Recombinant Conus vexillum Conotoxin A11GB

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference during order placement for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please consult your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to ensure contents settle. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50% and can serve as a reference.
Shelf Life
Shelf life depends on storage conditions, buffer components, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If you require a specific tag, please inform us; we will prioritize its development.
Synonyms
; Conotoxin A11GB
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
55-81
Protein Length
Cytoplasmic domain
Purity
>85% (SDS-PAGE)
Species
Conus vexillum (Flag cone)
Target Protein Sequence
CQRANF VCDAFHHAAV CCEGVCVLVC A
Uniprot No.

Target Background

Protein Families
Conotoxin O1 superfamily
Subcellular Location
Secreted.
Tissue Specificity
Expressed by the venom duct.

Q&A

What are the structural characteristics of Conus vexillum conotoxins?

Conus vexillum conotoxins are small peptides typically characterized by specific cysteine frameworks that form disulfide bonds critical to their three-dimensional structure and biological function. These peptides generally contain between 10-30 amino acid residues with distinctive cysteine patterns that define their classification and folding properties . The specific arrangement of cysteine residues (such as -CC-CC-C-C- or -C-C-CC-) is a key determinant of structural diversity and functional specificity . These small peptides adopt rigid conformations stabilized by disulfide bridges, which allow precise interactions with their target ion channels.

Alpha-conotoxins from Conus vexillum, like many conotoxins, undergo post-translational modifications including C-terminal amidation, hydroxyproline formation, and occasionally N-terminal pyroglutamate modification, all of which contribute to their stability and target specificity . The structural integrity of these peptides is critical for their ability to interact with specific binding sites on ion channels and receptors, making them valuable tools for neuroscience research and potential therapeutic development.

How do Conus vexillum conotoxins differ functionally from other conotoxin families?

Conus vexillum conotoxins, like those from other cone snail species, exhibit remarkable target specificity depending on their structural framework and amino acid composition. While conotoxins as a group target various ion channels and receptors, individual Conus vexillum peptides have evolved specific targeting profiles, often reflecting the predatory strategies of this particular cone snail species .

Some Conus vexillum conotoxins primarily target voltage-gated sodium channels, similar to the μ-conotoxins that act as pore blockers or the δ-conotoxins that modify inactivation gating . Others may act on nicotinic acetylcholine receptors (nAChRs) as competitive antagonists, similar to α-conotoxins from other species . The functional diversity among Conus vexillum peptides reflects their evolutionary adaptation to immobilize prey through sophisticated neuropharmacology, targeting multiple components of the nervous system simultaneously.

What distinguishes Conus vexillum conotoxins from other species' toxins are subtle differences in amino acid sequences that confer specificity for particular subtypes of ion channels. For example, certain residues may determine whether a conotoxin preferentially blocks NaV1.4 (skeletal muscle) versus NaV1.6 (neuronal) sodium channels, as observed with the μ-conotoxin GIIIC variants . These functional differences make each conotoxin family valuable for investigating specific channel subtypes in research contexts.

What expression systems are most effective for recombinant Conus vexillum conotoxin production?

E. coli expression systems are commonly used for recombinant conotoxin production due to their efficiency and cost-effectiveness, as evidenced by the recombinant Conus vexillum Alpha-conotoxin VxXXC product information . When utilizing E. coli for conotoxin expression, researchers typically employ fusion tag strategies to enhance solubility and facilitate purification. The N-terminal 10X-His-SUMO tag approach has proven particularly effective for maintaining peptide solubility during expression while providing a convenient purification handle .

For more complex conotoxins with multiple disulfide bonds, eukaryotic expression systems such as Pichia pastoris or mammalian cell lines may offer advantages in proper disulfide bond formation and post-translational modifications. The choice of expression system should be guided by the specific structural complexity of the target conotoxin and required post-translational modifications. Researchers should consider that while E. coli systems are simpler to implement, they may require additional refolding steps to achieve the correct disulfide bond arrangement.

When expressing recombinant conotoxins, optimization of induction conditions (temperature, IPTG concentration, induction time) is critical for maximizing yield while maintaining proper folding. Typically, lower temperatures (16-20°C) and longer induction times produce better results for complex disulfide-rich peptides. Additionally, the inclusion of oxidative folding buffers containing appropriate redox reagents (glutathione, cysteine/cystine) during purification significantly improves the yield of correctly folded peptides.

What purification strategies maximize yield and biological activity of recombinant conotoxins?

Effective purification of recombinant conotoxins requires a multi-step approach that preserves the critical disulfide bonds while removing contaminants. Immobilized metal affinity chromatography (IMAC) utilizing the His-tag is typically employed as the initial capture step, followed by precise tag removal using SUMO protease cleavage . This strategy allows for native N-terminal sequence generation without residual amino acids that might interfere with biological activity.

Reverse-phase high-performance liquid chromatography (RP-HPLC) is essential for achieving high purity (>85% as typically required for research applications) . The selection of appropriate column chemistry (C18 or C8) and optimization of acetonitrile gradients with trifluoroacetic acid (TFA) or formic acid modifiers is critical for separating correctly folded peptides from misfolded variants. Ion exchange chromatography may serve as an orthogonal purification step for removing endotoxins and other charged contaminants.

For maintaining biological activity, buffer formulation during final purification steps is crucial. Tris/PBS-based buffers with 5-50% glycerol have proven effective for liquid formulations , while lyophilized preparations benefit from the inclusion of stabilizers such as trehalose (6%) to prevent aggregation during freeze-drying and reconstitution . Quality control through mass spectrometry, circular dichroism, and bioactivity assays is essential to confirm correct folding and functional activity of the purified peptide.

What analytical methods best characterize the structural integrity of recombinant conotoxins?

Comprehensive structural characterization of recombinant conotoxins requires a combination of complementary analytical techniques. High-resolution mass spectrometry is indispensable for confirming the exact molecular weight and sequence integrity of the purified peptide . Tandem MS/MS analysis following enzymatic digestion can verify the presence of post-translational modifications and confirm the disulfide bonding pattern, which is crucial for structure-function correlations in conotoxin research.

Nuclear Magnetic Resonance (NMR) spectroscopy provides the gold standard for determining the three-dimensional solution structure of conotoxins . 2D techniques such as TOCSY, NOESY, and HSQC can elucidate the specific folding pattern and confirm the correct formation of disulfide bridges. Circular dichroism (CD) spectroscopy serves as a rapid screening tool for secondary structure elements and can monitor conformational stability under various experimental conditions.

X-ray crystallography, though challenging with small peptides, can provide atomic-resolution structures when conotoxins are co-crystallized with their target proteins. This approach has been particularly valuable for understanding the precise binding interactions between μ-conotoxins and sodium channel pore domains . Combining these structural analyses with molecular dynamics simulations allows researchers to predict binding orientations and rationalize the effects of sequence variations on target selectivity.

How can researchers validate the functional activity of recombinant conotoxins?

Electrophysiological techniques provide the most direct validation of conotoxin functional activity. Patch-clamp recordings using recombinant cell lines expressing specific ion channel subtypes allow precise determination of IC50 values and kinetic parameters of inhibition . For sodium channel-targeting conotoxins, recordings from dorsal root ganglion (DRG) neurons can assess isoform selectivity against multiple endogenously expressed channels simultaneously.

Radioligand binding assays offer complementary approaches for quantifying binding affinities. Competition binding experiments using radiolabeled reference toxins (such as 125I-labeled α-bungarotoxin for nAChR-targeting conotoxins) can determine the binding site overlap and relative affinities . These assays are particularly valuable for high-throughput screening of conotoxin variants to establish structure-activity relationships.

Functional calcium imaging in neuronal cultures provides a medium-throughput approach to assess the physiological effects of conotoxins on cellular excitability. This method can reveal subtle effects on neuronal firing patterns that might not be apparent in isolated channel recordings. For in vivo validation, behavioral assays measuring antinociceptive, antiepileptic, or neuroprotective activities in animal models provide the ultimate confirmation of therapeutic potential, as has been demonstrated for numerous conotoxins .

How can recombinant conotoxins be utilized as molecular probes for ion channel research?

Recombinant conotoxins serve as exquisite molecular probes for investigating ion channel structure-function relationships. Their high subtype selectivity enables researchers to pharmacologically isolate specific channel isoforms in complex biological systems . For example, μ-conotoxins can distinguish between skeletal muscle (NaV1.4) and neuronal (NaV1.6) sodium channel isoforms, allowing targeted investigation of channel contributions to cellular excitability .

Mutational analysis combined with electrophysiological characterization has proven invaluable for mapping the molecular determinants of channel-toxin interactions. Mutant cycle analysis involving systematic mutations in both the conotoxin and its target channel can quantify energetic contributions of specific residue interactions, as demonstrated with μ-conotoxins GIIIA and GIIIB . These approaches have revealed that conotoxins often interact with multiple regions of the channel pore vestibule rather than binding exclusively to the selectivity filter.

Fluorescently labeled conotoxins enable visualization of receptor distribution and trafficking in living cells. Site-specific conjugation strategies using maleimide chemistry at engineered cysteine residues or bio-orthogonal reactions at incorporated unnatural amino acids minimize interference with biological activity. Such modified conotoxins have facilitated studies of receptor internalization dynamics and have proven particularly valuable for investigating nAChR subtypes in neuronal systems .

What considerations are critical when designing experiments with recombinant conotoxins?

When designing experiments with recombinant conotoxins, researchers must carefully consider proper storage and handling to maintain peptide integrity. Lyophilized preparations should be reconstituted in appropriate buffers (typically phosphate-buffered solutions with minimal exposure to reducing agents) and aliquoted to prevent repeated freeze-thaw cycles that can compromise disulfide bond integrity . For long-term storage, temperatures of -20°C or below are recommended .

Proper experimental controls are essential for interpreting conotoxin effects. These should include negative controls using heat-denatured peptide, scrambled sequence peptides, or competitive displacement with known ligands. Dose-response relationships should be established across a wide concentration range to determine both potency (IC50) and efficacy (maximum inhibition). Time-course experiments are crucial for distinguishing between direct pore blockade (typically rapid onset) and modulatory effects on channel gating (often showing use-dependence).

The experimental preparation must be carefully matched to the research question. Heterologous expression systems provide clean backgrounds for detailed mechanistic studies but may lack relevant auxiliary subunits or post-translational modifications. Primary neuronal cultures or acute tissue preparations offer more physiologically relevant contexts but introduce greater complexity in data interpretation. In vivo studies require careful consideration of peptide delivery methods, as conotoxins generally have poor blood-brain barrier penetration and often require direct administration to the central nervous system .

How can recombinant conotoxin analogs be designed for enhanced stability and target specificity?

Developing enhanced conotoxin analogs requires strategic modification approaches while preserving critical structural elements. Backbone cyclization has emerged as a powerful strategy for improving proteolytic stability without compromising biological activity. This approach involves connecting the N- and C-termini via a linker sequence, creating a cyclic peptide with enhanced resistance to exoproteases while maintaining the critical disulfide framework .

Incorporation of unnatural amino acids at strategic positions can enhance both stability and target specificity. D-amino acid substitutions at susceptible proteolytic sites can significantly extend the peptide's half-life in biological fluids. Meanwhile, introducing N-methylated amino acids can modulate backbone flexibility and membrane permeability. Computational modeling guided by known structure-activity relationships can predict which positions can tolerate such modifications without disrupting the binding interface .

Disulfide bond engineering represents another sophisticated approach for optimizing conotoxin properties. Selective replacement of native disulfide bridges with more stable thioether linkages can enhance stability while maintaining the critical three-dimensional scaffold. Alternatively, strategic addition of hydrophobic residues at solvent-exposed positions can improve membrane association and target accessibility. These rational design approaches have led to development of several conotoxin-derived compounds with improved pharmacokinetic properties while maintaining exquisite selectivity for their targets .

What are the challenges in translating recombinant conotoxin research to therapeutic applications?

Translating recombinant conotoxins to therapeutic applications faces several significant challenges, beginning with scalable manufacturing. While E. coli expression systems work well for research quantities, pharmaceutical-scale production requires development of consistent, high-yield processes that maintain precise disulfide bond formation . Regulatory considerations are substantial, as recombinant peptides must demonstrate consistent post-translational modification profiles across manufacturing batches.

Delivery and bioavailability present major hurdles due to the peptidic nature of conotoxins. Poor oral bioavailability necessitates alternative administration routes, with several conotoxin-based therapeutics requiring intrathecal delivery . Development of novel formulations incorporating cell-penetrating peptide tags, encapsulation in nanoparticles, or conjugation to antibody fragments represents active areas of research to overcome these limitations.

What are common challenges in recombinant conotoxin expression and how can they be overcome?

Incorrect disulfide bond formation represents the most significant challenge in recombinant conotoxin production. This often manifests as multiple chromatographic peaks during purification, each representing different disulfide isomers with varying biological activity . To address this issue, researchers can implement oxidative refolding protocols using optimized glutathione redox systems (typically 1:10 ratio of reduced:oxidized glutathione) and include folding additives such as L-arginine to prevent aggregation during the refolding process.

Low expression yields frequently occur with highly disulfide-rich conotoxins. This can be mitigated by using specialized E. coli strains engineered for disulfide bond formation in the cytoplasm (such as Origami or SHuffle strains) or by directing expression to the periplasmic space where natural disulfide bond formation machinery exists . Codon optimization of the synthetic gene for the expression host and reduction of expression temperature to 16-18°C can significantly improve yields of correctly folded product.

Proteolytic degradation during expression can compromise yields, particularly for smaller conotoxins. This can be addressed by co-expression with protease inhibitors, use of protease-deficient host strains, or strategic fusion partner selection. The SUMO fusion approach has proven particularly effective, as it both enhances solubility and provides steric protection against proteolytic attack . Following purification, proper buffer formulation with stabilizing agents such as trehalose for lyophilized products helps maintain integrity during storage .

How can researchers optimize experimental conditions for accurate functional characterization?

Accurate functional characterization of recombinant conotoxins requires careful consideration of experimental conditions that can influence peptide-target interactions. Buffer composition significantly impacts conotoxin activity, with divalent cation concentrations (particularly calcium and magnesium) modulating the apparent potency in electrophysiological assays . Standardized recording solutions should be established and maintained across experiments to ensure comparability of results.

Non-specific binding to experimental apparatus can lead to apparent loss of activity and inconsistent dose-response relationships. This can be minimized by including low concentrations (0.1-0.5%) of bovine serum albumin or other carrier proteins in experimental solutions and by using low-binding pipette tips and containers for peptide handling. Serial dilutions should be prepared fresh for each experiment to avoid concentration errors from adsorptive losses.

Temperature dependence of conotoxin binding kinetics can significantly impact experimental outcomes. While room temperature recordings are common for convenience, maintaining consistent temperature control (typically 22-23°C) across experiments is essential for reproducibility. For in-depth kinetic analyses, recordings at physiological temperature (37°C) provide more relevant data for translational applications, though this may accelerate peptide degradation in prolonged experiments. Researchers should verify batch-to-batch consistency through quality control assays such as mass spectrometry and activity testing against reference standards .

What emerging technologies might advance recombinant conotoxin research?

Cryo-electron microscopy (cryo-EM) has revolutionized structural biology of membrane proteins and holds tremendous promise for visualizing conotoxin-channel complexes at near-atomic resolution. Recent advances in single-particle analysis have enabled structure determination of several voltage-gated ion channels bound to toxins . This technology will likely provide unprecedented insights into the precise binding modes of conotoxins with their molecular targets, facilitating rational design of optimized analogs.

Cell-free protein synthesis systems offer exciting possibilities for rapid production of conotoxin libraries. These systems bypass cellular viability constraints and can be supplemented with specialized folding chaperones and disulfide isomerases to enhance correct folding. High-throughput cell-free expression platforms coupled with activity screening assays could dramatically accelerate the discovery and optimization of novel conotoxin-based channel modulators.

CRISPR/Cas9 gene editing technologies enable precise engineering of cell lines expressing modified ion channels. This approach allows investigation of conotoxin interactions with chimeric or mutant channels that would be difficult to study using traditional methods. Creation of knock-in animal models with humanized ion channel binding sites could bridge the translational gap between in vitro pharmacology and in vivo efficacy, addressing a significant challenge in conotoxin drug development .

What new therapeutic applications might emerge from advanced conotoxin research?

Conotoxin-based pain management represents one of the most promising therapeutic applications. Unlike opioids, conotoxins targeting specific ion channel subtypes can potentially provide effective analgesia without addiction liability or respiratory depression . Advanced research into selective Nav1.7 or Nav1.8 inhibitors derived from μO-conotoxins could yield transformative treatments for chronic pain conditions that are resistant to current therapies.

Neurodegenerative disease treatment represents an emerging frontier for conotoxin applications. Conotoxins that modulate calcium influx through selective inhibition of particular channels might provide neuroprotection in conditions like Alzheimer's disease, where calcium dysregulation contributes to neuronal death . Similarly, conotoxins targeting specific nicotinic acetylcholine receptor subtypes could have applications in Parkinson's disease, where cholinergic dysfunction plays a significant role.

Epilepsy therapy could benefit from conotoxin-derived precision medicines. Recombinant conotoxins with high selectivity for sodium channel subtypes implicated in particular forms of epilepsy could provide targeted seizure control with fewer side effects than current broad-spectrum anticonvulsants . The reversible mechanism of action and potential for isoform-specific targeting make conotoxins particularly attractive candidates for developing next-generation antiepileptic drugs with improved therapeutic windows.

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.