Recombinant Conus leopardus Alpha-conotoxin-like Lp1.2

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Conus leopardus Alpha-conotoxin-like Lp1.2

Alpha-conotoxins are a family of compounds found in the venom of Conus snails, known for their ability to target nicotinic acetylcholine receptors (nAChRs) . These toxins are of significant interest due to their potential as pharmacological tools and therapeutic agents . Among these, α-conotoxin Lp1.1, derived from Conus leopardus, has been identified and studied for its unique structural and functional characteristics . The Conus leopardus is a worm-hunting marine snail species collected from the South China Sea .

Discovery and Cloning of α-Conotoxin Lp1.1

Researchers have successfully cloned α-conotoxin Lp1.1 from both genomic DNA and cDNA of Conus leopardus . The deduced toxin is considered to be a typical α4/7 conotoxin containing 16 amino acid residues . The process involves analyzing the signal peptide and cysteine patterns, which are highly conserved in conotoxins . Cloning from genomic DNA is considered a better approach to acquire more conotoxins than cDNA cloning .

Synthesis and Folding

The α-conotoxin Lp1.1 can be synthesized using solid-phase peptide synthesis, followed by oxidative folding to form disulfide bonds . The synthetic version of Lp1.1 typically achieves high purity, exceeding 95%, as confirmed by HPLC analysis . The synthetic Lp1.1 has at least 95% purity as analysed by HPLC . Mass spectrometry confirms the accurate assembly of the peptide . The disulfide connectivity of Lp1.1 is assumed to be Cys2-Cys8 and Cys3-Cys16, consistent with the conserved α-conotoxin framework .

Receptor Selectivity

The Conus snails produce toxins that can target different subtypes of the same membrane receptor or ion channel with high affinity, due to their slow mobility and broad diet . α-Conotoxins are known for their selective and potent competitive antagonism of nicotinic acetylcholine receptors . α-Conotoxin Lp1.1 displays a specific blocking effect on α3β2 and α6α3β2 rat nAChR subtypes .

Structure-Function Relationship

The unique primary structure of α-conotoxin Lp1.1, particularly the absence of the conserved Ser and Pro residues, contributes to its specific receptor recognition profile . Comparing the sequences of mature peptides with the nucleotide sequences of the cDNAs encoding conotoxins helps in establishing structure/function relationships . Functional analyses of Lp1.1 provide a basis for designing more potent and selective molecules .

Analgesic Activity

Conotoxin-Ac1, a different conotoxin isolated from Conus achatinus, has shown inhibitory effects on the N-methyl-D-aspartate receptor subunit 2B (NR2B) and displays antinociceptive activity . While α-conotoxin Lp1.1 itself has not been directly linked to analgesic activity, the broader context of conotoxins suggests potential therapeutic applications .

Therapeutic Potential

Conotoxins, including α-conotoxins, have therapeutic potential, as demonstrated by Vc1.1, which is being developed to treat neuropathic pain by accelerating the recovery of injured neurons . The detailed functional analyses of Lp1.1 will provide a basis for establishing structure/function relationships and may facilitate structure-based design studies to produce more potent and selective molecules . Further research into α-conotoxin Lp1.1 and related peptides may uncover additional therapeutic applications .

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes 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: Standard shipping includes 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 collect the contents. 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%, which can serve as a guideline.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The specific tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its inclusion.
Synonyms
Alpha-conotoxin-like Lp1.2
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
49-65
Protein Length
Cytoplasmic domain
Purity
>85% (SDS-PAGE)
Species
Conus leopardus (Leopard cone)
Target Protein Sequence
GC CSHPACSVNN PYFCG
Uniprot No.

Target Background

Function
Alpha-conotoxins exert their effects on postsynaptic membranes by binding to and inhibiting nicotinic acetylcholine receptors (nAChRs).
Protein Families
Conotoxin A superfamily
Subcellular Location
Secreted.
Tissue Specificity
Expressed by the venom duct.

Q&A

What is the primary structure of Conus leopardus alpha-conotoxin Lp1.2?

Alpha-conotoxins from Conus leopardus typically contain 14-18 amino acid residues with the characteristic α4/7 framework, featuring a conserved disulfide connectivity pattern. While specific data on Lp1.2 is limited in the literature, we can infer its structure based on other characterized α-conotoxins from C. leopardus, such as Lp1.4. These peptides contain two disulfide bridges (Cys I-III and Cys II-IV) that create a compact globular structure with a C-terminal amidation .

For Lp1.2, researchers should expect a sequence containing the hallmark cysteine pattern and likely sharing homology with other members of the Lp1.x series. Comprehensive structural characterization requires techniques such as mass spectrometry, circular dichroism, and NMR spectroscopy to confirm the exact sequence and three-dimensional conformation.

How does Lp1.2 compare to other alpha-conotoxins from Conus leopardus?

Conus leopardus produces several alpha-conotoxins, with Lp1.4 being among the better characterized. Lp1.4 shows specificity for the mouse fetal muscle α1β1γδ nicotinic acetylcholine receptor, which is unusual for an α4/7-conotoxin as most members of this subclass target neuronal rather than muscle subtypes .

Based on naming conventions, Lp1.2 likely belongs to the same α4/7 subfamily as Lp1.4 but may exhibit different receptor subtype selectivity. Research comparing Lp1.2 with related toxins would involve:

  • Sequence alignment to identify conserved and variable regions

  • Competitive binding assays against multiple nAChR subtypes

  • Electrophysiological characterization using Xenopus oocyte expression systems

  • Structure-activity relationship studies to identify key pharmacophore elements

What expression systems are optimal for recombinant production of Lp1.2?

Recombinant production of alpha-conotoxins requires special consideration for proper disulfide bond formation. For successful expression of functional Lp1.2, consider these methodological approaches:

  • E. coli expression systems: Use specialized strains with enhanced disulfide formation capability such as Origami or SHuffle. Fusion tags like thioredoxin or SUMO can improve solubility and folding. The oxidative folding environment must be carefully controlled.

  • Yeast expression systems: Pichia pastoris offers advantages for disulfide-rich peptides due to its eukaryotic secretory pathway. Optimization of induction conditions and culture media composition is critical.

  • Chemical synthesis followed by in vitro folding: While not strictly recombinant, solid-phase peptide synthesis allows precise control over the peptide sequence, with subsequent oxidative folding using various buffer systems.

The choice between these systems depends on the required yield, downstream applications, and available resources. For structural studies requiring isotopic labeling, E. coli systems offer cost-effective 15N and 13C incorporation.

What are the critical parameters for optimizing recombinant Lp1.2 folding and disulfide formation?

Proper disulfide bond formation represents the most significant challenge in producing functional recombinant alpha-conotoxins. For Lp1.2, consider these methodological approaches:

In vitro oxidative folding protocol:

  • Synthesize linear peptide with appropriate Cys protection groups

  • For two-step directed folding:

    • First form Cys II-IV using 2% DMSO in buffer (pH 7.5-8.0)

    • Then form Cys I-III using oxidized/reduced glutathione (100:10 ratio)

  • For one-step folding, use glutathione buffer system (1-3 mM GSH:0.1-0.3 mM GSSG)

  • Incubate at room temperature with gentle stirring for 24-48 hours

  • Monitor folding by RP-HPLC and confirm by mass spectrometry

Analysis of folding intermediates is recommended using HPLC profiles at different time points. Alternative folding conditions may include:

Folding ConditionBuffer CompositionTemperatureDurationSpecial Considerations
Standard0.1 M Tris-HCl pH 7.5, 1 mM EDTA, 1 mM GSH, 0.1 mM GSSG25°C24hArgon atmosphere
Rapid0.1 M Tris-HCl pH 7.5, 2 mM EDTA, 2% DMSO4°C48hSlow stirring
High yield0.1 M NH4HCO3 pH 8.0, 0.5 M GuHCl, 0.1 mM GSH, 1 mM GSSG25°C72hInitial denaturation step

A critical quality control step is co-elution studies between synthetic and recombinant products to confirm identical folding patterns.

How can I optimize electrophysiological protocols to characterize Lp1.2 activity on nicotinic acetylcholine receptors?

For electrophysiological characterization of Lp1.2, the Xenopus oocyte expression system offers several advantages. The following protocol provides a methodological framework:

  • nAChR expression in oocytes:

    • Prepare cRNA from linearized cDNA templates encoding target nAChR subunits

    • Inject 5-50 ng of cRNA mixture into defolliculated Xenopus oocytes

    • Incubate at 18°C for 1-3 days in ND96 medium supplemented with gentamicin

  • Two-electrode voltage clamp recordings:

    • Hold oocytes at -70 mV in ND96 recording solution (96 mM NaCl, 2 mM KCl, 1.8 mM CaCl2, 1 mM MgCl2, 5 mM HEPES, pH 7.4)

    • Apply acetylcholine (ACh) at EC50 concentration for 1-2 seconds

    • After stable responses are established, preincubate with Lp1.2 for 3-5 minutes before co-application with ACh

    • Use increasing concentrations of Lp1.2 (1 nM to 10 μM) to generate dose-response curves

  • Data analysis:

    • Normalize responses to control ACh applications

    • Plot concentration-response data and fit with a logistic equation to determine IC50 values

    • Assess on-rate and off-rate kinetics through washout experiments

For comprehensive characterization, test Lp1.2 against multiple nAChR subtypes including:

  • Neuronal: α7, α3β2, α3β4, α6α3β2, α6α3β4

  • Muscle: adult (α1β1εδ) and fetal (α1β1γδ) subtypes

What strategies can resolve challenges in distinguishing Lp1.2 activity from other alpha-conotoxins in mixed samples?

When working with complex venom fractions or heterogeneous expression systems, distinguishing the specific activity of Lp1.2 from other alpha-conotoxins requires specialized approaches:

  • Immunological discrimination:

    • Develop epitope-specific antibodies targeting unique regions of Lp1.2

    • Employ immunodepletion to selectively remove Lp1.2 from complex mixtures

    • Use Western blotting with epitope-specific antibodies for detection and quantification

  • Biochemical separation strategies:

    • Multi-dimensional chromatography combining ion-exchange, RP-HPLC, and size-exclusion

    • Capillary electrophoresis with MS detection for improved resolution

    • Affinity chromatography using immobilized nAChR subunits or receptor fragments

  • Pharmacological discrimination:

    • Competitive binding assays with known alpha-conotoxin subtypes

    • Comparative IC50 determination across multiple receptor subtypes to generate a "fingerprint" profile

    • Use of mutant receptors with altered binding sites for specific conotoxins

  • Genetic approaches:

    • Site-directed mutagenesis of key residues in Lp1.2

    • Introduction of epitope or affinity tags for selective purification

    • Selective knockdown of Lp1.2 expression using RNAi if working with native toxin sources

Which residues in Lp1.2 are critical for selectivity between neuronal and muscle nicotinic acetylcholine receptors?

Based on studies of related alpha-conotoxins, several key positions likely determine the selectivity profile of Lp1.2:

  • Loop 1 residues (residues between the first and second cysteine): Amino acids in this region, particularly at positions 5 and 6, often influence subtype selectivity. Positively charged residues at these positions typically enhance interaction with muscle subtypes.

  • Loop 2 residues (between the third and fourth cysteine): The composition of this loop significantly affects receptor subtype discrimination. Hydrophobic residues in specific positions often contribute to neuronal receptor binding.

  • C-terminal extension: If Lp1.2 possesses a C-terminal extension beyond the final cysteine (similar to Lp1.4 and Lo1a), this region may play a crucial role in subtype selectivity .

To experimentally determine the critical residues:

  • Generate alanine scanning mutants across the Lp1.2 sequence

  • Create chimeric peptides combining segments from Lp1.2 and other alpha-conotoxins with known selectivity profiles

  • Perform computational docking studies to predict binding interface residues

  • Develop structure-activity relationship matrices based on electrophysiological characterization of mutants

The following matrix approach can guide systematic investigation:

PositionWild-type residueConservative mutationNon-conservative mutationExpected impact on selectivity
N-terminal-Extension/deletion-Minimal impact on binding
Loop 1XX to AX to chargedMajor impact on α7 binding
Loop 2YY to F/LY to D/EAffects muscle subtype specificity
C-terminalZDeletionAddition of RRRModifies neuronal vs. muscle selectivity

How does the three-dimensional structure of Lp1.2 contribute to its pharmacological properties?

The three-dimensional structure of alpha-conotoxins like Lp1.2 is critical to their function. Based on studies of homologous peptides, we can infer several structural features:

  • Global fold: Lp1.2 likely adopts a compact globular structure with two disulfide bridges creating a rigid framework. The characteristic "W-shaped" backbone conformation is stabilized by the Cys 3–Cys 9 and Cys 4–Cys 17 disulfide connectivity.

  • Surface electrostatics: The distribution of charged residues creates an electrostatic fingerprint that influences receptor subtype selectivity. Unlike many neuronal-selective alpha-conotoxins, Lp1.2 may possess a unique charge distribution if it shows the mixed activity profile observed in Lp1.4.

  • Loop conformations: The conformation of loops between conserved cysteines presents specific pharmacophore elements to receptor binding sites. NMR studies of related conotoxins suggest limited flexibility in these regions due to disulfide constraints .

To determine structure-function relationships:

  • Perform solution NMR spectroscopy to determine the 3D structure

  • Develop computational models based on homologous structures

  • Conduct molecular dynamics simulations to explore conformational flexibility

  • Map functional data from mutational studies onto the 3D structure

What strategies can enhance the stability of recombinant Lp1.2 for structural studies and therapeutic development?

Enhancing the stability of recombinant Lp1.2 is essential for both structural studies and potential therapeutic applications. Consider these methodological approaches:

  • Disulfide engineering:

    • Introduce non-native disulfide bridges to enhance thermal stability

    • Explore alternative disulfide connectivity patterns to identify more stable isomers

    • Consider selenocysteine substitution for enhanced oxidative stability

  • Backbone modifications:

    • N-methylation of specific amide bonds to reduce proteolytic susceptibility

    • Introduction of D-amino acids at vulnerable positions

    • Cyclization strategies to create head-to-tail cyclic variants

  • Formulation strategies:

    • Develop optimized buffer compositions to maximize long-term stability

    • Explore lyophilization with appropriate cryoprotectants

    • Investigate polymer encapsulation methods

  • Site-directed mutagenesis:

    • Replace oxidation-prone residues (Met, Trp) with more stable alternatives

    • Optimize surface charge distribution for reduced aggregation propensity

    • Introduce stabilizing salt bridges or hydrophobic interactions

Stability assessment should include:

  • Thermal denaturation studies using circular dichroism

  • Accelerated degradation testing under various pH and temperature conditions

  • Long-term storage studies with periodic activity testing

  • Freeze-thaw cycle tolerance evaluation

What analytical methods are most effective for characterizing the purity and structure of recombinant Lp1.2?

A comprehensive analytical strategy for recombinant Lp1.2 characterization should include:

  • Chromatographic methods:

    • Reversed-phase HPLC for purity assessment and isomer separation

    • Size-exclusion chromatography to detect aggregates

    • Ion-exchange chromatography to separate charge variants

    • Hydrophilic interaction chromatography (HILIC) for glycoform analysis if glycosylated

  • Mass spectrometry approaches:

    • ESI-MS for intact mass determination and confirmation of disulfide formation

    • MALDI-TOF for rapid screening and quality control

    • Tandem MS with enzymatic digestion for sequence confirmation

    • Top-down proteomics for comprehensive characterization

  • Spectroscopic techniques:

    • Circular dichroism for secondary structure assessment

    • NMR spectroscopy for tertiary structure determination

    • Fluorescence spectroscopy for conformational analysis

    • FTIR for complementary secondary structure information

  • Functional characterization:

    • Competitive binding assays

    • Electrophysiological measurements

    • Cell-based functional assays

For disulfide mapping, this methodology is recommended:

  • Partial reduction using TCEP at controlled concentrations

  • Alkylation of free thiols with iodoacetamide or NEM

  • Enzymatic digestion with specific proteases

  • LC-MS/MS analysis of resulting fragments

  • Connectivity assignment based on mass shifts

How can I differentiate between correctly folded Lp1.2 and misfolded isomers?

Differentiating correctly folded Lp1.2 from misfolded isomers requires a multi-technique approach:

  • Chromatographic discrimination:

    • RP-HPLC typically separates different disulfide isomers based on subtle hydrophobicity differences

    • Use reference standards from directed synthesis if available

    • Co-elution studies with synthetic standards of known connectivity

  • Functional assessment:

    • Perform electrophysiological characterization on nAChR subtypes

    • Only correctly folded peptides will exhibit the expected activity profile

    • Compare IC50 values with literature data for related conotoxins

  • Structural comparison:

    • Circular dichroism can rapidly identify gross structural differences

    • NMR fingerprinting can detect conformational heterogeneity

    • Disulfide connectivity mapping using partial reduction and MS

  • Stability assessment:

    • Thermal denaturation profiles often differ between correctly folded and misfolded isomers

    • Susceptibility to proteolytic degradation typically higher in misfolded variants

    • Chemical denaturation curves using urea or guanidinium hydrochloride

The following decision tree approach can guide isomer identification:

  • First level: RP-HPLC retention time comparison with standards

  • Second level: Mass confirmation by ESI-MS

  • Third level: Bioactivity screening on relevant nAChR subtypes

  • Fourth level: Structural characterization by CD and/or NMR

  • Final confirmation: Disulfide mapping by MS

What are the best approaches to evaluate Lp1.2 binding kinetics and affinity for different nicotinic acetylcholine receptor subtypes?

Comprehensive evaluation of Lp1.2 binding kinetics and affinity requires multiple complementary techniques:

  • Two-electrode voltage clamp electrophysiology:

    • Provides functional IC50 values and on/off rates

    • Can distinguish between competitive and non-competitive mechanisms

    • Allows analysis of use-dependence and state-dependence of inhibition

  • Radioligand binding assays:

    • Direct measurement of binding constants (Kd, Ki)

    • Saturation binding to determine Bmax

    • Competition binding against known ligands

    • Association/dissociation kinetics studies

  • Surface plasmon resonance (SPR):

    • Label-free real-time measurement of binding kinetics

    • Determination of kon and koff rates

    • Detection of complex binding modes and conformational changes

    • Requires immobilization of purified receptor or receptor fragments

  • Isothermal titration calorimetry (ITC):

    • Direct measurement of binding thermodynamics

    • Provides enthalpy and entropy contributions

    • May reveal binding stoichiometry

    • No immobilization or labeling required

For comprehensive receptor subtype profiling, this panel is recommended:

nAChR SubtypeExperimental SystemRecommended TechniqueExpected Potency Range
α7Xenopus oocytesTEVC1-10 μM (estimate)
α3β2Xenopus oocytesTEVC1-50 μM (estimate)
α3β4Xenopus oocytesTEVC10-100 μM (estimate)
α6α3β2Xenopus oocytesTEVC1-50 μM (estimate)
α1β1γδXenopus oocytesTEVC0.1-10 μM (estimate)
α1β1εδXenopus oocytesTEVC1-50 μM (estimate)

What preclinical models are appropriate for evaluating Lp1.2 as a potential analgesic agent?

Based on the activity profiles of related alpha-conotoxins, several preclinical models may be appropriate for evaluating Lp1.2's potential analgesic properties:

  • Inflammatory pain models:

    • Complete Freund's adjuvant (CFA)-induced hyperalgesia

    • Carrageenan-induced inflammatory pain

    • Formalin test for both neurogenic and inflammatory pain phases

  • Neuropathic pain models:

    • Chronic constriction injury (CCI)

    • Spared nerve injury (SNI)

    • Chemotherapy-induced peripheral neuropathy (CIPN)

  • Visceral pain models:

    • Acetic acid-induced writhing

    • Colorectal distension

    • Mustard oil-induced visceral hyperalgesia

The methodological approach should include:

  • Dose-response studies with both central and peripheral administration

  • Comparison with standard analgesics (morphine, gabapentin)

  • Assessment of motor function to distinguish analgesia from motor impairment

  • Pharmacokinetic studies to determine CNS penetration and tissue distribution

If Lp1.2 shows α7 nAChR activity (like Mr1.1), it may suppress inflammatory response to pain in vivo, suggesting potential applications in conditions with neuroinflammatory components .

How can the blood-brain barrier penetration of Lp1.2 be assessed and potentially enhanced?

Alpha-conotoxins like Lp1.2 face challenges in blood-brain barrier (BBB) penetration due to their size and hydrophilicity. To assess and enhance BBB penetration:

  • BBB penetration assessment methods:

    • In vitro BBB models using polarized endothelial cell cultures

    • Radioisotope or fluorophore labeling for in vivo tracking

    • Cerebrospinal fluid sampling after systemic administration

    • Brain microdialysis for direct measurement of brain penetration

  • Chemical modification strategies:

    • Addition of lipophilic moieties to increase passive diffusion

    • Glycosylation to potentially access glucose transporters

    • Reduced disulfide bonds with stable thioether replacements

    • N-methylation of selected backbone positions

  • Drug delivery approaches:

    • Conjugation to cell-penetrating peptides (CPPs) like TAT or Penetratin

    • Encapsulation in nanoparticles or liposomes

    • Antibody-based delivery using transferrin receptor targeting

    • Intranasal delivery to bypass BBB via olfactory pathways

  • Pharmacokinetic considerations:

    • Stability studies in serum and cerebrospinal fluid

    • Characterization of plasma protein binding

    • Assessment of efflux transporter susceptibility (P-glycoprotein)

    • Half-life determination and clearance mechanisms

Success in BBB penetration will significantly expand the therapeutic potential of Lp1.2 for centrally-mediated conditions.

What are the potential challenges in translating Lp1.2 from preclinical studies to clinical applications?

Translating recombinant Lp1.2 from bench to bedside involves addressing several challenges:

  • Pharmaceutical development challenges:

    • Large-scale GMP production of correctly folded peptide

    • Formulation stability for long-term storage

    • Development of suitable administration routes

    • Bioavailability and tissue distribution optimization

  • Regulatory considerations:

    • Documentation of impurity profiles

    • Establishment of release specifications

    • Immunogenicity assessment

    • Development of appropriate bioanalytical methods

  • Pharmacological challenges:

    • Narrow therapeutic window given the endogenous role of nAChRs

    • Potential off-target effects on non-target receptor subtypes

    • Species differences in receptor pharmacology

    • Target engagement biomarkers for clinical studies

  • Clinical development strategy:

    • Patient population selection based on mechanism of action

    • Dose selection from preclinical PK/PD modeling

    • Definition of appropriate clinical endpoints

    • Biomarker strategy for proof-of-mechanism studies

Mitigation strategies include:

  • Comprehensive receptor profiling across human and animal receptors

  • Detailed toxicology studies addressing potential cholinergic adverse effects

  • Rational modification to improve therapeutic index

  • Development of companion diagnostics to identify responsive patient populations

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.