recognizing different domains of VAMP2 may yield varying results, particularly if post-translational modifications or proteolytic cleavage affect certain epitopes . For instance, N-terminal-directed antibodies won't detect C-terminal fragments after tetanus toxin cleavage at position ~76 .
Cross-reactivity with other VAMP isoforms should be carefully evaluated, especially between closely related VAMP2 and VAMP3, which share high sequence homology. Batch-to-batch variation in antibody production can significantly impact results, necessitating lot number tracking and consistent use of the same lot for comparative studies.
Consider the application context—some antibodies perform well in Western blotting but poorly in immunohistochemistry due to epitope accessibility differences in fixed versus denatured samples . Fixation and sample preparation methods dramatically affect epitope preservation and antibody recognition; for example, certain fixatives may mask VAMP2 epitopes or create artifacts.
To resolve discrepancies, implement verification strategies including:
Using multiple antibodies targeting different VAMP2 epitopes
Validating with genetic approaches (siRNA knockdown, CRISPR knockout)
Employing complementary techniques like mass spectrometry
Testing antibodies on recombinant VAMP2 protein standards
Including appropriate positive and negative controls with each experiment
When publishing results, transparently report antibody details including catalog numbers, dilutions, and validation methods to support experimental reproducibility and help explain potential discrepancies across studies.
The most reliable validation methods for VAMP2 antibodies combine multiple orthogonal approaches to ensure specificity and reproducibility. Genetic validation using siRNA/shRNA knockdown or CRISPR/Cas9 knockout models provides definitive evidence of antibody specificity when signal reduction corresponds with protein depletion . Published research demonstrates successful validation through ~54% reduction in VAMP2 protein following shRNA treatment, with corresponding functional effects on exocytosis .
Peptide competition assays using the immunizing peptide (such as PEP-101 for PA1-766 antibody) to block antibody binding provide direct evidence of epitope specificity . Western blotting should show bands at the expected molecular weight (12.7-19 kDa for VAMP2), with recombinant protein standards serving as size references . Cross-validation across multiple applications (Western blot, immunofluorescence, immunoprecipitation) strengthens confidence in antibody performance.
For newly developed antibodies, heterologous expression systems overexpressing tagged VAMP2 provide controlled validation environments. Antibody validation should also include cross-reactivity testing against other VAMP family members, particularly the highly homologous VAMP3. To ensure reproducibility, researchers should maintain detailed records of antibody performance across batches and experimental conditions, while following reporting guidelines such as providing complete antibody information (catalog numbers, RRID identifiers, dilutions) in publications .
The future of VAMP2 antibody research will likely address several current limitations through technological and methodological innovations. Development of highly selective monoclonal antibodies with validated specificity for different VAMP2 post-translational modifications (phosphorylation at S61, S75, T79; ubiquitination at K52, K59; methylation at R66) will enable more nuanced studies of regulatory mechanisms . Antibodies capable of distinguishing between free VAMP2 and SNARE complex-incorporated VAMP2 would significantly advance our understanding of vesicle fusion dynamics.
Advances in antibody engineering will likely produce smaller antibody fragments (nanobodies, single-chain antibodies) with enhanced tissue penetration and reduced steric hindrance for super-resolution microscopy applications. These smaller probes will enable more precise localization of VAMP2 at the synapse and other sites of vesicle fusion. For live-cell applications, non-interfering antibody-based biosensors that report on VAMP2 conformational changes during SNARE complex assembly would provide unprecedented insights into fusion kinetics.
Standardization initiatives will establish validated antibody panels and protocols for VAMP2 detection across different experimental systems, enhancing reproducibility across laboratories. Integration with complementary technologies such as spatial transcriptomics and proteomics will provide comprehensive views of VAMP2 regulation in different cellular contexts. Finally, development of translational applications, including VAMP2-targeted diagnostic tools for neurological or metabolic disorders, will bridge basic research advances to clinical applications, expanding the impact of this field beyond fundamental mechanisms to therapeutic interventions.
Application | Recommended Antibody Type | Optimal Dilution Range | Critical Controls |
---|---|---|---|
Western Blot | Polyclonal or Monoclonal | 1:500-1:2000 | Recombinant protein, VAMP2 knockout tissue |
Immunohistochemistry | Monoclonal preferred | 1:100-1:500 | Peptide competition, VAMP2 knockout tissue |
Immunofluorescence | Monoclonal preferred | 1:100-1:500 | Primary omission, VAMP2 knockdown cells |
Immunoprecipitation | Monoclonal preferred | 1-5 μg per sample | IgG control, VAMP2 knockout lysate |
Flow Cytometry | Directly conjugated | 1:50-1:200 | Isotype control, VAMP2 knockdown cells |
ELISA | High-affinity monoclonal | 1:1000-1:5000 | Standard curve, VAMP2 knockout samples |
Modification Type | Site | Function | Detection Method |
---|---|---|---|
Ubiquitination | K52, K59 | Protein turnover regulation | IP-Western with ubiquitin antibodies |
Phosphorylation | S61 | Modulates fusion kinetics | Phospho-specific antibodies |
Methylation | R66 | Regulates protein interactions | Methylation-specific antibodies |
Phosphorylation | S75 | Activity regulation | Phospho-specific antibodies |
Phosphorylation | T79 | Response to signaling | Phospho-specific antibodies |
Issue | Possible Causes | Solutions |
---|---|---|
No signal in Western blot | Protein degradation, low expression | Use fresh samples, enrich vesicular fraction |
Multiple bands | Cross-reactivity, PTMs, degradation | Use more specific antibodies, include protease inhibitors |
High background in IF | Non-specific binding, autofluorescence | Optimize blocking, use monoclonal antibodies |
Signal in knockout controls | Antibody cross-reactivity | Validate knockouts, try alternative antibodies |
Inconsistent results | Antibody batch variation | Use same lot, include internal standards |
Poor signal in fixed tissue | Epitope masking | Try alternative fixation methods, antigen retrieval |
Synaptobrevin-2, also known as Vesicle-associated membrane protein 2 (VAMP2), is a crucial component of the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex. This protein plays a pivotal role in the process of membrane fusion, which is essential for neurotransmitter release at synapses .
Synaptobrevin-2 is an 18 kDa integral membrane protein localized to the cytoplasmic surface of synaptic vesicles . It consists of a proline-rich N-terminal region, a highly conserved hydrophilic domain, followed by a transmembrane anchor and a C-terminal . The protein is largely unstructured in the absence of interaction partners but stabilizes upon interaction with other SNARE proteins, forming a four-helix bundle .
The primary function of Synaptobrevin-2 is to mediate the fusion of synaptic vesicles with the plasma membrane, facilitating the release of neurotransmitters into the synaptic cleft . This process is critical for the transmission of signals between neurons in the central nervous system (CNS) .
The Mouse Anti Human Synaptobrevin-2 antibody is a monoclonal antibody derived from the hybridization of mouse SP2/O myeloma cells with spleen cells from BALB/c mice immunized with recombinant human VAMP2 . This antibody is used in various laboratory research applications, including ELISA and Western blot analysis, to study the expression and function of Synaptobrevin-2 .