Lysenin-related protein 2 Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Characteristics and Development

The Lysenin-related protein 2 antibody (e.g., PA5-144514) is a polyclonal antibody raised in rabbits, designed for research applications such as Western blot (WB), immunohistochemistry (IHC), and enzyme-linked immunosorbent assay (ELISA). Key features include:

ParameterDetails
Host SpeciesRabbit
ClonalityPolyclonal
Target ProteinLysenin-related protein 2 (UniProt ID: Q9U2S1)
ApplicationsWB, IHC, ELISA
Biological ActivityDetects LRP-2’s sphingomyelin-binding and hemolytic functions
Research UseExclusively for experimental studies; not for diagnostics or commercial use

This antibody enables precise detection of LRP-2 in cellular and biochemical assays, facilitating studies on its pore-forming mechanisms and pathogen interactions .

Sphingomyelin Binding and Hemolysis

LRP-2 binds sphingomyelin (SM) with high specificity, similar to lysenin, and induces hemolysis by forming oligomeric pores in erythrocyte membranes. Antibody-based assays (e.g., Western blot) confirmed that LRP-2’s activity is 10-fold stronger than its homolog LRP-1, attributed to conserved aromatic residues like phenylalanine 210 . Mutagenesis studies using antibodies revealed that tryptophan residues in LRP-2 are essential for SM recognition and pore stability .

Antibacterial Activity

The antibody has been employed to validate LRP-2’s antibacterial effects against Bacillus megaterium, suggesting its role in earthworm innate immunity . This aligns with lysenin family proteins’ broader function in combating microbial pathogens .

Oligomerization and Membrane Insertion

Antibody labeling experiments demonstrated that LRP-2 forms SDS-resistant oligomers upon binding to SM-rich membranes. Cryo-EM and AFM studies, supported by antibody validation, revealed a prepore-to-pore transition mechanism involving pH-dependent conformational changes . These oligomers create 3 nm pores, disrupting membrane integrity and ion homeostasis .

Role of Aromatic Residues

Systematic mutagenesis coupled with antibody detection highlighted the necessity of conserved tryptophan residues (e.g., Trp-20, Trp-112) for LRP-2’s pore-forming activity. Substitution of phenylalanine 210 in LRP-1 with isoleucine reduced hemolytic efficacy, underscoring structural nuances detectable via antibody assays .

Biosensor Development

LRP-2’s pore-forming properties, validated through antibody-assisted studies, enable its use in stochastic sensing platforms. These channels detect ions, peptides, and DNA molecules, with applications in analytical chemistry and diagnostics .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Description

The production of the lysenin-related protein 2 polyclonal antibody involves a rigorous process. It begins with the repeated immunization of a rabbit with recombinant Eisenia fetida lysenin-related protein 2 (1-300aa) until a satisfactory antibody titer is achieved. Following this, blood is collected from the rabbit and the antibodies are isolated from the serum using protein G. The functionality of the resulting lysenin-related protein 2 antibody is meticulously assessed through ELISA and Western Blot applications, confirming its precise interaction with the Eisenia fetida lysenin-related protein 2 protein.

LRP-2 is a member of the lysenin family, a group of pore-forming proteins derived from the coelomic fluid of the earthworm Eisenia foetida. Lysenin exhibits a specific interaction with sphingomyelin (SM). LRP-2, similarly, demonstrates specific binding to SM and, like lysenin, induces hemolysis.

Form
Liquid
Lead Time
Typically, we are able to ship products within 1-3 business days following receipt of your order. Delivery times may vary depending on the mode of purchase and location. For specific delivery information, please contact your local distributors.
Synonyms
Lysenin-related protein 2 (LRP-2) (Fetidin) (Hemolysin) (Lysenin-3) (efL3)
Target Names
N/A
Uniprot No.

Target Background

Function

Lysenin-related protein 2 (LRP-2) is a pore-forming toxin that specifically binds to sphingomyelin in the plasma membrane of various cells. This binding activity confers hemolytic properties. Additionally, LRP-2 possesses antibacterial activity against B. megaterium.

Protein Families
Lysenin family
Subcellular Location
Secreted. Target cell membrane.
Tissue Specificity
Expressed by coelomocytes.

Q&A

What is Lysenin-related protein 2 and how does it differ from other lysenin family proteins?

Lysenin-related protein 2 (LRP-2 or lysenin 3) is a member of the lysenin protein family isolated from the coelomic fluid of the earthworm Eisenia foetida. It shares significant structural and functional similarities with lysenin, specifically in its ability to bind sphingomyelin (SM) and induce hemolysis. LRP-2 demonstrates binding and hemolytic activities comparable to lysenin, while another family member, LRP-1 (lysenin 2), shows approximately 10 times less activity in these areas .

The molecular basis for these functional differences lies in their amino acid composition. Lysenin and LRP-2 share 30 common sites of aromatic amino acids, which are crucial for their biological activity. In contrast, LRP-1 has a single critical substitution at position 210, where phenylalanine is replaced with isoleucine, significantly reducing its activity . Experimental evidence has shown that restoring phenylalanine at position 210 in LRP-1 dramatically increases its activity, underscoring the importance of this aromatic amino acid in the biological functions of these proteins .

What are the primary applications of Lysenin-related protein 2 antibodies in research?

Antibodies against LRP-2 serve multiple research purposes, particularly in studying sphingomyelin distribution and dynamics in cells. These applications include:

  • Visualization of sphingomyelin distribution: LRP-2 antibodies can be used in immunofluorescence studies to investigate the localization and dynamics of sphingomyelin in cellular membranes .

  • Cancer research applications: Recent research has explored the potential of lysenin proteins in cancer therapy, including their ability to induce necrosis, autophagy, and immunogenic cell death in melanoma cells .

  • Structural and functional studies: Antibodies against LRP-2 are valuable tools for investigating the structure-function relationships of lysenin family proteins, particularly the role of specific amino acid residues in sphingomyelin binding and pore formation .

  • Detection of protein expression: In gene therapy and transfection studies, LRP-2 antibodies can be used to confirm the expression of recombinant lysenin proteins .

How do researchers differentiate between antibodies specific for LRP-2 versus those that cross-react with other lysenin family proteins?

Differentiating between antibodies specific for LRP-2 and those that might cross-react with other lysenin family members requires careful consideration of several factors:

  • Epitope mapping: Identifying the specific epitopes recognized by antibodies is crucial. Antibodies targeting unique regions of LRP-2 that differ from lysenin and LRP-1 will have higher specificity .

  • Validation techniques: Multiple validation techniques should be employed, including:

    • Western blot analysis with recombinant proteins to assess cross-reactivity

    • Immunoprecipitation followed by mass spectrometry

    • Competitive binding assays with purified proteins

  • Focus on distinguishing regions: The critical position 210 (phenylalanine in lysenin and LRP-2, isoleucine in LRP-1) represents a key distinguishing feature. Antibodies targeting this region can differentiate between LRP-1 and the other family members .

  • Pre-absorption controls: Pre-absorbing antibodies with recombinant lysenin family proteins can help determine specificity by eliminating signals from cross-reactive antibodies.

What methodological considerations are important when using LRP-2 antibodies to study sphingomyelin distribution in cellular membranes?

When utilizing LRP-2 antibodies for sphingomyelin (SM) distribution studies, researchers should consider several critical methodological factors:

  • Fixation and permeabilization protocols: SM distribution can be altered by different fixation methods. Paraformaldehyde fixation followed by careful permeabilization is often recommended to preserve membrane structure .

  • Control for non-specific binding: Include appropriate negative controls (isotype-matched control antibodies) and positive controls (cells with known SM distribution patterns).

  • Co-localization studies: Combine LRP-2 antibody staining with markers for specific cellular compartments such as endoplasmic reticulum (BAP31), Golgi apparatus (GM130), mitochondria (TOMM20), lysosomes (LAMP1), early endosomes (EEA1), late endosomes (CD63), and recycling endosomes (TfnR) .

  • Quantification methods: Employ digital image analysis with appropriate software to quantify distribution patterns rather than relying solely on visual assessment.

  • Cholesterol depletion controls: SM organization in membranes is influenced by cholesterol. Consider including cholesterol depletion/repletion experiments (using methyl-β-cyclodextrin) to understand the relationship between cholesterol levels and SM distribution as detected by LRP-2 antibodies.

  • Live versus fixed cell imaging: Be aware that fixation can alter membrane structure. When possible, validate findings using both fixed and live cell approaches with fluorescently-tagged LRP-2 derivatives.

How can researchers troubleshoot specificity issues when using LRP-2 antibodies in immunodetection experiments?

When encountering specificity issues with LRP-2 antibodies, researchers should implement the following troubleshooting strategies:

  • Validation with knockout/knockdown controls:

    • Use cells with LRP-2 knockdown/knockout as negative controls

    • Include cells overexpressing LRP-2 as positive controls

  • Sequential immunoprecipitation approach:

    • Perform sequential immunoprecipitation with antibodies against different lysenin family members

    • Analyze the resulting fractions to determine if LRP-2 is being specifically pulled down

  • Epitope competition assays:

    • Pre-incubate antibodies with recombinant LRP-2 protein before applying to samples

    • If the signal disappears, it confirms specificity for LRP-2

  • Cross-adsorption techniques:

    • Pre-adsorb antibodies with recombinant lysenin and LRP-1

    • This removes antibodies that cross-react with other family members

  • Western blot analysis with specific controls:

    • Include recombinant lysenin, LRP-1, and LRP-2 proteins on the same blot

    • Evaluate antibody reactivity against each protein

  • Altered antibody dilutions and incubation conditions:

    • Optimize antibody concentrations and incubation conditions

    • Higher dilutions often improve specificity at the cost of sensitivity

  • Secondary antibody controls:

    • Include controls with only secondary antibody to rule out non-specific binding

What are the key considerations when interpreting data from experiments using both LRP-2 antibodies and recombinant LRP-2 proteins?

Interpreting data from experiments using both LRP-2 antibodies and recombinant LRP-2 requires careful consideration of several factors:

  • Protein conformation and epitope accessibility:

    • Recombinant proteins may not always fold identically to native proteins

    • Some epitopes might be masked or exposed differently between native and recombinant forms

  • Post-translational modifications:

    • Native LRP-2 may undergo post-translational modifications not present in recombinant versions

    • These modifications can affect antibody recognition and protein function

  • Oligomerization effects:

    • LRP-2, like lysenin, can form oligomers that are SDS-resistant (>170 kDa)

    • Antibody recognition may differ between monomeric (~43 kDa) and oligomeric forms

  • Fusion tags influence:

    • Tags on recombinant proteins (like maltose-binding-protein tags mentioned in ) may affect protein folding or function

    • Consider using both tagged and untagged versions in validation studies

  • Protein-lipid interactions:

    • The presence of sphingomyelin can alter LRP-2 conformation and potentially affect antibody binding

    • Include lipid-free and lipid-bound states in controls when possible

  • Cross-reactivity with endogenous proteins:

    • Cells may express endogenous lysenin-like proteins that could complicate interpretation

    • Include appropriate controls to distinguish between endogenous and recombinant protein signals

How should researchers design experiments to study the role of specific amino acid residues in LRP-2 function?

Designing experiments to investigate the importance of specific amino acid residues in LRP-2 function requires a systematic approach:

What controls should be included when using LRP-2 antibodies to study sphingomyelin dynamics in cancer cells versus normal cells?

When studying sphingomyelin dynamics in cancer versus normal cells using LRP-2 antibodies, researchers should include the following controls:

  • Cell type-matched controls:

    • Use normal cells that correspond to the tissue of origin of the cancer cells

    • For example, when studying melanoma cells (like B16-F10), include normal melanocytes

  • Sphingomyelin depletion/supplementation controls:

    • Include cells treated with sphingomyelinase to deplete sphingomyelin

    • Include cells supplemented with exogenous sphingomyelin

    • These controls help verify that the antibody signal correlates with sphingomyelin levels

  • Blocking controls:

    • Pre-incubate cells with unlabeled lysenin proteins to block sphingomyelin binding sites

    • This confirms the specificity of antibody binding to sphingomyelin-associated epitopes

  • Cholesterol manipulation controls:

    • Include conditions with cholesterol depletion/enrichment to assess the impact on sphingomyelin organization

    • This is important because sphingomyelin and cholesterol interact in membrane microdomains

  • Cell cycle synchronization:

    • Synchronize cells at different cell cycle stages

    • Cancer cells often show altered cell cycle regulation, which may affect membrane composition

  • Dead cell discrimination:

    • Include methods to distinguish between live and dead cells

    • Cancer treatments can induce cell death, which alters membrane integrity and could affect results

  • Treatment response controls:

    • If studying treatment effects, include time-course samples to track dynamic changes in sphingomyelin distribution

How can LRP-2 antibodies be used to study immunogenic cell death in cancer therapy research?

LRP-2 antibodies can serve as valuable tools in studying immunogenic cell death (ICD) in cancer therapy research through several applications:

  • Monitoring sphingomyelin redistribution during ICD:

    • ICD involves changes in plasma membrane composition and organization

    • LRP-2 antibodies can track sphingomyelin redistribution during the ICD process, potentially revealing membrane reorganization patterns specific to immunogenic versus non-immunogenic cell death

  • Investigating the relationship between sphingomyelin exposure and DAMPs release:

    • Use LRP-2 antibodies in combination with markers for damage-associated molecular patterns (DAMPs) like HMGB1 and calreticulin

    • This can help determine if sphingomyelin exposure correlates with or precedes DAMPs release during ICD

  • Assessing membrane permeabilization mechanisms:

    • LRP-2, like lysenin, can form pores in sphingomyelin-containing membranes

    • Antibodies can help track the formation of these structures during therapeutic interventions

    • This could illuminate how different treatments induce membrane permeabilization leading to ICD

  • Flow cytometry applications:

    • Develop flow cytometry panels combining LRP-2 antibodies with ICD markers

    • This allows quantitative assessment of sphingomyelin exposure in relation to ICD markers at the single-cell level

  • Therapeutic response monitoring:

    • Use LRP-2 antibodies to monitor sphingomyelin dynamics in response to ICD-inducing therapies

    • This may help identify biomarkers predictive of successful ICD induction

  • Dendritic cell interaction studies:

    • Investigate how sphingomyelin exposure (detected by LRP-2 antibodies) affects tumor cell recognition and engulfment by dendritic cells

    • This relates to findings showing that lysenin-treated cancer cells promote dendritic cell maturation

What methodological approaches can researchers use to study the role of LRP-2 in gene therapy applications for cancer treatment?

Based on recent research exploring lysenin in gene therapy for melanoma , several methodological approaches can be applied to study LRP-2 in similar contexts:

  • Plasmid construction strategies:

    • Design expression vectors containing LRP-2 with and without secretory signal peptides

    • Create fusion constructs with reporter proteins (e.g., GFP) to track expression and localization

    • Include appropriate controls such as empty vectors and non-functional mutants

  • Nanoparticle-mediated gene delivery:

    • Develop nanoparticle formulations using polymers like mPEG-PDLLA and DOTAP for efficient delivery

    • Compare transfection efficiency across different cancer cell lines

    • Monitor intracellular distribution of expressed LRP-2 using antibodies or fluorescent tags

  • Cell death analysis protocol:

    • Implement multiple assays to characterize cell death mechanisms:

      • LDH release assay for membrane integrity assessment

      • Calcein AM staining for viability measurement

      • Electron microscopy for ultrastructural analysis

      • Immunoblotting for detection of cell death markers

  • Autophagy detection methods:

    • Monitor autophagy markers like LC3-I to LC3-II conversion by Western blot

    • Use fluorescent reporters for autophagic flux

    • Examine autophagic ultrastructures by electron microscopy

  • Immunogenic cell death evaluation:

    • Assess DAMP release (HMGB1 translocation, calreticulin exposure)

    • Measure dendritic cell maturation in co-culture experiments

    • Analyze T-cell activation markers

  • In vivo experimental design:

    • Establish tumor models with primary and distant (metastatic-like) tumors

    • Implement intratumoral administration protocols

    • Monitor both tumor growth and immune cell infiltration

    • Assess systemic toxicity through histopathology, blood counts, and biochemical analyses

What are the recommended validation steps for confirming the specificity of newly developed LRP-2 antibodies?

Validating the specificity of newly developed LRP-2 antibodies requires a comprehensive approach:

  • Recombinant protein panel testing:

    • Test antibodies against recombinant lysenin, LRP-1, and LRP-2

    • Include both native and denatured forms to assess conformation-dependent recognition

    • Quantify cross-reactivity to determine specificity ratios

  • Western blot validation:

    • Confirm detection of monomeric LRP-2 (~43 kDa) and oligomeric forms (>170 kDa)

    • Compare migration patterns with known controls

    • Test antibody recognition of both native and recombinant proteins

  • Immunoprecipitation followed by mass spectrometry:

    • Perform immunoprecipitation using the antibody

    • Analyze pulled-down proteins by mass spectrometry

    • Confirm identity as LRP-2 rather than other lysenin family members

  • Genetic knockout/knockdown controls:

    • Test antibody in systems with CRISPR/Cas9 knockout or siRNA knockdown of LRP-2

    • Signal should be significantly reduced or eliminated in these systems

  • Peptide competition assays:

    • Synthesize peptides corresponding to the antibody epitope

    • Pre-incubate antibodies with these peptides before application

    • Signal should be blocked if the antibody is specific to the intended epitope

  • Immunofluorescence correlation with sphingomyelin distribution:

    • Compare LRP-2 antibody staining with other established sphingomyelin markers

    • Test co-localization in cells with altered sphingomyelin levels

  • Documentation of validation:

    • Record all validation steps according to antibody reporting standards

    • Include batch information, optimal working dilutions, and applications tested

How can researchers distinguish between technical artifacts and genuine biological signals when using LRP-2 antibodies in complex experimental systems?

Distinguishing between technical artifacts and genuine biological signals requires systematic controls and validation:

  • Multiple antibody approach:

    • Use multiple antibodies targeting different epitopes of LRP-2

    • Consistent results across different antibodies increase confidence in biological relevance

  • Orthogonal detection methods:

    • Complement antibody-based detection with non-antibody methods

    • For example, use fluorescently labeled recombinant LRP-2 to confirm sphingomyelin binding patterns

  • Biological validation experiments:

    • Manipulate sphingomyelin levels and observe corresponding changes in antibody signal

    • Treatment with sphingomyelinase should reduce signal if it's specific to sphingomyelin-bound LRP-2

  • Concentration gradient testing:

    • Test multiple antibody concentrations to identify optimal signal-to-noise ratio

    • Plot signal intensity versus antibody concentration to identify non-specific binding thresholds

  • Secondary antibody-only controls:

    • Include controls with only secondary antibody to identify background or non-specific binding

    • This is particularly important in tissues with high autofluorescence

  • Tissue/cell type-specific considerations:

    • Validate antibodies separately for each tissue or cell type

    • What works well in one system may produce artifacts in another

  • Image acquisition standardization:

    • Use consistent exposure settings across experimental and control samples

    • Document image processing steps to ensure reproducibility

  • Signal quantification methods:

    • Employ automated, unbiased quantification methods

    • Use appropriate statistical tests to distinguish signal from background

What are the emerging applications of LRP-2 antibodies in studying membrane dynamics and cellular responses to therapeutic interventions?

Emerging applications of LRP-2 antibodies in membrane dynamics and therapeutic response research include:

  • Super-resolution microscopy of sphingomyelin microdomains:

    • LRP-2 antibodies can be used with techniques like STORM or PALM to visualize sphingomyelin at nanoscale resolution

    • This could reveal previously undetected changes in membrane organization during therapeutic interventions

  • Live-cell imaging of sphingomyelin dynamics:

    • Development of non-toxic, cell-permeable antibody fragments or nanobodies targeting LRP-2

    • These tools could enable real-time visualization of sphingomyelin redistribution during drug treatment

  • Correlation with lipid raft markers:

    • Combined use of LRP-2 antibodies with other lipid raft markers to study membrane microdomain reorganization

    • This could help understand how membrane composition affects therapeutic response

  • Study of exosome membrane composition:

    • Analysis of sphingomyelin content and distribution in exosomes using LRP-2 antibodies

    • This could provide insights into intercellular communication in cancer

  • Biomarker development:

    • Exploration of sphingomyelin exposure patterns (detected by LRP-2 antibodies) as potential predictive biomarkers for treatment response

    • Correlation of these patterns with clinical outcomes

  • Combination therapy evaluation:

    • Use of LRP-2 antibodies to study how membrane composition changes during combination therapies

    • This could help optimize drug sequencing and dosing

  • Immunotherapy response prediction:

    • Investigation of the relationship between sphingomyelin distribution and response to immune checkpoint inhibitors

    • This builds on findings that lysenin treatment can activate anti-tumor immune responses

How might comparative studies of LRP-1, LRP-2, and lysenin contribute to our understanding of structure-function relationships in sphingomyelin-binding proteins?

Comparative studies of lysenin family proteins can provide valuable insights into structure-function relationships through several approaches:

  • Site-directed mutagenesis comparative analysis:

    • Create parallel mutations in all three proteins (lysenin, LRP-1, LRP-2)

    • Compare functional effects to identify residues with consistent or divergent roles

    • Focus particularly on position 210 (phenylalanine in lysenin and LRP-2, isoleucine in LRP-1)

  • Domain swapping experiments:

    • Create chimeric proteins by swapping domains between family members

    • Test these chimeras for sphingomyelin binding and pore formation

    • This can identify functional domains responsible for specific activities

  • Evolutionary analysis of sequence conservation:

    • Compare conservation patterns across species

    • Identify residues under positive or negative selection

    • Correlate evolutionary conservation with functional importance

  • Structural biology approaches:

    • Solve crystal or cryo-EM structures of all three proteins

    • Compare binding pockets and oligomerization interfaces

    • Identify structural determinants of sphingomyelin recognition specificity

  • Lipid binding specificity profiling:

    • Compare binding profiles of all three proteins across lipid arrays

    • Identify subtle differences in lipid recognition

    • Correlate with amino acid differences between the proteins

  • Pore formation kinetics and electrophysiology:

    • Compare pore formation rates and conductance properties

    • Investigate how specific residues affect pore stability and ion selectivity

    • This can reveal the molecular basis for functional differences between family members

  • Molecular dynamics simulations:

    • Perform in silico analysis of protein-membrane interactions

    • Compare simulation results between family members

    • Identify dynamic interactions that may not be apparent in static structures

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 2024 Thebiotek. All Rights Reserved.