EDAR (Ectodysplasin A Receptor) Human, Sf9 is a tumor necrosis factor (TNF) receptor superfamily member expressed in Sf9 insect cells via baculovirus vectors . It binds ectodysplasin-A1 (EDA-A1) to activate NF-κB, JNK, and caspase-independent pathways, regulating hair, teeth, and sweat gland development . Mutations in EDAR cause hypohidrotic ectodermal dysplasia (HED), characterized by defective ectodermal derivatives .
Property | Specification |
---|---|
Amino acid length | 413 residues (27-187a.a.) |
Molecular mass | 45.6 kDa (theoretical); 40–57 kDa (SDS-PAGE) |
Glycosylation | Yes (post-translational modification) |
Purity | >90% via SDS-PAGE |
Feature | EDAR Human, Sf9 | EDAR Human, E. coli |
---|---|---|
Expression system | Baculovirus/Sf9 | E. coli |
Glycosylation | Yes | No |
Molecular mass | 45.6 kDa | 48.2 kDa |
Tag position | C-terminal His-tag | N-terminal His-tag |
Surface plasmon resonance (SPR): Demonstrated KD values for WT EDA-A1 binding to EDAR CRDS at nanomolar affinity, while mutants (A259E, D265G) showed no detectable binding .
Pull-down assays: EDAR CRDS-MBP failed to interact with mutant EDA-A1 THD proteins .
In vivo models: CRISPR-generated Eda mutant mice exhibited dose-dependent defects in hair follicles, teeth, and sweat glands .
Ectodysplasin A Receptor, Ectodysplasin 1, Anhidrotic Receptor, Anhidrotic Ectodysplasin Receptor 1, Ectodermal Dysplasia Receptor, Downless Homolog, EDA-A1 Receptor, DL, Tumor Necrosis Factor Receptor Superfamily Member EDAR, Downless, Mouse, Homolog Of, Ectodysplasin-A Receptor, ECTD10A, ECTD10B, EDA-A1R, EDA1R, ED1R, EDA3, HRM1, ED5, ED3.
Sf9, Baculovirus cells.
ADPEYSNCGE NEYYNQTTGL CQECPPCGPG EEPYLSCGYG TKDEDYGCVP CPAEKFSKGG YQICRRHKDC EGFFRATVLT PGDMENDAEC GPCLPGYYML ENRPRNIYGM VCYSCLLAPP NTKECVGATS GASANFPGTS GSSTLSPFQH AHKELSGQGH LATAAAAFES ACSLEPKSC DKTHTCPPCP APELLGGPSV FLFPPKPKDT LMISRTPEVT CVVVDVSHED PEVKFNWYVD GVEVHNAKTK PREEQYNSTY RVVSVLTVLH QDWLNGKEYK CKVSNKALPA PIEKTISKAK GQPREPQVYT LPPSRDELTK NQVSLTCLVK GFYPSDIAVE WESNGQPENN YKTTPPVLDS DGSFFLYSKL TVDKSRWQQG NVFSCSVMHE ALHNHYTQKS LSLSPGKHHH HHH.
EDAR Human produced in Sf9 Baculovirus cells is a single, glycosylated polypeptide chain containing 413 amino acids (residues 27-187) with a molecular mass of 45.6kDa. When analyzed via SDS-PAGE, it typically appears at approximately 40-57kDa. The commercially available form is expressed with a 249 amino acid hIgG-His-tag at the C-Terminus and is purified using proprietary chromatographic techniques .
Structurally, the extracellular region of EDAR consists of three cysteine-rich domains (CRDs) and adopts an extended conformation spanning approximately 65 Å in length. Each EDAR CRD binds across the convex surface of a single EDA·A1 THD (TNF homology domain) in the trimer, burying about 1340 Ų of total solvent-accessible surface area at the intermolecular interface .
EDAR plays a conserved role in the development of ectodermally derived organs across species. In both humans and mice, loss of function mutations in the genes coding for the ligand EDA-A1, EDAR, and EDARADD lead to strikingly similar phenotypes characterized by defective hair development, absence of eccrine glands, and missing or misshapen teeth .
The receptor is part of the TNF receptor superfamily and has a unique 1:1 binding mode with its ligand EDA·A1, which is distinctly different from other TNF-receptor complex structures where each receptor typically binds into the concaved groove formed by two adjacent ligand protomers .
When expressing EDAR in Sf9 baculovirus cells, researchers should consider the following methodological approach:
Vector selection: The protein coding sequence should be optimized for expression in insect cells and cloned into a suitable vector containing a strong promoter (such as polyhedrin or p10) for high-level expression.
Protein tags: Expression with a C-terminal tag (such as the 249 amino acid hIgG-His-tag used in commercial preparations) facilitates purification while preserving the native N-terminal structure critical for ligand binding .
Post-translational modifications: Monitor glycosylation status, as EDAR typically exhibits glycosylation when expressed in Sf9 cells. PNGase F treatment can be used to assess glycosylation patterns, as demonstrated in EDA·A1 studies where both glycosylated and non-glycosylated forms were observed .
Culture conditions: Maintain Sf9 cells at 27°C in serum-free medium optimized for insect cell expression to maximize protein yield while preserving proper folding.
Purification strategy: Implement a multi-step chromatography approach, typically involving affinity chromatography (utilizing the His-tag), followed by size exclusion and/or ion exchange chromatography to achieve high purity.
Functional validation of purified EDAR should include:
Binding assays: Surface plasmon resonance (SPR) or pull-down assays to measure the interaction between EDAR and its ligand EDA·A1. The binding affinity and kinetics should be consistent with published values .
Structural integrity assessment: Circular dichroism (CD) spectroscopy to confirm proper folding of the protein's secondary structure.
Signaling pathway activation: Cell-based assays measuring NF-κB activation following ligand binding, as EDAR is known to signal through this pathway .
Thermal stability testing: Differential scanning fluorimetry (DSF) to assess protein stability and batch-to-batch consistency.
Glycosylation analysis: Western blot analysis with and without PNGase F treatment to characterize glycosylation patterns, which may affect function .
EDAR variants, particularly the EDARV370A allele, represent one of the strongest candidates for recent positive selection in human evolution. Researchers can use purified EDAR Human, Sf9 in several advanced applications:
Comparative binding studies: Using purified wildtype and variant (e.g., V370A) EDAR proteins to quantitatively compare binding affinities with EDA·A1 via surface plasmon resonance or isothermal titration calorimetry.
Structural analysis: Crystallography studies comparing wildtype and variant EDAR structures, particularly focusing on the Death Domain (DD) required for interaction with the downstream signal transducer EDARADD .
Signaling pathway analysis: In vitro assessment of NFκB signaling activation differences between variants, which has been reported to be up-regulated with the 370A variant compared to 370V .
Protein-protein interaction screens: Using purified EDAR to identify novel interaction partners that might differ between populations expressing different EDAR variants.
Evolutionary timeline reconstruction: Molecular dating analysis suggests the EDARV370A allele arose in Central China approximately 30,000 years ago, providing a framework for population genetics studies using modern EDAR variants .
Research into EDAR's role in eccrine gland development should consider:
Quantitative phenotyping: Development of precise methods for counting eccrine glands, similar to the approaches used in the EDARV370A mouse model studies where gland numbers were quantitatively assessed .
Rescue experiments: Testing the ability of purified EDAR proteins (wildtype vs. variants) to rescue eccrine gland phenotypes in models with EDAR loss-of-function mutations, such as the downless (dlj, E379K) mouse model .
Signaling pathway manipulation: Modulating the EDAR-NF-κB pathway at different developmental timepoints to determine critical windows for eccrine gland specification and development.
Human association studies: Correlating EDAR genotypes with eccrine gland density in diverse human populations, expanding on findings that EDARV370A is associated with increased active eccrine gland numbers in Han Chinese populations .
Single-cell transcriptomics: Analyzing gene expression changes in developing eccrine gland progenitors following EDAR pathway activation to identify downstream effectors specific to this tissue.
The EDAR-EDA·A1 interface contains several critical residues essential for binding:
Key EDA·A1 residues:
Key EDAR residues:
Functional studies have demonstrated that mutations at the interface completely disrupt binding. For example, the A259E and D265G mutations in EDA·A1 THD abolished interaction with EDAR CRDs in pull-down and surface plasmon resonance assays. These mutations are clinically associated with non-syndromic tooth agenesis (NSTA) and X-linked hypohidrotic ectodermal dysplasia (XL-HED), respectively .
The binding specificity between EDAR and EDA·A1 is defined by several unique features:
1:1 binding mode: Unlike other TNF-receptor complexes (such as TNF-TNFR2 and RANKL-RANK) where each receptor binds into the concaved groove formed by two adjacent ligand protomers, EDAR binds in a 1:1 fashion across a single EDA·A1 THD in the trimer .
Structural determinants of specificity: Comparing EDA·A1 with the related ligand EDA·A2 reveals that two extra amino acids (Val307 and Glu308) in EDA·A1 are critical for reshaping local geometry to enable EDAR binding. These residues influence the positioning of key aromatic residues like Tyr310, Tyr311, and Phe314 .
Receptor discrimination: EDAR specifically binds EDA·A1 but not EDA·A2, while the related receptor XEDAR binds EDA·A2 but not EDA·A1. This specificity is mediated by key differences, including:
These structural differences establish the molecular basis for ligand-receptor specificity in the EDA signaling system and provide insight into the evolution of this developmentally critical pathway.
Disease-causing mutations in EDAR can impact protein structure and function through several mechanisms:
Interface disruption: Mutations at the EDA-EDAR interface disrupt ligand binding. For example:
Signaling impairment: Mutations in the EDAR Death Domain (DD) required for interaction with EDARADD, such as the V370A variant, can alter downstream NFκB signaling efficiency .
Functional consequences: The severity of phenotypic effects correlates with the degree of binding and signaling disruption:
Complete disruption of EDA-EDAR interaction leads to severe X-linked hypohidrotic ectodermal dysplasia (XL-HED)
Partial weakening of interaction results in milder non-syndromic manifestations
Some variants like V370A may actually enhance signaling, leading to phenotypes associated with thicker hair and increased eccrine gland number
Structure-function relationships: Crystal structures of the EDA·A1 THD-EDAR CRDS complex reveal that disease-causing mutations cluster at functionally significant regions of the protein-protein interface, providing mechanistic explanations for the observed phenotypes .
Several experimental approaches have proven valuable for modeling EDAR variants:
Knock-in mouse models: Generation of precise genetic modifications to create mouse models expressing human EDAR variants, such as the EDARV370A knock-in mouse. This approach offers the advantage of evaluating phenotypes on a genetically homogeneous background, isolating the effects of a single variant .
In vitro binding assays: Pull-down assays and surface plasmon resonance (SPR) using purified ectodomains of EDA·A1 and EDAR to quantitatively measure how variants affect binding affinity and kinetics .
Signaling pathway assessment: NF-κB reporter assays to measure downstream signaling activation in response to variant EDAR proteins .
Protein glycosylation analysis: Deglycosylation studies using PNGase F to determine if variants affect post-translational modifications .
Cross-species validation: Testing conserved function of variants across species (e.g., human and mouse) to validate evolutionary conservation of critical residues .
Human genotype-phenotype correlation: Association studies in human populations to correlate EDAR variants with quantitative traits (hair thickness, tooth morphology, eccrine gland number) .
Rescue experiments: Testing the ability of variant EDAR proteins to rescue phenotypes in loss-of-function models, such as the experiment demonstrating that 370A/379K heterozygous animals had more eccrine glands than 370V/379K animals .
Rigorous quality control measures for EDAR Human, Sf9 research should include:
Purity assessment: SDS-PAGE analysis to confirm protein size (expected 40-57kDa range) and purity .
Glycosylation characterization: Western blot analysis with and without PNGase F treatment to identify glycosylated versus non-glycosylated forms, as observed in related studies where EDA·A1 ectodomain showed two close bands, with the higher molecular-weight band corresponding to a glycosylated form .
Functional validation: Binding assays with known ligand (EDA·A1) to confirm biological activity, using techniques such as pull-down assays or surface plasmon resonance .
Structural integrity: Circular dichroism or thermal shift assays to confirm proper folding and stability.
Batch consistency: Lot-to-lot comparison of critical quality attributes including purity, activity, and glycosylation patterns.
Endotoxin testing: Limulus amebocyte lysate (LAL) assay to ensure preparations are endotoxin-free for cell-based applications.
Storage stability: Accelerated and real-time stability testing to establish appropriate storage conditions and shelf-life.
Effective protocols for studying EDAR's role in hair follicle development should include:
Histological analysis: Standardized sectioning and staining protocols to quantitatively assess hair shaft thickness, follicle density, and morphology in model systems .
Ex vivo hair follicle culture: Organ culture systems allowing manipulation of EDAR signaling in isolated follicles to study direct effects on growth and development.
Genotype-phenotype correlation studies: Methods for precise phenotyping of hair traits (thickness, density, shape) in human populations with different EDAR variants, building on findings that EDARV370A has been associated with increased scalp hair thickness .
Developmental timing studies: Stage-specific manipulation of EDAR signaling to determine critical windows for hair follicle induction, morphogenesis, and cycling.
Single-cell transcriptomics: Analysis of gene expression changes in different hair follicle cell populations following EDAR pathway activation.
3D skin organoid models: Development of three-dimensional skin models incorporating different EDAR variants to study hair follicle morphogenesis in a controlled environment.
Comparative models: Cross-species analysis leveraging the conserved role of the Ectodysplasin pathway in the development of ectodermally derived organs across vertebrates .
The Ectodysplasin A Receptor (EDAR) is a crucial protein in human biology, encoded by the EDAR gene. It is a cell surface receptor for ectodysplasin A (EDA), a member of the tumor necrosis factor (TNF) family of ligands. EDA plays a significant role in the development of ectodermal tissues, such as skin, hair, teeth, and sweat glands .
EDA exists in two main splice variants: 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) . The binding of EDA-A1 to EDAR triggers the recruitment of the intracellular EDAR-associated death domain (EDARADD) adapter protein, leading to the activation of the NF-κB signaling pathway .
The EDA/EDAR signaling pathway is essential for the formation of various ectodermal derivatives during prenatal development. Mutations in the EDA gene or its receptors can result in hypohidrotic ectodermal dysplasia (HED), a condition characterized by sparse hair (oligotrichosis), missing teeth (oligodontia), and reduced ability to sweat (oligohidrosis or anhidrosis) .
Recombinant EDAR (Human, Sf9) refers to the human EDAR protein produced using the Sf9 insect cell expression system. This system is commonly used for producing recombinant proteins due to its high yield and proper post-translational modifications. The recombinant EDAR is utilized in various research applications to study the EDA/EDAR signaling pathway and its implications in development and disease.
The EDA/EDAR signaling pathway has been implicated in several diseases beyond ectodermal dysplasia. Elevated levels of EDA have been observed in metabolic diseases such as non-alcoholic fatty liver disease (NAFLD), obesity, and insulin resistance . Additionally, EDA and its receptors play a role in tumor pathogenesis by regulating tumor cell proliferation, apoptosis, differentiation, and migration .