EDAR Antibody

Ectodysplasin A Receptor, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Development of Agonist Anti-EDAR Antibodies

Pioneering work generated monoclonal antibodies through hybridoma technology:

  • Immunization: Mice immunized with EDAR-Fc fusion proteins (human/mouse)

  • Hybridoma screening: ELISA-based selection identified clones with high EDAR affinity

  • Key clones:

    • EDAR12: Chimeric rabbit antibody with cross-species reactivity

    • EPR8020: Recombinant rabbit monoclonal for WB/flow cytometry

    • 18032-1-AP: Polyclonal antibody used in Western blot/IP

Variable region analysis revealed limited heavy/light chain gene diversity among effective clones :

CloneVH GeneVL GeneCross-reactivity
1VH1-53*01Vκ4-80*01Human, mouse, chicken
3VH1-69*01Vκ1-117*01Human, dog, rat
8VH1-53*01Vκ4-80*01Mouse-specific

Functional Mechanisms

EDAR antibodies exhibit dual signaling capabilities:

  1. Developmental signaling: Activates NF-κB pathway through EDARADD adaptor

  2. Apoptotic induction: Triggers caspase-independent death via JNK activation

Key functional parameters:

  • EC50: 0.1-0.7 mg/kg for sweat gland induction in vivo

  • Half-life: ~11 days in murine models

  • Divalency requirement: Essential for therapeutic efficacy

Research Applications

Validated experimental uses include:

ApplicationProtocol DetailsReference
Western Blot1:500-1:1000 dilution, 48 kDa band
ImmunohistochemistryCitrate antigen retrieval, 1:500 dilution
Flow CytometrySurface staining of live keratinocytes
ELISAEDAR-Fc coated plates, 1 μg/ml detection
Functional NeutralizationEC50 5-10 ng/ml in Fas-reporter assay

Therapeutic Potential in XLHED

X-linked hypohidrotic ectodermal dysplasia (XLHED) models demonstrate:

  • Neonatal intervention: 0.35 mg/kg antibody restores:

    • Tail hair growth in Tabby mice

    • Molar cusp formation in canines

  • Prenatal treatment: Single in utero dose prevents:

    • Palatal clefting

    • Tracheal gland hypoplasia

Long-term effects:

  • Sweat gland functionality maintained post-treatment

  • Tooth morphology correction persists into adulthood

Oncological Implications

Emerging research links EDAR signaling to tumorigenesis:

  • Mammary cancer: Transgenic mice with EDAR overexpression show:

    • 73% tumor incidence in multiparous females

    • Squamous differentiation in ER-negative tumors

  • Therapeutic target: Anti-EDAR antibodies reduce:

    • Tumor sphere formation in vitro

    • Cancer stem cell markers CD44+/CD24- population

Product Specs

Introduction
Ectodysplasin A Receptor, also known as EDAR, is a member of the tumor necrosis factor receptor family. This receptor binds to the soluble ligand ectodysplasin A and can activate various signaling pathways, including nuclear factor-kappaB, JNK, and caspase-independent cell death pathways. EDAR plays a crucial role in developing hair, teeth, and other ectodermal derivatives. Mutations in the EDAR gene have been linked to autosomal dominant and recessive forms of hypohidrotic ectodermal dysplasia.
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
The solution contains 1 mg of EDAR antibody per milliliter of buffer consisting of PBS at pH 7.4, 10% glycerol, and 0.02% sodium azide.
Storage Procedures
For storage up to 1 month, maintain the antibody at 4°C. For extended storage, store at -20°C. Avoid repeated freeze-thaw cycles.
Stability / Shelf Life
The antibody is stable for 12 months when stored at -20°C and for 1 month at 4°C.
Applications
The EDAR antibody has undergone testing by ELISA and Western blot analysis to confirm its specificity and reactivity. However, optimal working dilutions should be determined by titration for each specific application. A starting dilution of 1:1000 is recommended.
Synonyms
Ectodysplasin A Receptor, DL, Ectodysplasin 1, Anhidrotic Receptor, Anhidrotic Ectodysplasin Receptor 1, Ectodermal Dysplasia Receptor, Ectodysplasin-A Receptor, Downless Homolog, EDA-A1 Receptor, ECTD10A, ECTD10B, EDA3, HRM1, ED3, Tumor Necrosis Factor Receptor Superfamily Member EDAR, Downless, Mouse, Homolog Of, EDA-A1R, EDA1R, ED1R, ED5, Tumor necrosis factor receptor superfamily member EDAR.
Purification Method
EDAR antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
PAT19E8AT.
Immunogen
Anti-human EDAR mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with a recombinant human EDAR protein 27-448 amino acids purified from E. coli.
Ig Subclass
Mouse IgG2a heavy chain and k light chain.

Q&A

What is EDAR and why are antibodies against it important in research?

EDAR (ectodysplasin A receptor) is a membrane-bound protein that serves as a receptor for EDA isoform A1, but not EDA isoform A2. In humans, the canonical EDAR protein consists of 448 amino acid residues with a molecular mass of approximately 48.6 kDa . EDAR is notably expressed in fetal kidney, lung, skin, and cultured neonatal epidermal keratinocytes . The protein undergoes post-translational modifications, including glycosylation, which can affect its function .

Antibodies against EDAR are important research tools because they enable detection and characterization of this receptor in various experimental settings. More importantly, the EDAR gene has been associated with ectodermal dysplasia, making EDAR antibodies valuable for studying this developmental disorder's pathophysiology . Additionally, agonist anti-EDAR antibodies have been developed that mimic the action of EDA1, opening therapeutic possibilities for conditions like X-linked hypohidrotic ectodermal dysplasia (XLHED) .

What are the common experimental applications for EDAR antibodies?

EDAR antibodies are employed in multiple experimental applications:

  • Western Blotting: For detecting EDAR protein in cell or tissue lysates under denatured or non-denatured conditions. This technique helps researchers quantify EDAR expression levels and verify protein size .

  • Immunohistochemistry (IHC): For visualizing EDAR distribution in tissue sections, particularly in developmental studies of skin, hair follicles, and other ectodermal tissues .

  • ELISA (Enzyme-Linked Immunosorbent Assay): For quantitative detection of EDAR in solution and for epitope mapping studies. ELISA is also commonly used during antibody development to screen hybridoma supernatants for antibody secretion .

  • Functional Studies: Agonist anti-EDAR antibodies can be used to activate EDAR signaling pathways in vitro and in vivo, mimicking the effects of the natural ligand EDA1 .

  • Animal Model Research: Anti-EDAR antibodies have been used to correct developmental abnormalities in EDA-deficient mice and dogs, demonstrating their potential for therapeutic applications .

How do researchers distinguish between different types of anti-EDAR antibodies?

Researchers distinguish between different types of anti-EDAR antibodies through several analytical approaches:

  • Functional Classification: Anti-EDAR antibodies can be categorized as neutral (detection only), agonist (activating EDAR signaling), or antagonist (blocking EDAR signaling). Agonist antibodies mimic the action of EDA1 and can induce developmental effects, such as hair growth in EDA-deficient mice .

  • Isotype Determination: ELISA plates coated with anti-EDAR antibodies can be probed with peroxidase-coupled antibodies against different mouse immunoglobulin isotypes (IgG1, IgG2a, IgG2b) to determine the antibody class .

  • Epitope Mapping: Different anti-EDAR antibodies bind to distinct regions (epitopes) of the EDAR protein. Researchers map these epitopes using ELISA techniques with various EDAR-Fc constructs .

  • Cross-Reactivity Analysis: Some anti-EDAR antibodies cross-react with EDAR from multiple species (mammals and birds), while others are species-specific. This property is important for selecting antibodies for particular experimental or therapeutic applications .

  • Structural Analysis: Native gel electrophoresis can confirm the monoclonal nature of purified antibodies by demonstrating their sharp migration pattern .

What are the most effective methods for generating and screening anti-EDAR monoclonal antibodies?

The generation and screening of anti-EDAR monoclonal antibodies involve several sophisticated techniques:

  • Immunization Strategy:

    • Effective immunization uses EDAR-Fc fusion proteins (human or mouse EDAR extracellular domain fused to an Fc fragment)

    • Typical protocol: Initial immunization followed by boosting at days 10-14 with antigen in PBS/STIMUNE adjuvant, then a final boost with antigen in PBS at day 40

    • The extracellular domain (amino acid residues 29-183) of EDAR is typically used as the immunogen

  • Hybridoma Production:

    • Lymph node cells from immunized mice are harvested 3 days after the final boost

    • Fusion with myeloma cells creates hybridomas

    • Selection in hypoxanthine/aminopterin/thymidine-containing medium identifies viable hybridomas

  • Primary Screening:

    • ELISA using plates coated with hEDAR-Fc identifies antibody-secreting hybridomas

    • Supernatants are tested for their ability to recognize EDAR protein

  • Functional Screening:

    • In vitro cell-based assays assess the agonistic/antagonistic activity of the antibodies

    • In vivo testing in EDA-deficient mice (e.g., ability to induce tail hair) confirms biological activity

  • Subcloning and Production:

    • Positive hybridomas are subcloned to ensure monoclonality

    • Adaptation to serum-free medium facilitates antibody purification

    • Native protein electrophoresis confirms monoclonal nature

This systematic approach yields characterized monoclonal antibodies with defined specificities and functional properties for research and potential therapeutic applications.

How can researchers determine the binding affinity and specificity of anti-EDAR antibodies?

Determining binding affinity and specificity of anti-EDAR antibodies requires multiple complementary approaches:

  • Surface Plasmon Resonance (SPR):

    • Gold standard for measuring binding kinetics and affinity

    • Allows determination of association (kon) and dissociation (koff) rate constants

    • Affinity (KD) is calculated as koff/kon

    • Fab fragments are often generated to measure monomeric binding

  • ELISA-Based Competition Assays:

    • Measures the ability of antibodies to compete with natural ligand (EDA1) for binding to EDAR

    • Can distinguish antibodies binding to different epitopes

    • ELISA plates are coated with EDAR-Fc constructs and probed with anti-EDAR antibodies in the presence/absence of FLAG-EDA1

  • Western Blot Analysis Under Different Conditions:

    • Testing antibody recognition under reducing vs. non-reducing conditions

    • Reveals whether antibodies recognize linear or conformational epitopes

    • Comparison with control proteins (e.g., hFas-Fc) assesses specificity

  • Cross-Reactivity Assessment:

    • Testing reactivity against EDAR from different species (human, mouse, rat, etc.)

    • Important for translational research and therapeutic development

    • Can be assessed by ELISA using species-specific EDAR-Fc constructs

  • Epitope Mapping:

    • Using truncated or mutated EDAR constructs to identify binding regions

    • Helps categorize antibodies and predict functional properties

    • Critical for understanding structure-function relationships

These methodological approaches provide comprehensive characterization of anti-EDAR antibodies, guiding their appropriate use in research and therapeutic applications.

What techniques are used to evaluate anti-EDAR antibody functionality in developmental biology research?

Evaluating anti-EDAR antibody functionality in developmental biology research employs specialized techniques:

  • In Vivo Phenotypic Rescue:

    • Administration of agonist anti-EDAR antibodies to EDA-deficient mice

    • Assessment of developmental rescue (e.g., tail hair induction, sweat gland formation)

    • Quantification of dose-response relationships to determine potency

    • Comparison with known effects of transgenic or recombinant EDA1

  • Cross-Species Testing:

    • Evaluation in multiple animal models (mice, dogs)

    • Assessment of cross-reactivity and conservation of functional activity

    • Important for translational research toward human applications

  • Histological Analysis:

    • Examination of tissue-specific effects (sweat glands, tracheal glands, tooth morphology)

    • Quantification of structural changes in ectodermal derivatives

    • Comparison with wild-type and untreated EDA-deficient controls

  • Timing-Dependent Studies:

    • Administration at different developmental stages to determine critical windows

    • Analysis of immediate vs. delayed effects

    • Important for understanding developmental mechanisms and therapeutic potential

  • Molecular Signaling Analysis:

    • Assessment of EDAR signaling pathway activation

    • Measurement of downstream transcriptional responses

    • Comparison with physiological EDA-EDAR signaling

These functional assessment techniques provide crucial insights into both the basic biology of EDAR signaling and the therapeutic potential of agonist anti-EDAR antibodies for developmental disorders.

How are computational methods used to predict and optimize anti-EDAR antibody structures?

Computational methods for predicting and optimizing anti-EDAR antibody structures involve sophisticated algorithms and approaches:

These computational methods accelerate antibody engineering and optimization, reducing the experimental burden while improving the likelihood of developing effective anti-EDAR antibodies for research and therapeutic applications.

What are the challenges in predicting anti-EDAR antibody properties using computational methods?

Despite advances in computational antibody design, several challenges remain in accurately predicting anti-EDAR antibody properties:

  • Varying Performance Across Properties:

    • Computational models show inconsistent performance across different antibody properties

    • Models may perform well for thermostability prediction (correlation coefficients of r = -0.84, ρ = -0.88, τ = -0.73) but poorly for immunogenicity prediction

    • For immunogenicity, models incorrectly assign both high and low confidence to therapeutics with 0% anti-drug antibody responses (r = 0.48, ρ = 0.32)

  • Limited Transferability:

    • Models trained on one dataset often perform poorly when applied to similar properties across different datasets

    • Suggests that fitness landscapes are antibody-specific and not easily generalized

  • Structural Prediction Limitations:

    • High-quality experimental structures remain superior to computational models

    • Assessment studies like AMA-II conclude that despite progress, accurate antibody structure prediction remains challenging

    • Particularly difficult for complementarity-determining regions (CDRs), especially the highly variable H3 loop

  • Docking Challenges:

    • Antibody-antigen interfaces are typically flat, limiting the effectiveness of shape complementarity in docking algorithms

    • General protein-protein docking procedures have limited application to antibody-antigen complexes

    • Requires specialized approaches like SnugDock that incorporate antibody-specific considerations

  • Data Limitations:

    • Relatively few experimental structures of EDAR-antibody complexes available

    • Limited experimental fitness data specific to anti-EDAR antibodies

    • Benchmarking datasets like FLAb aim to address this gap but more comprehensive data is needed

Understanding these limitations is crucial for researchers applying computational approaches to anti-EDAR antibody design and optimization, guiding appropriate interpretation of computational predictions.

How do researchers analyze the sequence-structure-function relationships of anti-EDAR antibodies?

Researchers employ integrated approaches to analyze sequence-structure-function relationships of anti-EDAR antibodies:

  • Variable Region Sequence Analysis:

    • RT-PCR amplification and sequencing of variable regions from hybridoma cells

    • Identification of gene usage patterns using specialized alignment software (e.g., IMGT)

    • Analysis of somatic mutations that contribute to specificity and affinity

    • Studies have revealed that while different variable genes can generate agonist anti-EDAR antibodies, the repertoire appears limited, as similar antibodies were found multiple times in analyzed panels

  • Structure-Function Correlation:

    • Generation of Fab fragments to analyze monomeric binding properties

    • Surface plasmon resonance to measure binding kinetics

    • Correlation of binding parameters with structural features and sequence variations

    • Assessment of how structural elements contribute to agonist vs. antagonist activity

  • Epitope-Function Mapping:

    • ELISA techniques using various EDAR-Fc constructs to map binding regions

    • Correlation of epitope binding with functional outcomes

    • Investigation of how different epitopes relate to receptor activation or inhibition

  • Molecular Dynamics Simulations:

    • Computational analysis of antibody-antigen complex dynamics

    • Investigation of allosteric effects during antibody-antigen recognition

    • Helps understand how binding events translate to functional outcomes

  • Mutational Analysis:

    • Systematic mutation of antibody sequences to identify critical residues

    • Correlation of mutations with changes in binding affinity, specificity, and function

    • Creates experimental fitness landscapes that can be compared with computational predictions

This multi-faceted approach provides deep insights into how the molecular features of anti-EDAR antibodies determine their biological functions, guiding rational antibody engineering efforts.

How are agonist anti-EDAR antibodies developed and evaluated for potential therapeutic applications?

Agonist anti-EDAR antibodies with therapeutic potential undergo a systematic development and evaluation process:

  • Generation and Initial Screening:

    • Immunization with EDAR-Fc fusion proteins

    • Hybridoma production and screening for antibody secretion

    • Initial selection based on binding to EDAR in ELISA

  • Functional Classification:

    • Cell-based in vitro assays to identify antibodies with agonist activity

    • Preliminary in vivo testing in EDA-deficient mice to confirm biological function

    • Focus on antibodies that mimic the action of EDA1 in development

  • Cross-Reactivity Assessment:

    • Evaluation of reactivity across species (humans, mammals, birds)

    • Important for translational research and preclinical testing

    • Most therapeutic candidates show cross-reactivity with EDAR of mammals and birds

  • Structural Characterization:

    • Analysis of antibody format activity (monomeric, divalent)

    • Many agonist anti-EDAR antibodies are active as divalent molecules

    • Structure-function relationships guide optimization

  • In Vivo Efficacy Studies:

    • Comprehensive evaluation in EDA-deficient mouse models

    • Assessment of multiple developmental parameters (sweat glands, tracheal glands, tooth morphology)

    • Testing in large animal models (EDA-deficient dogs)

    • Dose-response studies to determine therapeutic window

  • Therapeutic Potential Assessment:

    • Evaluation for XLHED (X-linked hypohidrotic ectodermal dysplasia) treatment

    • Investigation of other EDAR-related applications

    • Mouse monoclonal antibodies serve as proof-of-concept for human therapeutic development

This development pathway demonstrates how fundamental research on anti-EDAR antibodies translates into potential therapeutic applications for developmental disorders.

What are the main challenges in optimizing anti-EDAR antibodies for improved stability and reduced immunogenicity?

Optimizing anti-EDAR antibodies for therapeutic applications faces several key challenges:

These challenges highlight the need for integrated approaches combining computational prediction, experimental validation, and iterative optimization to develop anti-EDAR antibodies with improved therapeutic properties.

What experimental data conflicts exist in anti-EDAR antibody research, and how are they resolved?

Several experimental data conflicts and their resolution approaches exist in anti-EDAR antibody research:

  • In Vitro vs. In Vivo Activity Discrepancies:

    • Some hybridomas produce antibodies with in vivo activity but limited activity in cell-based in vitro assays

    • Resolution requires multiple assay systems to fully characterize antibody functionality

    • Comprehensive testing in both systems helps identify antibodies with consistent activity profiles

  • Computational Prediction vs. Experimental Results:

    • Computational models show variable performance across different antibody properties

    • Models may predict certain properties (e.g., thermostability) well but perform poorly for others (e.g., immunogenicity)

    • Resolution involves using computational models as guides rather than definitive predictors

    • Integration of multiple computational approaches with experimental validation

  • Cross-Species Functionality Variations:

    • Antibodies may show different activity levels across species despite binding to conserved epitopes

    • Resolution through detailed epitope mapping and cross-species testing

    • Understanding species-specific differences in EDAR structure and signaling

  • Monoclonal vs. Polyclonal Effects:

    • Initial studies with polyclonal antibodies may show effects not replicated by individual monoclonal antibodies

    • Resolution through comprehensive screening of multiple monoclonal candidates

    • Investigation of synergistic effects between antibodies binding different epitopes

  • Structure Prediction Discrepancies:

    • Different computational approaches may yield varying predictions for antibody structures

    • High-quality experimental structures remain superior to computational models

    • Resolution through benchmarking studies comparing different methods

    • Combination of homology modeling with knowledge-based and energy-based methods

These examples illustrate how researchers navigate conflicting data through comprehensive experimental approaches, integration of multiple methodologies, and careful interpretation of results in the context of the limitations of each technique.

How might emerging computational approaches advance anti-EDAR antibody design and optimization?

Emerging computational approaches hold significant promise for advancing anti-EDAR antibody design:

  • Advanced Deep Learning Architectures:

    • Integration of transformer-based language models with structure prediction

    • Models like AlphaFold and RosettaFold being adapted specifically for antibody design

    • Potential for improved prediction of stability, binding affinity, and immunogenicity

    • Enhanced ability to capture sequence-structure-function relationships

  • Multi-Property Optimization Algorithms:

    • Simultaneous optimization of multiple antibody properties (affinity, stability, solubility)

    • Machine learning approaches that balance trade-offs between competing properties

    • Identification of sequence modifications that improve therapeutic potential without compromising function

  • Molecular Dynamics Integration:

    • Enhanced simulation of antibody-antigen interactions

    • Better understanding of allosteric effects during binding

    • Prediction of binding kinetics (kon and koff) rather than just equilibrium binding (KD)

    • Insights into how antibody binding triggers or inhibits signaling

  • Expanded Training Datasets:

    • Freely accessible fitness landscape data for antibodies (e.g., FLAb repository)

    • Integration of high-throughput experimental data with computational predictions

    • Enhanced model training on diverse antibody properties and structures

  • Epitope-Specific Design Approaches:

    • Computational tools targeting specific EDAR epitopes associated with agonist activity

    • Structure-based design of antibodies with optimized binding to functional epitopes

    • Rational engineering of antibodies based on known structure-function relationships

These computational advances, particularly when integrated with experimental validation, promise to accelerate the development of optimized anti-EDAR antibodies for both research and therapeutic applications.

What novel experimental techniques are being developed to better characterize anti-EDAR antibody functions?

Novel experimental techniques are enhancing the characterization of anti-EDAR antibody functions:

  • High-Throughput Functional Screening:

    • Automated cell-based assays for EDAR activation

    • Parallel assessment of multiple antibody candidates

    • Quantitative measurement of signaling pathway activation

    • Correlation of structural features with functional outcomes

  • Advanced Imaging Techniques:

    • Super-resolution microscopy to visualize antibody-receptor interactions

    • Live-cell imaging to track EDAR clustering and signaling dynamics

    • Intravital microscopy to monitor antibody effects in living tissues

    • Better understanding of the spatiotemporal aspects of antibody function

  • Single-Cell Analysis:

    • Assessment of cell-to-cell variability in EDAR expression and antibody response

    • Single-cell RNA sequencing to identify transcriptional changes following antibody treatment

    • Correlation of cellular heterogeneity with developmental outcomes

    • Insights into mechanism of action at the single-cell level

  • CRISPR-Based Functional Genomics:

    • Genome-wide screens to identify factors influencing EDAR signaling

    • Precise genetic manipulation of EDAR pathway components

    • Identification of synergistic targets for combination therapy

    • Better understanding of the molecular context affecting antibody function

  • Organoid and Tissue Engineering Models:

    • 3D culture systems recapitulating ectodermal tissue development

    • Evaluation of anti-EDAR antibody effects in physiologically relevant models

    • Bridge between in vitro cell culture and in vivo animal models

    • Platform for personalized testing of antibody effects

These emerging techniques provide unprecedented insights into anti-EDAR antibody functions, facilitating more precise and effective antibody engineering for research and therapeutic applications.

How might anti-EDAR antibody research contribute to broader understanding of developmental biology and signaling pathways?

Anti-EDAR antibody research extends beyond its immediate applications to enhance broader understanding of developmental biology:

  • Spatiotemporal Control of Developmental Signaling:

    • Agonist anti-EDAR antibodies allow precise temporal control of pathway activation

    • Administration at specific developmental stages reveals critical windows for ectodermal development

    • Insights into how timing of signaling events shapes developmental outcomes

    • Better understanding of the sequential nature of tissue formation

  • Cross-Talk Between Signaling Pathways:

    • Investigation of how EDAR signaling interacts with other developmental pathways

    • Use of anti-EDAR antibodies with varying specificity and activity to probe pathway connections

    • Understanding of compensatory mechanisms when EDAR signaling is altered

    • Insights into the integrated nature of developmental signaling networks

  • Evolutionary Conservation and Divergence:

    • Cross-species studies with anti-EDAR antibodies that recognize conserved epitopes

    • Comparison of antibody effects across species (mouse, dog, potentially human)

    • Insights into evolutionary conservation of ectodysplasin pathway function

    • Understanding of species-specific adaptations in developmental mechanisms

  • Therapeutic Paradigms for Developmental Disorders:

    • Anti-EDAR antibodies as proof-of-concept for treating developmental disorders postnatally

    • Insights into the plasticity and correctability of developmental processes

    • Potential application of similar approaches to other developmental signaling pathways

    • New conceptual framework for intervention in congenital disorders

  • Structure-Function Relationships in Receptor Signaling:

    • Detailed understanding of how antibody binding to specific EDAR epitopes triggers signaling

    • Insights into receptor clustering, conformational changes, and downstream activation

    • Parallels with other TNF receptor family members and their signaling mechanisms

    • Broader principles of receptor activation applicable across multiple biological systems

This research thus contributes fundamental knowledge to developmental biology while simultaneously advancing therapeutic approaches for developmental disorders.

Table 1: Characteristics of Anti-EDAR Antibodies and Their Applications

PropertyDescriptionResearch ApplicationsCitation
Molecular TargetEctodysplasin A receptor (448 aa, 48.6 kDa)Developmental biology, ectodermal dysplasia research
Subcellular LocalizationMembrane-boundCell surface receptor studies, signaling pathway analysis
Expression PatternFetal kidney, lung, skin, epidermal keratinocytesDevelopmental studies, tissue-specific investigations
Post-translational ModificationsGlycosylationProtein maturation studies, receptor processing analysis
Primary ApplicationsWestern Blot, ELISA, IHC, functional studiesProtein detection, quantification, tissue localization
Therapeutic PotentialAgonist antibodies mimicking EDA1Treatment of XLHED, developmental disorder interventions

Table 2: Experimental Methods for Anti-EDAR Antibody Characterization

MethodPurposeTechnical DetailsOutcome MeasuresCitation
ELISAAntibody detection, epitope mappingPlates coated with hEDAR-Fc (1 μg/ml)Binding affinity, specificity
Western BlotRecognition of denatured EDARReducing vs. non-reducing conditionsEpitope type (linear vs. conformational)
Native Gel ElectrophoresisConfirm monoclonal natureStained with Amido BlackSharp migration pattern
Surface Plasmon ResonanceAffinity determinationUsing Fab fragmentsAssociation/dissociation kinetics
In Vivo TestingFunctional validationEDA-deficient miceTail hair induction, sweat gland development

Table 3: Computational Approaches for Antibody Design and Evaluation

Computational MethodApplicationPerformance MetricsLimitationsCitation
Homology ModelingAntibody structure predictionDependent on template qualityH3 loop prediction challenging
RosettaAntibodyCombined homology and ab initio modelingImproved accuracy for frameworks and CDRsHigh-quality experimental structures still superior
SnugDockAntibody-antigen complex predictionBetter than general protein-protein dockingLimited by flat epitope-paratope interfaces
Language ModelsFitness prediction (thermostability)r = -0.84, ρ = -0.88, τ = -0.73Variable performance across properties
Deep LearningImmunogenicity predictionr = 0.48, ρ = 0.32, τ = 0Poor correlation for immunogenicity

Table 4: Variable Region Characteristics of Agonist Anti-EDAR Antibodies

FeatureObservationImplicationsResearch ApplicationCitation
Gene UsageDifferent variable genes can generate agonist antibodiesLimited repertoire (similar antibodies found multiple times)Understanding molecular basis of agonist activity
Cross-ReactivityMost agonist antibodies cross-react with EDAR of mammals and birdsConservation of functional epitopesTranslational research potential
Structural FormatActive as monomeric, divalent moleculesFlexibility in antibody engineeringTherapeutic development
Functional EffectsCorrection of sweat glands, tracheal glands, tooth morphologyMultiple developmental processes affectedComprehensive developmental rescue
Species ActivityEffective in both mice and dogsTranslational potential toward human applicationsTherapeutic development pathway

Product Science Overview

Introduction

The Ectodysplasin A Receptor (EDAR) is a protein encoded by the EDAR gene in humans. It is a cell surface receptor for ectodysplasin A (EDA), a member of the tumor necrosis factor (TNF) family of ligands. EDAR plays a crucial role in the development of ectodermal tissues such as skin, hair, nails, teeth, and sweat glands .

Structure and Function

EDAR is structurally related to members of the TNF receptor superfamily. It is a transmembrane protein that binds specifically to the EDA-A1 isoform of ectodysplasin A. This binding results in the recruitment of the intracellular EDAR-associated death domain (EDARADD) adaptor protein and the activation of the NF-κB signaling pathway .

The EDA gene, located on the long arm of the X chromosome, encodes several splice variants, including EDA-A1 and EDA-A2. EDA-A1 binds to EDAR, while EDA-A2 binds to a different receptor known as EDA2R or X-linked ectodermal dysplasia receptor (XEDAR) .

Role in Development

EDAR and its ligand EDA are essential for the interaction between the ectoderm and mesoderm during embryonic development. These interactions are critical for the proper formation of several structures derived from the ectoderm, including the skin, hair, nails, teeth, and sweat glands . Mutations in the EDAR gene can lead to hypohidrotic ectodermal dysplasia, a disorder characterized by a lower density of sweat glands, sparse hair, and missing teeth .

Clinical Significance

Mutations in the EDAR gene have been associated with various developmental disorders. For instance, hypohidrotic ectodermal dysplasia is caused by inactivation mutations in the EDA gene or its receptors, leading to symptoms such as oligotrichosis (sparse hair), edentulosis or oligodontia (missing teeth), and oligohidrosis or anhidrosis (reduced or absent sweating) .

Additionally, the EDAR variant 370A, a derived G-allele point mutation, has been linked to several traits, including greater hair shaft diameter, more numerous sweat glands, and increased mammary gland density. This variant is most common in East Asian populations and is believed to have arisen around 35,000 years ago in central China .

Research and Therapeutic Potential

Recent research has explored the role of EDAR and its ligands in various diseases and conditions. For example, EDA levels have been observed to be upregulated in metabolic diseases such as non-alcoholic fatty liver disease (NAFLD), obesity, and insulin resistance . Furthermore, EDA and its receptors have been implicated in tumor pathogenesis through the regulation of tumor cell proliferation, apoptosis, differentiation, and migration .

In experimental settings, the use of mouse models has provided valuable insights into the function of EDAR. For instance, studies have shown that prenatal correction of EDAR signaling in mice can prevent conditions such as otitis media, rhinitis, and nasopharyngitis .

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.