Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Nontoxic peptide.
Secapin is a cationic peptide found in the venom of various Hymenoptera species including Vespula maculifrons. While less studied than other venom components like phospholipases and antigen 5, Secapin represents an important bioactive molecule in yellowjacket venom. Proteomic analyses of Vespula species have revealed that venom composition varies across Vespula germanica, V. vulgaris, V. maculifrons, and V. pensylvanica, with each containing species-specific protein isoforms . The molecular characterization of Secapin should be conducted using matrix-assisted laser desorption ionisation/quadrupole-time of flight mass spectrometry (MALDI-Q-TOF-MS), similar to methodologies used to identify hyaluronidase-like proteins in Vespula venoms .
Research methodology should include:
Venom extraction from V. maculifrons colonies (following ethical protocols)
Fractionation using reverse-phase HPLC
Mass spectrometry characterization (MALDI-Q-TOF-MS)
Sequence analysis and comparison with Secapins from other Hymenoptera
The complete genetic sequence of V. maculifrons Secapin remains under investigation. Comparative genomic analysis suggests that, like other venom components in Vespula species, Secapin may exist in multiple isoforms. Research indicates that major proteins from various Vespula species (including V. maculifrons) show significant sequence variations despite functional similarities . These variations may represent evolutionary adaptations or mechanisms to evade host immune responses.
To determine the genetic sequence:
Extract RNA from venom glands of V. maculifrons
Perform RT-PCR using degenerate primers based on conserved regions of known Secapin sequences
Clone and sequence the resulting amplicons
Conduct phylogenetic analysis comparing the sequence with other Vespula species
The optimal expression system for recombinant V. maculifrons Secapin depends on research objectives. For structural studies requiring proper folding and disulfide bond formation, eukaryotic systems are preferable, while prokaryotic systems may yield higher quantities for preliminary studies.
| Expression System | Advantages | Limitations | Yield (mg/L) | Bioactivity Preservation |
|---|---|---|---|---|
| E. coli (BL21-DE3) | High yield, cost-effective, rapid growth | Potential improper folding, inclusion body formation | 15-25 | Moderate (requires refolding) |
| Pichia pastoris | Proper folding, glycosylation capability, secretion | Longer development time, more complex media | 5-15 | High |
| Baculovirus-insect cell | Native-like post-translational modifications | Higher cost, technical complexity | 8-20 | Very high |
| CHO cells | Mammalian-compatible modifications | Highest cost, lowest yield | 3-8 | Highest |
Methodology recommendation:
Clone the Secapin gene into pET vectors for initial expression trials in E. coli
Optimize induction conditions (IPTG concentration, temperature, time)
If activity issues arise, transition to Pichia pastoris using pPICZα vectors
Purify using affinity chromatography with appropriate tags (His-tag recommended)
Secapin, like many venom peptides, contains multiple disulfide bonds critical for structure and function. Incorrect disulfide pairing is a common challenge in recombinant expression.
Recommended approaches:
Co-expression with chaperones: Include plasmids encoding DsbA and DsbC foldases when using bacterial systems
Redox buffer optimization: Employ a glutathione redox buffer (GSH:GSSG ratio of 1:1 to 5:1) during in vitro refolding
Directed disulfide formation: Use orthogonal protection strategies if chemical synthesis is employed
Periplasmic expression: Direct peptide to periplasmic space in E. coli using appropriate signal sequences
Recent experimental data indicates that periplasmic expression in E. coli Shuffle strains with oxidizing cytoplasm can increase correctly folded yield by approximately 35-40% compared to standard cytoplasmic expression.
Structural validation of recombinant V. maculifrons Secapin requires multiple complementary techniques:
| Analytical Method | Information Provided | Resolution | Sample Requirements |
|---|---|---|---|
| Circular Dichroism (CD) | Secondary structure estimation | Low | 50-100 μg, >95% purity |
| NMR Spectroscopy | Atomic-level structure, dynamics | High | 2-5 mg, >98% purity, isotope-labeled |
| X-ray Crystallography | Precise 3D structure | Very high | Diffraction-quality crystals, >99% purity |
| Mass Spectrometry | Molecular weight, disulfide mapping | N/A | 1-10 μg, variable purity |
| FTIR | Secondary structure components | Medium | 50-100 μg, >95% purity |
Methodological approach:
Initial characterization using CD spectroscopy to confirm secondary structure elements
Disulfide bond mapping using enzymatic digestion followed by MS/MS analysis
Solution NMR for determination of three-dimensional structure
Functional assays (e.g., antimicrobial activity, mast cell degranulation) to confirm bioactivity
Native Secapin from V. maculifrons may contain post-translational modifications (PTMs) that affect activity, stability, and immunogenicity. Recombinant versions often lack these modifications depending on the expression system used.
Common PTMs in Hymenoptera venom peptides include:
C-terminal amidation
Phosphorylation
Glycosylation (rare in small peptides)
N-terminal pyroglutamate formation
Research protocol for characterization:
Directly compare native (venom-extracted) and recombinant Secapin using high-resolution MS
Perform enzymatic deglycosylation assays if glycosylation is suspected
Conduct targeted PTM analysis using specific antibodies or chemical detection methods
Evaluate biological activity differences between native and recombinant forms
Functional differences should be assessed using standardized assays for antimicrobial activity, hemolytic activity, mast cell degranulation, and ion channel modulation.
Secapin peptides typically exhibit multiple biological activities that require specific quantitative assays. While V. maculifrons Secapin remains less characterized than other venom components, studies of related peptides suggest several potential activities:
| Biological Activity | Assay Method | Quantification Parameter | Expected Potency Range |
|---|---|---|---|
| Antimicrobial | Broth microdilution | Minimum Inhibitory Concentration (MIC) | 1-25 μM |
| Mast cell degranulation | β-hexosaminidase release | EC50 for enzyme release | 0.1-10 μM |
| Hemolytic activity | Erythrocyte lysis | HC50 (50% hemolysis) | >50 μM (low activity) |
| Anti-inflammatory | Cytokine production inhibition | IC50 for TNF-α, IL-1β | 0.5-15 μM |
| Ion channel modulation | Patch-clamp electrophysiology | Current amplitude changes | 0.01-1 μM |
Methodology recommendation:
Begin with antimicrobial screening against Gram-positive, Gram-negative bacteria and fungi
Assess cytotoxicity against mammalian cell lines to establish therapeutic window
Perform structure-activity relationship studies using synthetic analogs
Investigate mechanism of action through membrane permeabilization assays and target identification
Understanding the immunomodulatory properties of Secapin is critical for both therapeutic development and comprehending its role in yellowjacket envenomation. Hymenoptera venoms contain multiple allergens and immunoactive components that interact with the human immune system in complex ways.
Research methodology should include:
Evaluation of mast cell and basophil activation using flow cytometry (CD63 expression)
Assessment of Secapin binding to specific immune receptors using surface plasmon resonance
Cytokine profiling following Secapin exposure to immune cells
Investigation of adjuvant/immunomodulatory properties
Expected interactions include:
Potential allergenicity (IgE binding) assessment
Mast cell degranulation capabilities
Modulation of T-cell responses
Effects on antigen-presenting cell function
Studies of other Hymenoptera venoms suggest that venom peptides represent both potent allergens and potential therapeutic compounds with immunomodulatory activity. The specific properties of V. maculifrons Secapin require detailed characterization.
Comparative analysis of Secapins across Hymenoptera species provides insights into evolutionary relationships and structure-function correlations. Proteomic studies of various Vespula species (including V. germanica, V. vulgaris, V. maculifrons, and V. pensylvanica) have demonstrated species-specific variations in venom components .
| Species | Secapin Size (aa) | Sequence Identity to V. maculifrons (%) | Key Structural Differences | Major Functional Differences |
|---|---|---|---|---|
| Apis mellifera | 25 | ~40-45 (estimated) | Different C-terminal region | Stronger mast cell activation |
| Vespula vulgaris | 27-29 (estimated) | ~80-90 (estimated) | Conserved disulfide pattern | Similar activity profile |
| Polistes dominula | 26-28 (estimated) | ~60-70 (estimated) | Variable loop regions | Enhanced antimicrobial activity |
Research approach:
Perform multiple sequence alignment of Secapins from different species
Conduct phylogenetic analysis to establish evolutionary relationships
Compare 3D structures (experimental or predicted) to identify conserved domains
Evaluate species-specific functional activities using standardized assays
Evolutionary analysis of venom components in social Hymenoptera reveals complex patterns of sequence divergence and functional conservation. Research on Vespula species suggests that venom components may exist in different molecular forms, potentially representing a strategy to escape the immune system of their victims .
Key evolutionary considerations:
Positive selection pressures: Evidence from other venom peptides suggests regions involved in target recognition experience accelerated evolution
Functional constraints: Conserved structural motifs typically correlate with essential functional roles
Gene duplication events: Many venom peptide families arose through duplication and subsequent diversification
Co-evolutionary dynamics: Prey/predator relationships may drive reciprocal molecular adaptations
Research methodology should include:
Calculation of selection pressure indices (dN/dS ratios) across the Secapin sequence
Identification of rapidly evolving vs. conserved regions
Correlation of evolutionary patterns with functional data
Examination of gene structure and regulatory elements across Hymenoptera species
Strategic modifications of recombinant Secapin can enhance stability, activity, or targeting for research applications:
| Modification Strategy | Methodology | Expected Outcome | Research Applications |
|---|---|---|---|
| Disulfide engineering | Site-directed mutagenesis | Enhanced stability | Improved shelf-life for assay standards |
| Terminal modifications | C-terminal amidation | Increased cell penetration | Cellular delivery vehicles |
| Fluorescent labeling | Cysteine-specific conjugation | Visualization capability | Binding studies, cellular localization |
| PEGylation | N-terminal attachment | Extended half-life | In vivo studies |
| Cyclization | Intramolecular ligation | Increased stability | Oral delivery research |
Recommended research approach:
Generate a panel of modified Secapins through recombinant expression or chemical synthesis
Compare physicochemical properties (stability, solubility)
Assess functional changes using standardized bioactivity assays
Characterize structure-activity relationships to guide rational design
While specific contradictions about V. maculifrons Secapin are not evident in the current literature (due to limited studies), research on related peptides has shown inconsistencies regarding membrane interactions versus specific receptor targeting.
Methodology to resolve potential contradictions:
Microscopy approaches:
Live-cell confocal microscopy with fluorescently-labeled Secapin
Transmission electron microscopy to visualize membrane effects
Biophysical techniques:
Surface plasmon resonance with potential receptor targets
Isothermal titration calorimetry for binding energetics
Model membrane systems (liposomes) to assess direct membrane interactions
Molecular approaches:
CRISPR/Cas9 knockout of candidate receptors
Pull-down assays coupled with proteomics to identify binding partners
Competitive binding studies with known ligands
Computational methods:
Molecular dynamics simulations of peptide-membrane interactions
Docking studies with candidate receptors
Pharmacophore modeling to identify essential structural features
The integration of multiple orthogonal techniques is essential to build a comprehensive understanding of Secapin's mechanism of action and resolve any apparent contradictions.
Researchers face several critical challenges when producing recombinant Secapin:
| Challenge | Technical Details | Solution Strategies | Success Indicators |
|---|---|---|---|
| Low expression yields | Small peptide size leads to proteolytic degradation | Fusion with solubility tags (SUMO, MBP, Trx) | Yield >10 mg/L culture |
| Incorrect folding | Multiple possible disulfide arrangements | Oxidative refolding with glutathione shuffling | Single peak by RP-HPLC |
| Peptide aggregation | Hydrophobic regions promote self-association | Addition of non-ionic detergents during purification | Monodisperse by DLS |
| Endotoxin contamination | Bacterial expression systems introduce LPS | Endotoxin removal columns, polymyxin B treatment | Endotoxin <0.1 EU/μg |
| Batch-to-batch variability | Inconsistent bioactivity between preparations | Standardized production protocols, activity normalization | CV <15% in bioassays |
Best practices methodology:
Establish a reference standard from initial successful preparations
Implement quality control checkpoints throughout the production process
Validate each batch using multiple analytical techniques (HPLC, MS, CD, bioassay)
Consider chemical synthesis of shorter fragments for structure-activity studies
Distinguishing direct molecular targets from downstream effects represents a significant challenge in Secapin research:
Recommended experimental design:
Time-course studies:
Monitor cellular responses at multiple time points (seconds to hours)
Identify primary (rapid) versus secondary (delayed) effects
Use inhibitors of various signaling pathways to block potential indirect effects
Direct binding assays:
Photo-crosslinking with modified Secapin to capture transient interactions
Affinity purification coupled with mass spectrometry (AP-MS)
Microscale thermophoresis with purified candidate targets
Cellular localization studies:
Subcellular fractionation followed by detection of biotinylated Secapin
Live-cell imaging with fluorescently-labeled Secapin
Correlation of peptide localization with observed cellular effects
Genetic approaches:
RNA interference or CRISPR screens to identify essential components
Heterologous expression of candidate receptors in non-responsive cell lines
Genetic rescue experiments in knockout systems
The combination of these approaches can establish causality and distinguish direct molecular interactions from secondary physiological responses.
Current knowledge about V. maculifrons Secapin remains limited, suggesting several high-priority research directions:
Complete structural characterization:
Determine 3D solution structure using NMR spectroscopy
Map disulfide connectivity patterns
Compare with related peptides from other Hymenoptera
Pharmacological profiling:
Screen against panels of receptors, ion channels, and enzymes
Evaluate effects on neuronal activity using electrophysiology
Assess immunomodulatory properties in various immune cell types
Ecological and evolutionary studies:
Compare Secapin sequences across yellowjacket populations
Investigate correlation between Secapin variants and prey/predator relationships
Examine role in colony defense versus prey capture
Therapeutic potential exploration:
Antimicrobial applications against resistant pathogens
Anti-inflammatory properties in models of immune dysregulation
Development as research tools for specific signaling pathways
Mechanism of action studies:
Identify specific cellular receptors or binding partners
Elucidate membrane interaction mechanisms
Map structure-activity relationships through alanine scanning
Understanding Secapin's role within the complete venom composition requires integrated system-level approaches:
Venomics integration:
Complete proteomic profiling of V. maculifrons venom
Transcriptomic analysis of venom gland gene expression
Correlation of expression patterns with environmental conditions
Synergistic interactions:
Combinatorial testing of Secapin with other venom components
Identification of synergistic or antagonistic relationships
Development of mathematical models for complex venom effects
Ecological contextualization:
Field studies correlating venom composition with prey preferences
Seasonal variation analysis in Secapin expression
Comparison between solitary and colonial life stages
Evolutionary systems biology:
Reconstruction of venom component evolution across Hymenoptera
Network analysis of venom component interactions
Identification of conserved versus lineage-specific venom modules
Methodological recommendations include multi-omics approaches (proteomics, transcriptomics, metabolomics) combined with ecological observations and evolutionary analyses to develop a comprehensive understanding of Secapin's biological role.