Recombinant Bungarus caeruleus Phospholipase A2 isoform 1

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: Standard shipping includes blue ice packs. Dry ice shipping requires advance notice 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 settle the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50%, which may serve as a guideline for your application.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and the protein's inherent 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. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The specific tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its incorporation.
Synonyms
Acidic phospholipase A2 1; svPLA2; EC 3.1.1.4; Phosphatidylcholine 2-acylhydrolase; Fragment
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
20-137
Protein Length
Full Length of Mature Protein
Purity
>85% (SDS-PAGE)
Species
Bungarus caeruleus (Indian krait)
Target Protein Sequence
N LYQLMNMIQC ANTRTWPSYT NYGCYCGKGG SGTPVDDLDR CCYTHDHCYN DAKNIDGCNP VTKTYSYTCT EPTITCNDSK DKCARFVCDC DRTAAICFAK APYNTSNVMI RSTNSCQ
Uniprot No.

Target Background

Function

Snake venom phospholipase A2 (PLA2) exhibits anticoagulant and neurotoxic activities. PLA2 catalyzes the calcium-dependent hydrolysis of the 2-acyl groups in 3-sn-phosphoglycerides.

Protein Families
Phospholipase A2 family, Group I subfamily, D49 sub-subfamily
Subcellular Location
Secreted.
Tissue Specificity
Expressed by the venom gland.

Q&A

What is the primary structure of Phospholipase A2 isoform 1 from Bungarus caeruleus?

Phospholipase A2 (PLA2) isoform 1 from Bungarus caeruleus (common Indian krait) is characterized as a single chain polypeptide of approximately 118-125 amino acid residues with a molecular mass of 13-15 kDa. The protein belongs to Group I snake venom PLA2s, which feature seven disulfide bonds that are crucial for structural stability. The conserved catalytic site includes His-48, Asp-49, Tyr-28, Gly-30, Gly-32, and Asp-99 residues. These residues form the calcium-binding loop essential for enzymatic activity. Key structural elements include three long α-helices and two short antiparallel β-strands, which are typical of Group I PLA2 enzymes .

What is the mechanism of catalysis for PLA2 isoform 1?

PLA2 isoform 1 catalyzes the hydrolysis of membrane glycerophospholipids at the sn-2 position through a well-defined mechanism involving:

  • Substrate binding: The substrate binds in a hydrophobic cleft surrounded by lipophilic residues.

  • Calcium coordination: Ca²⁺ ions, coordinated by Asp-49 and other residues in the calcium-binding loop, stabilize the substrate positioning and the tetrahedral intermediate.

  • Nucleophilic attack: A water molecule activated by His-48 attacks the ester bond.

  • Tetrahedral intermediate: Formation of a tetrahedral intermediate stabilized by the Ca²⁺ ion.

  • Bond cleavage: The intermediate collapses, releasing free fatty acid and lysophospholipid.

Two mechanisms have been proposed based on crystallographic and biochemical studies: the "single-water mechanism" and the "assisted-water mechanism." Both emphasize the critical roles of His-48 as a base catalyst and Asp-49 for calcium coordination .

What are the optimal conditions for recombinant expression of B. caeruleus PLA2 isoform 1 in E. coli?

Based on successful expression strategies for similar snake venom PLA2s, the optimal conditions for recombinant expression of B. caeruleus PLA2 isoform 1 in E. coli include:

  • Expression system: BL21(DE3) strain transformed with a pET vector containing a 6xHis-tag fusion to facilitate purification.

  • Induction parameters: 0.5-1.0 mM IPTG at OD₆₀₀ of 0.6-0.8, with post-induction growth at 25-30°C for 4-6 hours (rather than 37°C) to minimize inclusion body formation.

  • Media optimization: Use of enriched media such as TB (Terrific Broth) supplemented with 1% glucose to repress basal expression.

  • Codon optimization: Adaptation of the cDNA sequence to E. coli codon usage preferences, especially important for rare codons.

The protein typically expresses as inclusion bodies, necessitating denaturation and refolding steps. The process should include careful optimization of disulfide bond formation during refolding, which is critical for obtaining functionally active enzyme .

What is the most effective refolding protocol for recombinant B. caeruleus PLA2 from inclusion bodies?

The most effective refolding protocol for recombinant B. caeruleus PLA2 from inclusion bodies involves:

  • Isolation and washing: Thorough washing of inclusion bodies with detergent-containing buffer (typically 0.5% Triton X-100) to remove membrane contaminants.

  • Solubilization: Complete denaturation using 6-8 M urea or 6 M guanidine hydrochloride, with 10 mM DTT to reduce disulfide bonds.

  • Refolding procedure: Gradual dilution into refolding buffer containing:

    • 50 mM Tris-HCl, pH 8.5

    • 0.5 M L-arginine (prevents aggregation)

    • 5 mM reduced glutathione and 0.5 mM oxidized glutathione (facilitates disulfide shuffling)

    • 5 mM CaCl₂ (stabilizes structure)

    • 1 mM EDTA (prevents metal-catalyzed oxidation)

  • Concentration and purification: Using tangential flow filtration followed by affinity chromatography and gel filtration.

Critical parameters include protein concentration during refolding (optimally 0.1-0.5 mg/ml), temperature (typically 4°C), and slow dilution rate to prevent aggregation. The refolded protein requires validation through enzymatic activity assays and circular dichroism analysis to confirm proper folding .

How can I verify the enzymatic activity of purified recombinant PLA2 isoform 1?

Verification of recombinant PLA2 enzymatic activity should employ multiple complementary approaches:

  • Fluorescent assay: Using 1-hexadecanoyl-2-(1-pyrene-decanoyl)-sn-glycero-3-phosphoglycerol as substrate. In a 96-well black plate, combine 50 μL of diluted enzyme (0.02 ng/μL) with 50 μL of substrate (100 μM in substrate buffer). Measure fluorescence at excitation and emission wavelengths of 345 nm and 395 nm in kinetic mode for 5 minutes .

  • Colorimetric assay: Using diheptanoyl phosphatidylcholine with subsequent detection of released fatty acids via ADIFAB or pH-sensitive indicators.

  • Specific activity calculation:

    Specific Activity (pmol/min/μg) = [Adjusted Vmax (RFU/min) × Conversion Factor (pmol/RFU)] / amount of enzyme (μg)

  • Comparative analysis: Always include positive control (native enzyme) and negative control (buffer only) to establish relative activity levels.

The expected specific activity for properly folded PLA2 should be >2,500 pmol/min/μg under standard assay conditions .

How can I determine the oligomeric state of recombinant B. caeruleus PLA2 isoform 1?

Determination of the oligomeric state of recombinant PLA2 requires multiple biophysical approaches:

  • Dynamic Light Scattering (DLS): Prepare samples in 50 mM Tris-HCl buffer (pH 8.0) at protein concentrations of 5-15 mg/ml. Filter through 0.1 μm polyvinylidene difluoride filters before analysis. Use a 30 mW, 660 nm laser diode at 303 K, collecting data in quintuplicate. This technique provides information about the hydrodynamic radius and size distribution in solution .

  • Size Exclusion Chromatography (SEC): Use Superdex 75 or equivalent column calibrated with appropriate molecular weight standards. Analyze the elution profile to determine the apparent molecular weight compared to standards.

  • Analytical Ultracentrifugation (AUC): Both sedimentation velocity and equilibrium experiments provide definitive data on the molecular weight and shape of the protein in solution.

  • Native PAGE: Compare migration patterns against known monomeric, dimeric, and trimeric PLA2 standards.

  • Cross-linking studies: Using chemical cross-linkers like glutaraldehyde or BS3 (bis(sulfosuccinimidyl)suberate) to capture transient oligomeric states.

Different isoforms of Bungarus PLA2s have been observed to form stable trimers or other oligomeric states. For example, a trimeric PLA2 from B. caeruleus showed approximately 12% of its total solvent-accessible surface area buried in the core of the trimer, with extensive water-mediated interactions between subunits .

What crystallization conditions are optimal for structural studies of recombinant B. caeruleus PLA2 isoform 1?

Based on successful crystallization of various Bungarus PLA2 isoforms, the following conditions are recommended:

  • Protein preparation: Purify protein to >95% homogeneity and concentrate to 10-15 mg/ml in 20 mM Tris-HCl, pH 7.5, 50 mM NaCl.

  • Initial screening: Employ commercial sparse matrix screens (e.g., Hampton Research Crystal Screens 1 and 2, Molecular Dimensions MemGold) using sitting-drop vapor diffusion method.

  • Optimized conditions: Based on successful conditions for similar proteins:

    • Precipitant: 2.5-3.0 M NaCl

    • Buffer: 0.1 M sodium acetate or Tris-HCl, pH 7.0-8.5

    • Additives: 5-10 mM CaCl₂ (stabilizes calcium-binding loop)

    • Temperature: 293 K (20°C)

    • Drop size: 1-2 μl protein + 1-2 μl reservoir solution

  • Setup: Use sitting-drop vapor diffusion in 24-well plates with 500 μl reservoir solution.

  • Crystal appearance: Crystals typically appear within 1-2 weeks and reach maximum size in 3-4 weeks.

  • Cryoprotection: For X-ray data collection, crystals should be transferred to a cryoprotectant solution containing the reservoir solution supplemented with 20-25% glycerol or ethylene glycol before flash-cooling in liquid nitrogen .

How do I interpret structural data to understand catalytic site variations between active and inactive PLA2 isoforms?

Interpreting structural data to understand catalytic site variations requires systematic analysis of key structural elements:

FeatureActive PLA2Inactive PLA2 (e.g., His-Ala47 mutant)
Ca²⁺ bindingProper coordination through Asp49Impaired or absent
His-48 positioningOptimally oriented for catalysisMay be displaced
Active site waterProperly positioned for nucleophilic attackDisplaced or absent
Substrate channelWell-formed hydrophobic channelMay be distorted
Secondary structureCanonical α-helices and β-strandsGenerally preserved

How does the enzymatic activity of recombinant B. caeruleus PLA2 isoform 1 compare to the native enzyme?

The comparison between recombinant and native B. caeruleus PLA2 isoform 1 requires comprehensive biochemical characterization:

  • Enzymatic parameters: Determine and compare kinetic parameters (Km, kcat, Vmax) using standardized assay conditions. Typically, properly folded recombinant PLA2 shows 70-90% of the specific activity of the native enzyme, with minor variations in substrate affinity.

  • Calcium dependence: Analyze enzymatic activity across a calcium concentration range (0-10 mM). Both enzymes should demonstrate absolute calcium dependence with similar EC50 values, typically in the micromolar range.

  • pH profile: Compare activity across pH 5-10, where both enzymes should exhibit a bell-shaped curve with optimal activity around pH 7.5-8.5.

  • Thermal stability: Assess activity after incubation at temperatures ranging from 4-60°C. Properly folded recombinant enzyme may show slightly reduced thermal stability compared to the native form.

  • Substrate specificity: Test activity against various phospholipids with different head groups and fatty acid compositions.

Any significant deviations in these parameters may indicate incorrect folding, incomplete post-translational modifications, or artifacts introduced by the recombinant expression system. The presence of the His-tag may also slightly alter the biochemical properties, which can be assessed by comparing the tagged protein with one where the tag has been removed .

What approaches can differentiate neurotoxic from non-neurotoxic PLA2 isoforms of B. caeruleus?

Differentiating neurotoxic from non-neurotoxic PLA2 isoforms requires a multi-faceted approach:

  • Ex vivo neuromuscular junction assays: Use isolated phrenic nerve-hemidiaphragm preparations from mice or rats. Neurotoxic PLA2s produce concentration-dependent inhibition of nerve-evoked muscle twitches. Parameters to measure include:

    • Time to 50% blockade (t50)

    • Concentration producing 50% blockade (EC50)

    • Reversibility following washout

  • Electrophysiological characterization:

    • Intracellular recordings to distinguish pre- vs post-synaptic effects

    • Miniature end-plate potential (MEPP) frequency analysis

    • End-plate potential (EPP) quantal content analysis

  • Receptor binding studies: Using radiolabeled or fluorescently labeled PLA2s to quantify binding to synaptic preparations. Neurotoxic PLA2s often show higher affinity for neuronal membranes.

  • Structural correlates: Analyze surface electrostatic potential and hydrophobic patches which may correlate with neurotoxicity. Neurotoxic PLA2s typically feature specific surface regions for target recognition distinct from the catalytic site.

  • Enzymatic activity correlation: Some neurotoxic PLA2s maintain their toxicity even when catalytically inactive, suggesting toxicity mechanisms independent of phospholipid hydrolysis .

How can I design site-directed mutagenesis experiments to understand structure-function relationships in B. caeruleus PLA2 isoform 1?

Strategic site-directed mutagenesis can elucidate structure-function relationships in PLA2 isoform 1:

  • Catalytic site mutations:

    • His48Gln: Expected to abolish catalytic activity while maintaining structural integrity

    • Asp49Ala/Asn/Lys: Different substitutions disrupt calcium binding to varying degrees

    • Tyr28Phe: Tests the role of the hydroxyl group in substrate positioning

  • Calcium-binding loop mutations:

    • Gly30Ser/Ala: Affects flexibility of the calcium-binding loop

    • Gly32Ala: Tests the importance of backbone conformational freedom

  • Interfacial binding surface mutations:

    • Replace surface hydrophobic residues that interact with membranes to alter substrate access

    • Modify charged residues at the putative membrane-binding interface

  • Oligomerization interface mutations:

    • Target residues involved in potential trimer formation (based on structural homology with known trimeric PLA2s)

    • Example: Modify Arg, Lys and Thr residues involved in water-mediated interactions across oligomeric interfaces

  • Experimental validation:

    • Structural analysis: Circular dichroism and thermal denaturation to confirm structural integrity

    • Enzymatic assays: Determine kinetic parameters for each mutant

    • Binding studies: Assess membrane/substrate binding using fluorescence techniques

    • Oligomerization analysis: SEC, DLS, and AUC to determine effects on quaternary structure

This systematic approach allows mapping of residues critical for activity, binding, and structural stability, with clear distinction between those affecting catalysis versus those involved in other functions .

How do I design experiments to compare B. caeruleus PLA2 isoform 1 with other Group I PLA2 enzymes?

Design comprehensive comparative analyses between B. caeruleus PLA2 isoform 1 and other Group I PLA2s using the following methodological approach:

  • Sequence alignment and phylogenetic analysis:

    • Perform multiple sequence alignment (using MUSCLE or CLUSTALW)

    • Calculate sequence identity and similarity percentages

    • Construct phylogenetic trees (Maximum Likelihood or Bayesian methods)

    • Map conserved and variable regions onto the alignment

  • Structural comparison:

    • Superimpose 3D structures (if available) using PyMOL or UCSF Chimera

    • Calculate RMSD values for backbone atoms

    • Compare secondary structure elements and loop regions

    • Analyze electrostatic surface potential differences

  • Functional comparison:

    • Standardized enzymatic assays under identical conditions

    • Side-by-side testing of substrate specificity profiles

    • Comparative calcium dependence and pH optima determination

    • Thermal stability and denaturation profiles

  • Membrane interaction studies:

    • Liposome binding assays with varied phospholipid compositions

    • Surface plasmon resonance to measure binding kinetics

    • Interfacial activation profiles on different substrate presentations

  • Pharmacological profiling:

    • Comparative testing in standardized bioassays (e.g., neuromuscular preparations)

    • Cross-reactivity with antibodies or inhibitors

    • Tissue and cell-type specificity evaluation

This comprehensive approach enables identification of unique structural and functional features of B. caeruleus PLA2 isoform 1 relative to other Group I enzymes .

What are the most promising applications of recombinant B. caeruleus PLA2 isoform 1 in neuroscience research?

Recombinant B. caeruleus PLA2 isoform 1 offers several valuable applications in neuroscience research:

  • Investigation of presynaptic neurotransmission mechanisms:

    • Selective modulation of neurotransmitter release machinery

    • Study of voltage-gated calcium channel functions

    • Analysis of vesicle recycling pathways

    • Synaptic vesicle-plasma membrane fusion mechanisms

  • Development of novel neuronal tracing tools:

    • Creation of fluorescently-tagged recombinant PLA2 variants

    • Using catalytically inactive mutants as specific molecular probes for neuronal membranes

    • Exploiting retrograde transport properties for neural circuit mapping

  • Neurodegenerative disease research:

    • Investigation of lipid metabolism in neuronal membranes

    • Studies on phospholipid dynamics in synaptic plasticity

    • Analysis of membrane remodeling in health and disease

  • Therapeutic target identification:

    • Screening platforms for novel PLA2 inhibitors with neuroprotective properties

    • Structure-based drug design approaches

    • Development of PLA2-neutralizing antibodies

  • Electrophysiological tools:

    • Selective modulation of neuromuscular junction transmission

    • Creation of precisely timed and localized synaptic blockades

    • Molecular dissection of different components of synaptic transmission

The unique structural features and pharmacological properties of this PLA2 make it particularly valuable for studying presynaptic mechanisms involving membrane phospholipids .

How can I use recombinant B. caeruleus PLA2 isoform 1 to develop novel enzyme inhibitors?

Developing novel enzyme inhibitors using recombinant B. caeruleus PLA2 isoform 1 involves a systematic approach:

  • High-throughput screening platform:

    • Optimize a fluorescence-based assay for 384-well format

    • Establish Z' factor >0.7 for assay robustness

    • Screen compound libraries at 10-20 μM concentration

    • Include appropriate positive controls (known PLA2 inhibitors)

  • Rational design using structural information:

    • Analyze the catalytic site and substrate-binding pocket

    • Target the critical His-48/Asp-99 catalytic dyad

    • Design compounds to compete with calcium binding

    • Exploit unique features of the hydrophobic channel

  • Structure-activity relationship studies:

    • Generate focused libraries based on initial hits

    • Test modifications to improve potency and selectivity

    • Establish clear SAR patterns to guide optimization

  • Binding validation methods:

    • Isothermal titration calorimetry to determine binding thermodynamics

    • Surface plasmon resonance for binding kinetics

    • Co-crystallization or soaking experiments for structural validation

    • NMR-based approaches for fragment screening

  • Selectivity profiling:

    • Test inhibitors against panel of PLA2s from different sources

    • Develop isoform-selective inhibitors based on unique structural features

    • Assess activity against mammalian PLA2s to minimize off-target effects

  • In silico approaches:

    • Molecular docking and virtual screening

    • Molecular dynamics simulations of enzyme-inhibitor complexes

    • Pharmacophore modeling based on known inhibitors

This structured approach enables development of potent and selective inhibitors with potential applications in both research and therapeutic contexts .

What are common challenges in recombinant expression of B. caeruleus PLA2 isoform 1 and how can they be addressed?

Researchers commonly encounter several challenges when expressing recombinant B. caeruleus PLA2 isoform 1:

  • Low expression yields:

    • Problem: Toxic effects on host cells due to phospholipase activity

    • Solution: Use tightly controlled expression systems (e.g., pET with T7 lysozyme co-expression) or express as fusion with solubilizing partners (SUMO, TRX, MBP)

  • Inclusion body formation:

    • Problem: Improper disulfide bond formation leading to aggregation

    • Solution: Lower induction temperature (16-20°C), reduce IPTG concentration (0.1-0.2 mM), co-express with chaperones (GroEL/ES, DsbC)

  • Incorrect folding:

    • Problem: Multiple disulfide bonds forming incorrectly during refolding

    • Solution: Optimize GSH/GSSG ratios, employ redox shuffling systems, use pulse refolding with sequential dilution

  • Proteolytic degradation:

    • Problem: Susceptibility to host proteases

    • Solution: Use protease-deficient strains (BL21(DE3) pLysS), include protease inhibitors, express with N- and C-terminal stabilizing domains

  • Toxicity management:

    • Problem: Phospholipase activity damaging host cells

    • Solution: Express catalytically inactive mutants (H48Q) or design constructs with removable pro-sequence to prevent activity during expression

  • Refolding efficiency:

    • Problem: Low yield of correctly folded protein after denaturation/refolding

    • Solution: Screen additives (L-arginine, glycerol, sucrose), try on-column refolding, employ step-wise dialysis with decreasing denaturant

ChallengeStandard ApproachOptimized Solution
Low yield0.5 mM IPTG, 37°C0.1 mM IPTG, 16°C, 18-24h induction
Inclusion bodiesStandard refoldingPulse refolding with 0.5M L-arginine
MisfoldingSingle buffer conditionsMatrix screening of pH, redox conditions
Low activitySimple refoldingAddition of phospholipid micelles during refolding
AggregationDirect dilutionStep-wise dialysis with stabilizing additives

Implementing these solutions can significantly improve the yield and quality of recombinant PLA2 .

How can I distinguish between catalytic and non-catalytic effects when studying B. caeruleus PLA2 isoform 1?

Distinguishing between catalytic and non-catalytic effects requires careful experimental design:

  • Site-directed mutagenesis approach:

    • Generate H48Q mutant (catalytically inactive but structurally intact)

    • Create D49K mutant (disrupts calcium binding and catalysis)

    • Produce Y28F mutant (reduced but not abolished activity)

    • Compare biological effects of these variants with wild-type enzyme

  • Chemical modification strategy:

    • Use p-bromophenacyl bromide (p-BPB) for selective modification of His-48

    • Employ EDTA/EGTA for calcium chelation to abolish activity

    • Apply reversible inhibitors to temporarily block catalytic activity

  • Experimental paradigms:

    • Direct comparison of wild-type and catalytically inactive mutants

    • Pre-treatment with specific PLA2 inhibitors before biological assays

    • Heat-inactivation of enzyme as negative control

    • Rescue experiments with addition of lysophospholipids and fatty acids

  • Binding vs. catalysis differentiation:

    • Fluorescence-based binding assays to assess membrane interaction

    • Surface plasmon resonance to quantify binding without catalysis

    • Co-localization studies using labeled inactive enzyme

  • Temporal analysis:

    • Rapid effects are often non-catalytic (binding-mediated)

    • Delayed effects may involve catalytic activity and downstream lipid mediators

    • Time-course studies with selective inhibition at different time points

These approaches help distinguish direct binding effects (often rapid and calcium-independent) from catalytic effects (typically calcium-dependent and linked to lipid mediator production) .

What are the most sensitive methods for detecting structural changes in PLA2 following site-directed mutagenesis?

Detecting subtle structural changes in PLA2 mutants requires combining multiple high-sensitivity biophysical techniques:

  • High-resolution spectroscopic methods:

    • Circular Dichroism (CD): Far-UV (190-260 nm) for secondary structure; near-UV (250-350 nm) for tertiary structure environment of aromatic residues

    • Fourier Transform Infrared Spectroscopy (FTIR): Complementary to CD for secondary structure analysis, particularly for β-sheet content

    • Nuclear Magnetic Resonance (NMR): 2D HSQC spectra to monitor chemical shift perturbations indicating structural changes; requires ¹⁵N-labeled protein

  • Fluorescence-based techniques:

    • Intrinsic tryptophan fluorescence: Monitors changes in microenvironment of Trp residues

    • 8-Anilino-1-naphthalenesulfonic acid (ANS) binding: Detects exposure of hydrophobic patches

    • Time-resolved fluorescence: Measures fluorescence lifetime changes indicating altered dynamics

  • Thermal stability assessment:

    • Differential Scanning Calorimetry (DSC): Provides thermodynamic parameters of unfolding

    • Thermal shift assays: Using SYPRO Orange or similar dyes in a real-time PCR instrument

    • CD thermal melting curves: Monitoring secondary structure changes during thermal denaturation

  • Hydrodynamic methods:

    • Analytical Ultracentrifugation (AUC): Detects changes in shape and oligomeric state

    • Size Exclusion Chromatography with Multi-Angle Light Scattering (SEC-MALS): Accurate molecular weight and shape determination

    • Small-Angle X-ray Scattering (SAXS): Low-resolution shape information in solution

  • Hydrogen-deuterium exchange mass spectrometry (HDX-MS):

    • Maps regions of altered solvent accessibility and dynamics

    • Provides peptide-level resolution of structural changes

    • Particularly powerful for detecting allosteric effects distant from mutation sites

Combining these methods provides comprehensive characterization of structural perturbations, from local changes at the mutation site to global effects on protein conformation and dynamics .

How can B. caeruleus PLA2 isoform 1 be used to develop better antivenoms?

Development of improved antivenoms using recombinant B. caeruleus PLA2 isoform 1 involves several methodological approaches:

  • Epitope mapping strategy:

    • Express overlapping peptide fragments covering the complete PLA2 sequence

    • Identify immunodominant epitopes using existing antivenom binding assays

    • Map epitopes onto 3D structure to identify surface-exposed regions

    • Design multi-epitope constructs focusing on neutralizing epitopes

  • Rational immunogen design:

    • Generate catalytically inactive mutants (H48Q) that maintain structural epitopes

    • Create chimeric constructs containing epitopes from multiple PLA2 isoforms

    • Develop consensus sequences covering epitope variations across isoforms

    • Express structurally stabilized variants with enhanced immunogenicity

  • Immunization protocols:

    • Compare different adjuvant formulations for optimal antibody response

    • Establish prime-boost strategies with different construct combinations

    • Monitor antibody titers and neutralizing capacity against native toxins

    • Assess cross-reactivity with related PLA2s from other venomous species

  • In vitro neutralization assessment:

    • Develop standardized enzymatic neutralization assays

    • Establish ex vivo neurotoxicity neutralization models

    • Quantify neutralizing potency using dose-response curves

    • Compare with existing antivenoms for enhanced coverage

  • Monoclonal antibody development:

    • Generate monoclonal antibodies against critical epitopes

    • Select based on neutralizing activity rather than just binding

    • Create antibody cocktails targeting multiple epitopes

    • Engineer antibodies for improved stability and reduced immunogenicity

This approach addresses the limitations of current antivenom production, which typically uses crude venom and produces antibodies against many non-toxic components .

What novel experimental approaches can reveal the molecular mechanisms of PLA2 neurotoxicity?

Investigating PLA2 neurotoxicity mechanisms requires advanced experimental approaches:

  • High-resolution imaging of neuromuscular junctions:

    • Super-resolution microscopy (STORM/PALM) with fluorescently labeled PLA2

    • Real-time tracking of PLA2 binding and internalization

    • Correlative light and electron microscopy to visualize ultrastructural changes

    • Multi-color imaging to track co-localization with potential targets

  • Molecular target identification:

    • Chemical cross-linking followed by mass spectrometry

    • Proximity labeling approaches (BioID, APEX) to identify interacting proteins

    • Pull-down assays with catalytically inactive mutants

    • Genome-wide CRISPR screens for toxicity modulators

  • Advanced electrophysiological approaches:

    • Patch-clamp recordings of presynaptic terminals

    • Optical measurements of calcium dynamics using genetically encoded indicators

    • Simultaneous pre- and post-synaptic recordings

    • High-throughput multi-electrode array recordings

  • Single vesicle fusion assays:

    • Reconstituted systems with defined lipid compositions

    • TIRF microscopy to monitor individual fusion events

    • Real-time monitoring of lipid remodeling

    • Direct visualization of PLA2 effects on fusion kinetics

  • Lipidomic analysis:

    • Targeted and untargeted mass spectrometry of synaptic membranes

    • Spatial lipidomics using imaging mass spectrometry

    • Temporal profiling of lipid changes after PLA2 exposure

    • Correlation of specific lipid alterations with functional deficits

These approaches can identify precise molecular targets and distinguish between direct binding effects and consequences of phospholipid hydrolysis .

How can structural information from B. caeruleus PLA2 isoform 1 contribute to understanding evolutionary relationships among snake venom PLA2s?

Structural analysis of B. caeruleus PLA2 provides key insights into evolutionary relationships through several methodological approaches:

  • Integrated phylogenetic analysis:

    • Combine primary sequence data with structural information

    • Weight conserved structural elements in phylogenetic reconstructions

    • Identify structurally constrained regions versus variable regions

    • Create structure-guided sequence alignments for improved accuracy

  • Structure-based clustering:

    • Compare 3D structures using distance matrix analysis

    • Measure root-mean-square deviation (RMSD) between backbone atoms

    • Generate structure-based dendrograms

    • Identify structural synapomorphies (shared derived features)

  • Functional surface analysis:

    • Map electrostatic potential on molecular surfaces

    • Compare hydrophobic patches and binding interfaces

    • Analyze active site geometry across evolutionary lineages

    • Identify convergent and divergent functional adaptations

  • Quaternary structure comparison:

    • Analyze oligomerization patterns (monomers, dimers, trimers)

    • Compare interface residues between oligomeric forms

    • Identify evolutionary transitions between oligomeric states

    • Assess stability of oligomeric interfaces across species

  • Molecular dynamics simulations:

    • Compare dynamic properties across evolutionary lineages

    • Identify conserved motion patterns versus lineage-specific dynamics

    • Simulate ancestral sequences in structural contexts

    • Predict evolutionary constraints on protein flexibility

This integrated approach reveals how structural constraints and functional diversification have shaped PLA2 evolution, providing insights beyond what sequence analysis alone can offer. The structural data can identify cases of convergent evolution and help distinguish between phylogenetic signal and homoplasy in sequence alignments .

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.