EREG demonstrates remarkable species-specific expression patterns that have significant research implications:
EREG is expressed in the developing neocortex of humans and gorillas but is undetectable in mouse neocortex
In human fetal neocortex (12-19 gestation weeks), EREG is expressed at higher levels in radial glia (both apical and basal) than in neurons
The mouse Ereg gene locus shows repressive H3K27me3 modifications and lacks active chromatin marks, while the human EREG locus exhibits active H3K27ac marks in the fetal neocortex
RT-qPCR analysis confirms this inter-species difference in EREG mRNA expression
These expression differences suggest that EREG may play an important role in primate-specific aspects of brain development, making it a crucial target for comparative developmental neurobiology research .
When working with recombinant EREG Human, His protein, several factors can influence its experimental performance:
Protein conformation: Commercial EREG is often available in both native and denatured forms, with the latter primarily suitable for antibody production and immunoassays rather than functional studies
Storage conditions: Repeated freeze-thaw cycles can diminish biological activity, necessitating single-use aliquots
Buffer composition: The presence of carrier proteins (e.g., BSA) can prevent non-specific binding to surfaces and improve stability
Concentration effects: EREG demonstrates dose-dependent effects, typically requiring 1-100 ng/mL for biological responses
Receptor expression: The magnitude of cellular responses correlates with EGFR/ERBB4 receptor abundance on target cells
The purity of the recombinant protein is also critical, with most research applications requiring >80% purity as typically confirmed by SDS-PAGE analysis .
EREG plays a significant role in primate-specific aspects of brain development:
EREG supports proliferation of basal progenitor cells in the developing neocortex of primates
Addition of EPIREGULIN to mouse neocortex (which naturally lacks Ereg expression) increases proliferation of basal progenitor cells
EREG ablation in human cortical organoids reduces proliferation in the subventricular zone
Treatment with EPIREGULIN promotes a further increase in proliferation of gorilla but not human basal progenitor cells, suggesting species-tailored sensitivity
EPIREGULIN competes with EGF to promote proliferation, and inhibition of the EGF receptor abrogates the EPIREGULIN-mediated increase in basal progenitor cells
These findings suggest that species-specific regulation of EPIREGULIN expression may contribute to the increased neocortex size of primates by providing a tunable pro-proliferative signal to basal progenitor cells in the subventricular zone .
Researchers investigating EREG's role in neurological development can employ several experimental approaches:
Cerebral organoid models: Human or non-human primate cerebral organoids can be treated with recombinant EREG to study effects on progenitor proliferation
CRISPR/Cas9 gene editing: EREG can be ablated in human cortical organoids to study loss-of-function effects on neurodevelopment
Cross-species comparative studies: Adding EREG to mouse neocortical cultures (where endogenous Ereg is absent) enables examination of evolutionary differences in receptor responses
Pharmacological inhibition: Combining EREG treatment with EGFR inhibitors confirms pathway specificity
Epigenetic analysis: Examining histone modifications (H3K27ac, H3K27me3, H3K4me3) at the EREG locus provides insights into expression regulation
When designing these experiments, researchers should consider developmental timing, concentration-dependent effects, and the need for appropriate controls including other growth factors to establish EREG-specific effects.
The species-specific expression of EREG is regulated by complex epigenetic and genomic mechanisms:
Chromatin state: In mouse neocortex, the Ereg locus is marked by repressive H3K27me3 and lacks active modifications like H3K4me3 and H3K27ac. In contrast, the human EREG locus shows enrichment in active H3K27ac marks
Cis-regulatory elements (CREs): ATAC-seq and H3K27ac ChIP-seq data of fetal human neocortex identified 11 regions of open chromatin within 100 kb of the EREG gene that may represent putative active enhancers
Genomic conservation: Of the 11 putative EREG CREs, only one shows even a small ATAC-seq peak in mouse neocortex, while the orthologous regions in mouse lack H3K27ac or ATAC-seq peaks
Sequence divergence: While most putative CREs are at least partially conserved in the mouse, they show higher sequence divergence compared to primate species
Interestingly, experimental reduction of repressive H3K27me3 at the mouse Ereg locus using CRISPR/Cas9-based epigenome editing did not result in upregulation of Ereg gene expression, suggesting the mouse locus is in a fully repressed rather than poised state .
EREG has emerged as a promising target in cancer research, particularly for colorectal cancer (CRC), with several strategic approaches:
Antibody-Drug Conjugates (ADCs): EREG-targeting ADCs show efficacy in both RAS wildtype and mutant colorectal cancer models
Monoclonal antibodies: High-specificity antibodies like H231 can bind EREG with Kd values as low as 0.01 μg/ml (0.1 nmol/L)
Expression targeting: CRISPR-Cas9-based knockout of EREG can help evaluate its role in cancer cell proliferation and survival
Pathway inhibition: Combined targeting of EREG and downstream EGFR signaling may enhance therapeutic efficacy
The development of EREG-targeting strategies is particularly significant because EREG is highly expressed in both RAS wildtype and mutant CRC with minimal expression in normal tissues, making it an attractive target for therapeutic development .
EREG-targeting antibody-drug conjugates (ADCs) represent a sophisticated approach to cancer therapy with several key components:
Target selection: EREG's high expression in colorectal cancers with minimal normal tissue expression provides tumor specificity
Antibody development: Monoclonal antibodies like H231 are developed with high specificity and affinity for human and mouse EREG
Internalization properties: Selected antibodies must internalize to lysosomes following EREG binding
Conjugation chemistry: Antibodies are conjugated to cytotoxic payloads like duocarmycin DM via cleavable dipeptide or tripeptide chemical linkers
Payload mechanism: Upon internalization, lysosomal enzymes cleave the linkers, releasing the DNA-alkylating payload
Cancer Model Type | ADC Configuration | Tumor Response | Mechanism of Action |
---|---|---|---|
RAS wildtype CRC | EREG ADC (tripeptide) | Significant regression | DNA alkylation, EGFR pathway neutralization |
RAS mutant CRC | EREG ADC (tripeptide) | Growth inhibition | DNA alkylation, independent of RAS status |
Patient-derived xenografts | EREG ADC (various) | Increased survival | Targeted cytotoxicity to EREG+ cells |
Preclinical studies demonstrate that EREG ADCs are well-tolerated, neutralize EGFR pathway activity, cause significant tumor growth inhibition or regression, and increase survival in various CRC models .
EREG-targeted therapies present several distinct advantages over conventional EGFR-targeted approaches:
RAS-independence: While the efficacy of clinically approved anti-EGFR mAbs is largely limited by RAS mutational status, EREG ADCs show promise for both RAS mutant and wildtype patients
Tumor specificity: EREG's restricted expression pattern (high in tumors, low in normal tissues) potentially reduces off-target effects compared to ubiquitous EGFR targeting
Novel mechanism: EREG-targeted ADCs combine EGFR pathway neutralization with direct cytotoxic effects through payload delivery
Resistance bypass: EREG targeting may overcome resistance mechanisms that develop against direct EGFR inhibition
These advantages suggest that EREG-targeted approaches could expand therapeutic options for colorectal cancer patients, particularly those with RAS mutations who derive limited benefit from current EGFR-targeted therapies .
Comprehensive validation of EREG-specific antibodies requires multiple complementary approaches:
Binding affinity determination: Surface plasmon resonance or biolayer interferometry to quantify Kd values (e.g., H231 antibody demonstrated Kd of 0.01 μg/ml or 0.1 nmol/L)
Cell-based binding assays: Testing antibody binding using cells with confirmed EREG expression versus control cells
Cross-reactivity testing: Evaluating binding to related EGF family members to confirm specificity
Internalization assessment: Confocal microscopy or flow cytometry to confirm antibody internalization (critical for ADC development)
Functional neutralization: Testing the antibody's ability to block EREG-mediated EGFR activation
Epitope mapping: Determining the specific binding region to ensure it doesn't interfere with intended applications
For example, the development of the H231 mAb included validation using 293T cells (which lack endogenous EREG) transfected with EREG expression constructs versus vector controls to confirm binding specificity .
Several methodological approaches are valuable for investigating EREG interactions with its receptors:
Binding kinetics: Surface plasmon resonance to measure association and dissociation rates
Receptor activation: Western blotting or ELISA to quantify EGFR and ERBB4 phosphorylation after EREG treatment
Downstream signaling: Monitoring activation of pathways including MAPK/ERK and PI3K/AKT
Competitive binding: Assessing EREG competition with other EGFR ligands like EGF
Structural studies: X-ray crystallography or cryo-EM of EREG-receptor complexes
Crosslinking: Chemical crosslinkers to stabilize complexes for co-immunoprecipitation
When designing these experiments, it's important to include appropriate controls such as other EGFR ligands for comparison and receptor-blocking antibodies to confirm specificity. Time-course studies (5-60 minutes post-treatment) are essential to capture transient signaling events.
For rigorous EREG genetic manipulation studies, several methodological considerations are critical:
CRISPR-Cas9 knockout: Design guide RNAs targeting early exons (e.g., the sgRNA sequence 5'-GACAGAAGACAATCCACGTG-3' has been validated for EREG knockout)
Validation of knockout: Confirm EREG deletion at both genomic (PCR, sequencing) and protein levels (Western blot, immunofluorescence)
Overexpression systems: Clone full-length EREG or specific domains into appropriate expression vectors with tags for detection
Expression verification: Use anti-tag antibodies (e.g., anti-myc) to confirm expression in transfected cells
Selection strategies: Establish stable lines using appropriate antibiotics for long-term studies
Controls: Include both vector-only controls and rescue experiments to confirm phenotype specificity
For EREG overexpression, researchers have successfully used the pIRESpuro3 vector with EREG fused to the CD8 signal peptide sequence (MALPVTALLLPLALLLHAA) followed by a Myc-tag at the N-terminus .
Researchers working with EREG Human, His protein frequently encounter several technical issues:
Protein aggregation: His-tagged EREG may form aggregates that reduce activity
Solution: Add carrier proteins (0.1% BSA) and avoid vigorous vortexing
Inconsistent cellular responses: Variability in EREG-induced signaling
Solution: Verify receptor expression levels and standardize cell density
Batch variation: Different preparations showing varying potency
Solution: Perform functional validation of each batch using standard assays
Signal-to-noise ratio: High background in phosphorylation assays
Solution: Optimize serum starvation conditions (16-24 hours) before EREG treatment
Species cross-reactivity: Human EREG may have different potency on cells from different species
Solution: Validate activity using species-matched positive controls
For cellular experiments, researchers should carefully control cell confluency, passage number, and starvation conditions, as these factors can significantly influence EREG responsiveness.
To ensure reproducible results in EREG functional studies, implement these methodological controls:
Standardized protocols: Develop detailed SOPs for all experimental procedures
Positive controls: Include established EGFR ligands (EGF, TGF-α) in parallel with EREG
Dose-response analysis: Test multiple EREG concentrations (typically 0.1-100 ng/mL)
Biological replicates: Perform experiments across multiple cell passages or donors
Technical replicates: Include at least triplicate measurements within each experiment
Receptor quantification: Regularly verify EGFR/ERBB4 expression levels on target cells
Time-course studies: Examine responses at multiple time points to capture both immediate and delayed effects
Reagent quality control: Use the same lot of recombinant EREG when possible, or validate new lots
Documentation of all experimental parameters, including cell source, passage number, culture conditions, and reagent details, is essential for troubleshooting inconsistencies and ensuring reproducibility.
Robust experimental design for EREG studies requires comprehensive controls:
Receptor dependency: Include EGFR/ERBB4 inhibitors (e.g., erlotinib) to confirm receptor-mediated effects
Specificity controls: Test other EGF family ligands to distinguish EREG-specific versus general EGFR effects
Genetic validation: Compare effects in EREG-knockout versus wildtype cells
Functional neutralization: Use EREG-neutralizing antibodies to block activity
Concentration controls: Demonstrate dose-dependent responses to establish specificity
Vehicle controls: Include buffer-only treatments with identical carrier proteins and solvents
Sequential treatment: Pre-treat with EGFR blockers before EREG to confirm receptor dependency
For developmental studies using organoids, additional controls should include treatment with other growth factors and comparison of effects across species (human vs. gorilla vs. mouse) to highlight species-specific responses .
Epiregulin (EREG) is a member of the epidermal growth factor (EGF) family, which plays a crucial role in various cellular processes, including proliferation, differentiation, and survival. Epiregulin functions as a ligand for the epidermal growth factor receptor (EGFR) and other members of the ERBB family of tyrosine-kinase receptors .
Human recombinant epiregulin is typically produced in Escherichia coli (E. coli) as a single, non-glycosylated polypeptide chain. The recombinant protein contains 69 amino acids (63-108 a.a) and has a molecular mass of approximately 7.7 kDa . It is fused to a 23 amino acid His-tag at the N-terminus, which facilitates purification using chromatographic techniques .
The recombinant epiregulin is provided as a sterile, filtered, colorless solution. It is formulated in a buffer containing 20 mM Tris-HCl (pH 8.0) and 10% glycerol . The protein solution is stable when stored at 4°C for short-term use (2-4 weeks) and at -20°C for long-term storage. To prevent degradation, it is recommended to avoid multiple freeze-thaw cycles and to add a carrier protein (0.1% HSA or BSA) for extended storage .
Epiregulin is expressed in various tissues, including the placenta and peripheral blood leukocytes. It is also found in specific carcinomas of the bladder, lung, kidney, and colon . Epiregulin stimulates the proliferation of keratinocytes, hepatocytes, fibroblasts, and vascular smooth muscle cells. Interestingly, it inhibits the growth of several tumor-derived epithelial cell lines .