EREG (Epiregulin) Human, HEK refers to the recombinant human Epiregulin protein produced in HEK293 (Human Embryonic Kidney 293) cells. Epiregulin is a member of the epidermal growth factor (EGF) family and functions as a ligand for EGFR (epidermal growth factor receptor) and ERBB4, influencing cell proliferation, differentiation, and oncogenesis . The HEK293 expression system ensures proper post-translational modifications, making it critical for studies requiring biologically active EREG .
EREG Human, HEK activates EGFR and ERBB4 receptors, driving downstream pathways such as:
Key Findings from Research:
High EREG expression correlates with poor prognosis in head and neck squamous cell carcinoma (HNSCC) by sustaining EGFR-Erk pathway activation .
EREG upregulates PD-L1 (programmed death-ligand 1) in HNSCC cells via c-Myc, enhancing immune evasion .
Stromal EREG in treatment-damaged tumor microenvironments induces resistance to chemotherapy by activating NF-κB and C/EBP pathways .
Cell Proliferation Assays: HEK293-derived EREG stimulates Balb/3T3 fibroblast proliferation (ED₅₀ = 0.125–0.75 ng/mL) .
Drug Sensitivity Testing: EREG overexpression increases sensitivity to EGFR inhibitors like erlotinib in HNSCC .
Antibody-Drug Conjugates: EREG-targeting ADCs show efficacy in colorectal cancer models .
Combination Therapies: Co-targeting EREG and PD-L1 enhances antitumor responses in preclinical trials .
Study Design: EREG knockdown in HNSCC cell lines (HN4, CAL27) reduced clonogenic survival and tumor growth in xenografts .
Key Result: EREG sustains EGFR-Erk activation independently of ligand degradation, mimicking EGFR mutations .
High serum EREG levels post-chemotherapy correlate with stromal activation and poor therapeutic outcomes .
EREG expression predicts response to cetuximab in metastatic colorectal cancer .
Epiregulin, a member of the EGF family, acts as a ligand for EGFR and most members of the ERBB (v-erb-b2 oncogene homolog) family of tyrosine-kinase receptors. Primarily found in the placenta, peripheral blood leukocytes, and certain carcinomas (bladder, lung, kidney, colon), Epiregulin promotes the proliferation of keratinocytes, hepatocytes, fibroblasts, and vascular smooth muscle cells while inhibiting growth in several tumor-derived epithelial cell lines. Synthesized initially as a 19.0 kDa glycosylated transmembrane precursor protein, human Epiregulin undergoes proteolytic cleavage to yield a mature secreted sequence of 6.0 kDa.
Recombinant Human EREG, produced in HEK293 cells, is a single, glycosylated polypeptide chain (amino acids 63-108a.a) with a molecular weight of 32.6 kDa. The protein comprises 289 amino acids. A 239 amino acid hIgG-His-Tag is fused to the C-terminus. Purification is achieved using proprietary chromatographic techniques.
Sterile filtered, colorless solution.
The EREG protein solution has a concentration of 0.25 mg/ml and contains 10% glycerol in Phosphate-Buffered Saline (pH 7.4).
For short-term storage (up to 2-4 weeks), keep at 4°C. For extended storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freezing and thawing.
Purity is determined to be greater than 90.0% using SDS-PAGE analysis.
Biological activity is assessed through a cell proliferation assay using Balb/3T3 mouse embryonic fibroblast cells. The ED50 is determined to be less than or equal to 1 µg/ml.
EPR, Epiregulin, Ep, ER, Proepiregulin, EREG.
HEK293 cells.
DGSMVSITKC SSDMNGYCLH GQCIYLVDMS QNYCRCEVGY TGVRCEHFFL LEPKSCDKTH TCPPCPAPEL LGGPSVFLFP PKPKDTLMIS RTPEVTCVVV DVSHEDPEVK FNWYVDGVEV HNAKTKPREE QYNSTYRVVS VLTVLHQDWL NGKEYKCKVS NKALPAPIEK TISKAKGQPR EPQVYTLPPS RDELTKNQVS LTCLVKGFYP SDIAVEWESN GQPENNYKTT PPVLDSDGSF FLYSKLTVDK SRWQQGNVFS CSVMHEALHN HYTQKSLSLS PGKHHHHHH
EREG (Epiregulin) is part of the EGF family that functions as a ligand of EGFR (Epidermal Growth Factor Receptor) and most members of the ERBB family of tyrosine-kinase receptors. Human Epiregulin is initially synthesized as a glycosylated 19.0 kDa transmembrane precursor protein, which undergoes proteolytic cleavage to produce a 6.0 kDa mature secreted sequence .
From a functional perspective, EREG exhibits a dual regulatory role in cellular processes. It stimulates the proliferation of keratinocytes, hepatocytes, fibroblasts, and vascular smooth muscle cells while simultaneously inhibiting the growth of several tumor-derived epithelial cell lines . This bifunctional nature distinguishes EREG from other EGF family members and makes it particularly interesting for developmental biology and cancer research.
HEK293 cells offer several methodological advantages for EREG production compared to bacterial, yeast, or insect cell expression systems:
Post-translational modifications: HEK293 cells perform human-specific glycosylation patterns and other post-translational modifications essential for EREG's biological activity
Protein folding efficiency: These cells contain appropriate chaperone systems for proper folding of complex human proteins
Secretion capability: HEK293 cells efficiently secrete recombinant proteins into the culture medium, simplifying purification
Scalability: HEK cultures can be scaled up using suspension cultures for larger protein yields
Transfection efficiency: HEK293 cells exhibit high transfection rates, making them suitable for both transient and stable expression
When specifically producing EREG Human Recombinant, HEK293 cells generate a properly folded, glycosylated polypeptide chain (63-108 a.a.) containing 289 amino acids with a molecular mass of 32.6 kDa . This mammalian expression system ensures the protein maintains its intended structure and function for research applications.
The following quality control parameters should be assessed for EREG Human produced in HEK cells:
For storage stability, EREG Human should be stored at 4°C if using within 2-4 weeks, or frozen at -20°C for longer periods. Addition of carrier proteins (0.1% HSA or BSA) is recommended for long-term storage, and multiple freeze-thaw cycles should be avoided to maintain protein integrity .
Optimizing transient transfection for EREG production in HEK293 cells requires systematic adjustment of several parameters based on design of experiments (DoE) principles. Research has demonstrated that yields can be maximized by implementing the following methodological approach:
Cell density optimization: Maintain cell density between 2.4-3.0 × 10^6 cells/mL at the time of transfection for optimal expression
Transfection reagent concentration: When using polyethyleneimine (PEI), a concentration range of 24-30 mg/L provides optimal transfection efficiency while minimizing cytotoxicity
DNA optimization:
Culture conditions post-transfection:
Temperature shift to 32-34°C after initial 24h at 37°C can increase protein yields
Supplement with nutrient feed 24h post-transfection
Harvest secreted protein from culture supernatant 3-5 days post-transfection
This optimized protocol has been shown to be generally applicable to recombinant antibodies and proteins expressed in HEK cells , making it a suitable starting point for EREG production that can be further refined through iterative optimization.
EREG and EGF elicit fundamentally different cellular responses in developmental organoid models, particularly in intestinal enteroids. These differences provide critical insights into the specialized roles of these growth factors in tissue development:
These differences emerge even when EREG is used at concentrations (1-100 ng/mL) comparable to standard EGF concentrations in enteroid culture media . The remarkable ability of EREG to create spatially organized structures suggests it activates developmental programs that EGF does not, making it particularly valuable for creating more physiologically relevant organoid models for research.
EREG-induced signaling exhibits significant cell type-specific variations that require careful methodological consideration when designing comparative studies:
Receptor transactivation mechanisms:
HB-EGF shedding dependency:
Receptor tyrosine involvement:
When designing comparative studies, researchers should implement these methodological controls:
Use multiple cell lines with well-characterized receptor expression profiles
Include pathway-specific inhibitors (Src inhibitors, MMP inhibitors)
Perform both dose-response and time-course experiments to capture the full signaling dynamics
Consider receptor expression levels when interpreting results
Use specific blocking agents for potential intermediaries (HB-EGF neutralizing antibodies)
These considerations ensure that observed differences reflect genuine biological variation rather than technical artifacts.
EREG Human has emerged as a critical factor in developmental biology and disease modeling, particularly in intestinal organoid systems. Recent research has revealed several innovative applications:
Developmental niche recreation: EREG has been identified as a growth factor robustly expressed in the stem cell domain of the developing human intestine . By incorporating EREG into enteroid culture media in place of the traditional EGF, researchers have successfully recreated the specialized developmental niche that supports proper intestinal epithelial organization.
Advanced organoid engineering: EREG-grown enteroids develop with remarkable spatial organization that recapitulates native intestinal architecture. This includes properly formed crypt domains containing proliferative stem cells and a differentiated villus-like central lumen . This spatial organization has been difficult to achieve with conventional organoid culture methods.
Rare cell population preservation: EREG-supplemented cultures maintain the full complement of epithelial cell types found in the native intestine, including difficult-to-preserve rare cell populations such as BEST4+ cells . This enables more comprehensive studies of specialized cell functions and rare disease phenotypes.
Signal transduction modeling: EREG can be used to study differential signaling mechanisms across cell types, providing insights into cell-specific responses that may contribute to developmental disorders or cancer progression . The finding that EREG activates different signaling pathways in different cell types offers opportunities to model context-dependent cellular responses.
These applications demonstrate how EREG is advancing our ability to create more physiologically relevant models for studying human development and disease, particularly in epithelial tissues where spatial organization is crucial for function.
Recent research has uncovered important connections between EREG signaling and immune system regulation:
Inflammatory gene expression regulation: RNA-sequencing analysis of EREG-responsive systems has identified significant effects on genes related to interferon and cytokine signaling, including IRF5, NFKBIZ, and SOCS1 . Ingenuity analysis of these datasets revealed high statistical significance for inflammatory pathways including:
Leukocyte recruitment and function: EREG expression is associated with biological processes including:
Expression pattern in immune tissues: EREG is expressed primarily in the placenta and peripheral blood leukocytes, suggesting immune-related functions . This expression pattern differs from other EGF family members and indicates potential specialized roles in immune homeostasis.
These findings suggest that EREG may serve as an important mediator in inflammatory responses and immune cell recruitment, making it a potential target for immunomodulatory interventions. The connection between EREG signaling and inflammatory gene expression also provides new avenues for understanding chronic inflammatory conditions and potential therapeutic approaches.
Accurately measuring EREG activity in complex biological systems requires a multi-faceted approach that distinguishes its effects from other EGF family members:
Receptor phosphorylation profiling:
Downstream signaling pathway analysis:
Biological activity assessment:
Specific neutralization:
Use EREG-specific neutralizing antibodies to confirm activity attribution
Compare with pan-EGF family inhibitors to determine relative contribution
Include EGF receptor blocking antibodies as controls
Gene expression analysis:
Identify EREG-specific transcriptional signatures through RNA-seq
Compare with signatures induced by other EGF family members
Focus on genes uniquely regulated by EREG signaling
These approaches, used in combination, allow researchers to specifically attribute biological effects to EREG rather than other EGF family members in complex systems such as tissue samples, patient-derived organoids, or in vivo models.
Researchers frequently encounter several challenges when producing EREG Human in HEK cells. The following table outlines these challenges and provides methodological solutions:
Additionally, researchers should implement systematic quality control testing throughout the production process, including SDS-PAGE for purity assessment, biological activity assays, and endotoxin testing to ensure consistent, high-quality EREG preparations suitable for sensitive research applications.
Designing experiments to investigate differential effects of EREG versus EGF requires careful consideration of multiple variables to ensure robust and interpretable results:
Experimental design framework:
Include matched concentrations of EREG and EGF (1-100 ng/mL range)
Perform complete dose-response curves to identify potential potency differences
Include time-course analyses (5 min to 24 h) to capture transient and sustained signaling events
Compare multiple cell types with characterized receptor expression profiles
Critical control experiments:
Receptor expression analysis: Quantify EGFR and ERBB2-4 levels in test cells
Pathway-specific positive controls: Include known activators of MAPK, PI3K, and other relevant pathways
Negative controls: Include receptor-specific inhibitors (e.g., gefitinib for EGFR)
Cellular context controls: Test effects in both 2D and 3D culture systems
Methodological approaches for pathway analysis:
Phosphorylation status: Measure receptor and downstream effector (ERK1/2, AKT) phosphorylation
Adapter recruitment: Assess Shc phosphorylation and complex formation
Gene expression: Perform RNA-seq to identify pathway-specific transcriptional signatures
Proteomics: Use phospho-proteomics to create comprehensive signaling profiles
Cell type-specific considerations:
This experimental design approach will allow researchers to distinguish the unique signaling properties of EREG from those of EGF across different cellular contexts, providing insights into their distinct biological roles.
When incorporating EREG into specialized research applications like organoid development, researchers should consider several critical methodological factors:
Concentration optimization:
Media formulation compatibility:
EREG must be used in the context of appropriate base media formulation
Confirm compatibility with other critical growth factors (e.g., Wnt, R-spondin, Noggin)
Test for potential synergistic or antagonistic interactions with other media components
Temporal considerations:
Validation approaches:
Compare morphology between EREG-grown and EGF-grown structures using brightfield microscopy
Perform immunostaining for domain-specific markers to confirm spatial organization
Use single-cell RNA sequencing (scRNA-Seq) to verify appropriate cell type composition
Validate functional properties specific to the organoid type (e.g., barrier function, secretion)
Specialized applications:
For developmental biology: Match EREG concentration to developmental stage being modeled
For disease modeling: Consider disease-specific alterations in EREG signaling
For drug screening: Standardize culture conditions to minimize variability
Quality control metrics:
Define objective criteria for successful organoid development (budding frequency, size distribution)
Implement quantitative image analysis to ensure consistency
Establish molecular and functional benchmarks for quality assessment
These considerations will help researchers successfully implement EREG in specialized applications, particularly those requiring proper spatial organization and cellular differentiation, such as advanced intestinal organoid models for developmental biology and disease research.
EREG Human presents several promising research opportunities at the intersection of disease modeling and therapeutic development:
Advanced cancer modeling:
EREG inhibits the growth of several tumor-derived epithelial cell lines , suggesting potential anti-cancer applications
The dual stimulatory/inhibitory nature of EREG could be exploited for targeted therapy approaches
EREG expression in specific carcinomas of the bladder, lung, kidney, and colon indicates potential as a biomarker
Spatially organized disease models:
EREG's ability to create spatially organized enteroids enables more accurate modeling of diseases affecting specific intestinal compartments
This provides platforms for:
Inflammatory bowel disease research with proper crypt-villus organization
Colorectal cancer progression models with defined compartmentalization
Developmental disorder investigations requiring proper tissue architecture
Immune modulation approaches:
EREG's expression in peripheral blood leukocytes and involvement in multiple immune-related pathways suggests immunomodulatory potential
Research opportunities include:
Targeting EREG-dependent inflammatory pathways in chronic inflammatory conditions
Exploiting EREG's role in leukocyte recruitment for immunotherapy approaches
Investigating EREG in viral resistance mechanisms
Regenerative medicine applications:
EREG stimulates proliferation of multiple cell types including keratinocytes, hepatocytes, and fibroblasts
This proliferative capacity could be harnessed for:
Wound healing and tissue regeneration therapies
Engineering of complex tissue grafts with proper spatial organization
Ex vivo expansion of primary tissue for transplantation
These research directions leverage EREG's unique properties to address unmet needs in disease modeling and therapeutic development, particularly where spatial organization and immune modulation are critical factors.
Emerging developments in cell signaling research are poised to transform our understanding of EREG's biological roles through several mechanisms:
Receptor heterodimer specificity:
Advanced imaging techniques now allow visualization of receptor dimer formation in real-time
This may reveal EREG's preference for specific EGFR/ERBB family heterodimer combinations
Such preferences could explain cell type-specific responses and differential effects compared to EGF
Understanding these preferences could clarify EREG's unique developmental roles
Signaling dynamics and computational modeling:
New approaches in signaling dynamics reveal that temporal patterns of pathway activation are as important as magnitude
EREG may induce distinctive temporal signaling signatures different from other EGF family members
Computational models integrating these dynamics could predict cell-specific responses to EREG
Such models may explain the remarkable spatial organization observed in EREG-grown enteroids
Single-cell signaling heterogeneity:
Single-cell analysis techniques have revealed substantial cell-to-cell variability in signaling responses
EREG may induce different response patterns across cell populations
This heterogeneity could drive important developmental processes requiring cellular diversity
Understanding this dimension may clarify EREG's role in creating organized tissue structures
Integration with mechanobiology:
Emerging research shows bidirectional relationships between mechanical forces and growth factor signaling
EREG signaling may be modulated by tissue mechanics differently than other growth factors
This interaction could explain EREG's unique effects in three-dimensional tissue contexts
Mechanistic insights could lead to improved organoid culture systems with more physiological organization
These advancing frontiers in signaling research will likely provide deeper insights into how EREG's distinctive signaling properties contribute to its specialized roles in development, homeostasis, and disease.
Several methodological advances are needed to more comprehensively characterize EREG's unique properties:
Advanced receptor binding studies:
Development of EREG-specific fluorescent probes for real-time binding visualization
Single-molecule tracking to determine receptor clustering and internalization dynamics
Quantitative binding assays across the full ERBB receptor family with comparative kinetics
These approaches would provide direct evidence of EREG's binding preferences and kinetics
Spatiotemporal signaling analysis:
Live-cell biosensors for real-time visualization of EREG-induced signaling events
Multi-parametric signaling analysis at single-cell resolution
Advanced phospho-proteomics with high temporal resolution to capture signaling dynamics
Integration of these datasets would reveal EREG's unique signaling "fingerprint"
Structural biology approaches:
High-resolution crystal structures of EREG-receptor complexes
Comparative structural analysis with other EGF family member-receptor complexes
Molecular dynamics simulations to predict ligand-specific conformational changes
These structural insights would explain mechanistic differences in receptor activation
Improved organoid analysis techniques:
Automated image analysis platforms for quantifying spatial organization in 3D cultures
Live imaging of developing organoids to track morphogenesis in real-time
Spatial transcriptomics to map gene expression patterns across organoid domains
Such techniques would provide quantitative metrics of EREG's unique organizational effects
Systems biology integration:
Multi-omics approaches combining transcriptomics, proteomics, and metabolomics
Network analysis to identify EREG-specific signaling hubs and feedback mechanisms
Comparative pathway analysis across multiple EGF family members
These integrative approaches would reveal emergent properties of EREG signaling networks
These methodological advances would move beyond the current descriptive understanding of EREG's effects to provide mechanistic insights into its unique properties and biological roles, particularly in complex developmental contexts.
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. The recombinant form of human epiregulin, expressed in HEK293 cells, is widely used in research to study its biological functions and therapeutic potential.
Epiregulin is a protein encoded by the EREG gene, located on human chromosome 4q13 . The recombinant human epiregulin protein expressed in HEK293 cells typically contains the amino acid sequence Val63-Leu108 and is often tagged with an Fc region of human IgG1 for purification and detection purposes . The molecular weight of the recombinant protein is approximately 33.8 kDa, but due to glycosylation, it may migrate to 37-42 kDa in SDS-PAGE .
The recombinant human epiregulin protein is expressed in HEK293 cells, a human embryonic kidney cell line commonly used for protein production due to its high transfection efficiency and ability to perform post-translational modifications. The protein is purified to a high degree, with a purity of over 95% as determined by SDS-PAGE and HPLC . The endotoxin level is kept below 1 EU per μg of protein, ensuring its suitability for various biological assays .
Epiregulin functions as a ligand for the epidermal growth factor receptor (EGFR) and other members of the ERBB family of tyrosine-kinase receptors . It exhibits bifunctional regulatory properties, inhibiting the growth of several epithelial tumor cells while stimulating the growth of fibroblasts and other cell types . Epiregulin binds to the EGF receptors of epidermoid carcinoma A431 cells more weakly than EGF but is more potent as a mitogen for rat primary hepatocytes and Balb/c 3T3 A31 fibroblasts .
The mechanism of action of epiregulin involves its interaction with EGFR, leading to receptor dimerization and autophosphorylation . This activation triggers downstream signaling pathways, including the RTK signaling, Fc-epsilon receptor signaling, and neurotrophin signaling pathways . These pathways regulate various cellular processes, such as muscle cell differentiation and proliferation .
Recombinant human epiregulin is used in various research applications, including cell proliferation assays, receptor binding studies, and signaling pathway analyses. It is particularly valuable in cancer research, where it helps to elucidate the role of EGFR signaling in tumor growth and progression .