Recombinant Human Proepiregulin (EREG), partial  (Active)

Shipped with Ice Packs
In Stock

Description

Biological Function and Relevance

Proepiregulin is a transmembrane precursor cleaved to release mature epiregulin, which acts as a ligand for EGFR and ERBB4 . Key functional attributes include:

  • Receptor Activation: Induces tyrosine phosphorylation of EGFR and ERBB4, albeit with weaker dimer stabilization compared to EGF .

  • Cellular Effects: Stimulates proliferation of fibroblasts, keratinocytes, and vascular smooth muscle cells while inhibiting epithelial tumor growth .

  • Pathway Involvement: Modulates ErbB signaling, PI3K/Akt/mTOR, and MAPK pathways .

In carcinomas of the bladder, lung, and colon, EREG overexpression correlates with tumor progression and treatment resistance .

Cancer Biology

  • Tumor Microenvironment: Stromal-derived EREG promotes chemotherapy resistance in prostate cancer by activating EGFR-dependent survival pathways .

  • Metastasis: Autocrine EREG signaling in salivary adenoid cystic carcinoma (SACC) stabilizes Snail/Slug proteins, driving epithelial-mesenchymal transition (EMT) .

Developmental Biology

  • Neurogenesis: EREG enhances basal progenitor cell proliferation in primate neocortex development, a mechanism absent in mice .

Drug Discovery

  • Biomarker Potential: EREG serves as a noninvasive indicator of DNA damage response in stromal cells .

EGFR Dimerization Dynamics

  • EREG stabilizes unique EGFR extracellular dimers, acting as a partial agonist with weaker dimerization strength than EGF .

  • FRET assays show EREG-induced EGFR oligomerization is density-dependent, unlike EGF’s maximal dimerization .

Therapeutic Implications

  • Stromal Targeting: Inhibiting stromal EREG in prostate cancer models reverses chemoresistance and metastasis .

  • Ligand Bias: EREG exhibits higher proliferative induction (~3x) in HeLa cells compared to other EGF family ligands .

Mechanistic Insights

  • Snail/Slug Stabilization: EREG treatment in SACC cells prolongs Snail/Slug protein half-life via EGFR-STAT3 signaling .

  • NF-κB Dependency: DNA damage-induced EREG expression in stromal cells requires NF-κB activation .

Product Specs

Buffer
0.2 µm filtered PBS, pH 7.4, lyophilized
Form
Liquid or Lyophilized powder
Lead Time
5-10 business days
Shelf Life
The shelf life is dependent on various factors, including storage conditions, buffer components, storage temperature, and the inherent stability of the protein.
Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C. The shelf life of the lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is recommended for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
Epiregulin; EPR; ER ; Ereg; EREG_HUMAN; Proepiregulin
Datasheet & Coa
Please contact us to get it.
Expression Region
60-108aa
Mol. Weight
5.6 kDa
Protein Length
Partial
Purity
>97% as determined by SDS-PAGE.
Research Area
Cancer
Source
E.Coli
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function
Epiregulin (EREG) is a ligand for the epidermal growth factor receptor (EGFR) and ERBB4. It stimulates EGFR and ERBB4 tyrosine phosphorylation, contributing to inflammation, wound healing, tissue repair, and oocyte maturation. EREG regulates angiogenesis and vascular remodeling and promotes cell proliferation.
Gene References Into Functions
  1. In Caco-2 CFTR-shRNA cells, EREG is overexpressed due to an active IL-1beta autocrine loop, indirectly activating EGFR. This constitutes a novel signaling pathway downstream of CFTR, Cl(-), and IL-1beta.PMID: 29091309
  2. Epiregulin has been shown to be constitutively activated in metastatic lung subtypes of salivary adenoid cystic carcinoma cells, suggesting its role in tumor growth and metastasis. PMID: 26958807
  3. A study elucidated how EREG and epigen (EPGN) stabilize different dimeric conformations of the EGFR extracellular region, revealing the mechanism by which different EGFR ligands elicit specific responses. This contributes to understanding receptor tyrosine kinase (RTK) signaling specificity. PMID: 28988771
  4. Elevated levels of EREG and MMP-1 were observed in nasal polyp and uncinate tissues of Chronic rhinosinusitis with nasal polyps patients, suggesting their involvement in this inflammatory condition. PMID: 28398769
  5. Upregulation of EREG expression through promoter demethylation might be crucial in activating the EGFR pathway during the development of colorectal adenocarcinoma (CRC) and potentially other cancers. PMID: 27270421
  6. EREG and AREG are strongly regulated by methylation, and their expression is associated with CIMP status and primary tumor site. PMID: 27272216
  7. The three-dimensional structure of the EPR antibody (the 9E5(Fab) fragment) was determined in the presence and absence of EPR. PMID: 26627827
  8. Research has identified a novel pathway involving EREG and MMP-1 that contributes to the initiation and development of early-stage breast cancer. PMID: 26215578
  9. Evidence indicates that EREG plays a role in the malignant progression of glioma. PMID: 24470554
  10. The effects of EREG and V-ATPase (TCIRG1) single nucleotide polymorphism (SNP) on pulmonary tuberculosis susceptibility, if present, are influenced by gene-gene interactions in West African populations. PMID: 24898387
  11. Patients homozygous for the minor allele A of EREG rs12641042 exhibited a significantly higher 3-year survival rate compared to patients with allele C (HR 0.48; P=0.034), but significance was lost in multivariable analysis. PMID: 25203737
  12. Epiregulin is a transcriptional target of TSC2 (tuberin). PMID: 24748662
  13. Epiregulin promotes liver progenitor cell proliferation and DNA synthesis by hepatocytes. It is upregulated in the serum of patients with liver injury. PMID: 24812054
  14. Plasma HGF and EREG levels are associated with resistance to anti-EGFR antibody treatment in KRAS wild-type patients with metastatic colorectal cancer. PMID: 24800946
  15. EREG, AREG, and BTC (betacellulin) induce prostaglandin E2 production by stimulating COX-2 (prostaglandin-endoperoxide synthase 2) through MAP kinase signaling in granulosa cells. PMID: 24092824
  16. In pre-treated K-ras wild-type colorectal cancer, patients with high EREG gene expression demonstrate greater benefit from cetuximab therapy compared to those with low expression. PMID: 24335920
  17. EREG might contribute to glioma progression under the control of IRE1a. PMID: 24330607
  18. Keratinocyte hyperproliferation in cholesteatoma is promoted through overexpression of epiregulin by subepithelial fibroblasts via epithelial-mesenchymal interactions, highlighting its role in the pathogenesis of middle ear cholesteatoma. PMID: 23826119
  19. Depletion of Epiregulin with shRNA inhibited SCAP proliferation. PMID: 23829318
  20. Research suggests a significant correlation between EREG expression and KRAS expression or KRAS copy number in KRAS-mutant non-small-cell lung cancer (NSCLC) cell lines. PMID: 22964644
  21. EREG-AREG and NRG1, members of the epidermal growth factor (EGF) family, appear to influence Behçet's Disease susceptibility through main effects and gene-gene interactions. PMID: 23625463
  22. No correlation was found between the presence of a K-ras mutation and the presence of Epiregulin and Amphiregulin in colon cancer tissue. PMID: 23885463
  23. Apical mistrafficking of EREG creates an apical EGFR signaling complex that may be uncoupled from basolateral regulatory restraints, potentially leading to cell transformation. PMID: 23671122
  24. FBXL11 inhibited osteo/dentinogenic differentiation potential in MSC cells by associating with BCOR, increasing histone K4/36 methylation in the Epiregulin promoter, and repressing Epiregulin transcription. PMID: 23074094
  25. EREG gene expression was low in 7 out of 11 gastric cancer cells, and this downregulation was mediated by aberrant CpG methylation of the EREG promoter. PMID: 22508389
  26. Epiregulin (EREG) variation is associated with susceptibility to tuberculosis. PMID: 22170233
  27. Expression status of AR and EPI mRNAs might be evaluated as dynamic predictors of response in KRAS WT patients receiving cetuximab-based therapy. PMID: 21161326
  28. Follow-up of Epiregulin expression might serve as a reliable early indication of the development of ovarian cancer. PMID: 21769422
  29. Blockade of epiregulin reduced the growth of hTERT-BJ cells and colony formation of hTERT-transformed fibroblasts. Inhibition of epiregulin function in immortal hTERT-BJ cells triggered a senescence program. PMID: 12702554
  30. Epiregulin might be a more important tumor growth regulator of malignant fibrous histiocytoma through autocrine or paracrine pathways, compared to betacellulin. PMID: 15274392
  31. Upregulation of epiregulin and amphiregulin expression is part of the signal transduction pathway leading to ovulation and luteinization in the human ovary. PMID: 15474502
  32. Findings demonstrated that PGE2 may mimic LH action, at least in part, by activating amphiregulin and epiregulin biosynthesis in human granulosa cells. PMID: 16888076
  33. Epiregulin, COX2, and MMP1 and 2 collectively facilitate the assembly of new tumor blood vessels, the release of tumor cells into the circulation, and the breaching of lung capillaries by circulating tumor cells to seed pulmonary metastasis. PMID: 17429393
  34. This study reports EREG expression in breast cancer (45.5% of breast cancers studied). It is preferentially expressed in breast tumors co-expressing HER2/HER4. PMID: 17962208
  35. Epiregulin plays an autocrine role in the proliferation of corneal epithelial cells, presumably through cross-induction with other EGF family members. PMID: 18079685
  36. Hamartomatous TSC skin tumors are induced by paracrine factors released by two-hit cells in the dermis, and proliferation with mTOR activation of the overlying epidermis is an effect of epiregulin. PMID: 18292222
  37. Increased epiregulin is associated with oral squamous cell carcinomas. PMID: 18497965
  38. Epiregulin has a protective effect against apoptosis in the human corpus luteum. PMID: 18835871
  39. The regulatory mechanism of epiregulin expression in Ki-ras-transformed 267B1 prostate epithelial cells was investigated. PMID: 18948081
  40. Epiregulin expression correlates with advanced disease, is EGFR dependent, and confers invasive properties on non-small cell lung cancer cells. PMID: 19138957

Show More

Hide All

Database Links

HGNC: 3443

OMIM: 602061

KEGG: hsa:2069

STRING: 9606.ENSP00000244869

UniGene: Hs.115263

Subcellular Location
[Epiregulin]: Secreted, extracellular space.; [Proepiregulin]: Cell membrane; Single-pass type I membrane protein.
Tissue Specificity
In normal adults, expressed predominantly in the placenta and peripheral blood leukocytes. High levels were detected in carcinomas of the bladder, lung, kidney and colon.

Q&A

What is recombinant human Epiregulin and what are its key structural features?

Recombinant human Epiregulin (EREG) is a 5.6 kDa monomeric protein belonging to the EGF family of growth factors. The mature form corresponds to amino acid residues Val63-Leu108 of the precursor protein, often with an N-terminal methionine when expressed in E. coli expression systems . Commercial preparations are typically derived from bacterial expression systems and are available in various formulations, including those with carrier proteins and carrier-free versions . The protein contains essential disulfide bonds that maintain its bioactive tertiary structure, which is crucial for receptor binding and downstream signaling activities .

What are the primary biological functions of Epiregulin in normal physiology?

Epiregulin functions as a ligand for the EGF receptor (EGFR/ErbB1) and ErbB4, but does not bind to ErbB2 or ErbB3 . It plays critical roles in multiple physiological processes including:

  • Stimulation of cell proliferation in keratinocytes, hepatocytes, fibroblasts, and vascular smooth muscle cells

  • Regulation of inflammation and tissue repair mechanisms

  • Contribution to wound healing through promotion of angiogenesis and vascular remodeling

  • Involvement in reproductive biology, particularly in oocyte maturation

  • Modulation of epithelial cell function in various tissues

Unlike some growth factors, Epiregulin has also been observed to inhibit the growth of certain tumor-derived epithelial cell lines, suggesting a context-dependent role in cell proliferation regulation .

What are the optimal reconstitution and storage conditions for maintaining EREG activity?

For optimal reconstitution of lyophilized recombinant human Epiregulin:

ParameterWith Carrier (BSA)Carrier-Free
Reconstitution SolutionSterile PBS containing at least 0.1% human or bovine serum albuminSterile PBS
Recommended Concentration100 μg/mL100 μg/mL
Storage Temperature−20°C to −80°C−20°C to −80°C
Special ConsiderationsUse manual defrost freezerUse manual defrost freezer
Freeze-Thaw CyclesAvoid repeated freeze-thaw cyclesAvoid repeated freeze-thaw cycles

The product is typically shipped at ambient temperature but should be stored immediately at recommended temperatures upon receipt . Working aliquots should be prepared to minimize freeze-thaw cycles, as repeated freezing and thawing can significantly reduce protein activity.

What concentration ranges of EREG are typically effective in cell-based assays?

The effective dose for Epiregulin varies by cell type and experimental endpoint. For proliferation assays using Balb/3T3 mouse embryonic fibroblast cell lines, the ED50 (effective dose for 50% maximal response) is typically in the range of 0.125-0.75 ng/mL . This concentration range provides a useful starting point for experimental design, though optimization may be necessary for specific cell types or experimental conditions.

When designing dose-response experiments, a recommended approach is to test a logarithmic series of concentrations (e.g., 0.01, 0.1, 1, 10, and 100 ng/mL) to establish the appropriate working range for your specific experimental system .

What is the difference between carrier-free and BSA-containing EREG preparations, and how does this impact experimental design?

The primary differences between carrier-free and BSA-containing EREG preparations are:

FeatureBSA-Containing PreparationCarrier-Free Preparation
CompositionIncludes BSA as carrier proteinNo carrier protein added
StabilityEnhanced protein stabilityMay have reduced stability at dilute concentrations
Shelf-lifeGenerally longerMay be shorter at equivalent concentrations
Recommended ApplicationsCell/tissue culture, ELISA standardsApplications where BSA might interfere
Reconstitution RequirementsPBS with additional serum albumin (≥0.1%)PBS only

How can EREG activity be verified in experimental systems?

To verify EREG activity in your experimental system, consider the following methodological approaches:

  • Proliferation assays: Measure cell proliferation in responsive cell lines like Balb/3T3 mouse embryonic fibroblasts, where the ED50 should be approximately 0.125-0.75 ng/mL .

  • Receptor phosphorylation: Assess EGFR and ErbB4 tyrosine phosphorylation by Western blotting after EREG stimulation .

  • Downstream signaling: Monitor activation of canonical pathways downstream of EGFR/ErbB4, including MAPK/ERK and PI3K/AKT pathways.

  • Gene expression analysis: Measure changes in expression of known EREG-responsive genes.

  • Biological response validation: For specific research contexts, verify the expected biological outcomes, such as cell migration, angiogenesis, or differentiation.

Comparison to a positive control (such as EGF) and inclusion of appropriate inhibitor controls (e.g., EGFR inhibitors) can provide additional validation of specificity .

How does EREG signaling differ from other EGF family ligands, and what implications does this have for experimental design?

Epiregulin demonstrates several unique signaling characteristics compared to other EGF family ligands:

  • Receptor specificity: While Epiregulin binds specifically to EGFR (ErbB1) and ErbB4, it does not bind ErbB2 or ErbB3 . This differs from ligands like Neuregulin-1, which predominantly signals through ErbB3 and ErbB4.

  • Receptor activation patterns: Epiregulin can activate homodimers of both ErbB1 and ErbB4, as well as all possible heteromeric combinations of the four ErbB family members .

  • Bimodal activity: Unlike most EGF family members, Epiregulin has been observed to inhibit the growth of certain epithelial tumor cell lines while stimulating others, suggesting unique signaling outcomes .

  • Binding affinity: Epiregulin typically demonstrates lower binding affinity to EGFR compared to EGF, potentially resulting in different signaling kinetics and dynamics.

For experimental design, these differences suggest that:

  • When comparing EGF family ligands, equivalent molar concentrations rather than mass-based concentrations should be used

  • Experiments should include appropriate receptor expression profiling in the cell systems used

  • Time-course studies may reveal important differences in signaling kinetics between EREG and other family members

  • Downstream pathway analyses should consider both canonical and non-canonical signaling outputs

What are the key methodological approaches for studying EREG's role in tumor microenvironments?

Investigating Epiregulin's role in tumor microenvironments requires multi-faceted experimental approaches:

  • Expression analysis in clinical samples:

    • Immunohistochemistry to localize EREG protein in tumor tissue

    • RNA-seq or qPCR to quantify EREG mRNA levels

    • Single-cell analysis to identify specific cell populations expressing EREG

  • Functional studies in co-culture systems:

    • Use of transwell or direct co-culture models combining tumor cells with stromal components

    • Selective inhibition of EREG signaling using neutralizing antibodies or genetic approaches

    • Assessment of paracrine effects on proliferation, migration, and invasion

  • In vivo models:

    • Xenograft models with EREG-overexpressing or EREG-knockout cells

    • Orthotopic models that better recapitulate tissue-specific microenvironments

    • Use of antibody-drug conjugates targeting EREG for therapeutic assessment

  • Mechanistic investigations:

    • Analysis of EREG-induced changes in tumor-associated macrophages or fibroblasts

    • Assessment of angiogenic responses through endothelial cell assays

    • Evaluation of immunomodulatory effects using immune cell functional assays

Several published studies have employed these approaches to demonstrate EREG's role in colorectal, bladder, lung, and kidney carcinomas .

What are common challenges in EREG-based experiments and how can they be addressed?

ChallengePossible CausesSolutions
Low or inconsistent activityProtein denaturation, Improper reconstitution, Repeated freeze-thaw cyclesUse fresh aliquots, Add carrier protein to dilutions, Validate activity in established bioassays
Non-specific effectsHigh concentration usage, Presence of contaminantsTitrate concentration, Use carrier-free preparation, Include appropriate controls
Cell type-specific variabilityDifferential receptor expression, Compensatory signaling mechanismsVerify receptor expression, Perform receptor blocking experiments, Use multiple cell lines
Poor reproducibilityVariability in cell density, Passage number effects, Serum lot variationsStandardize protocols, Use serum-free conditions when possible, Document all variables
Interference in downstream assaysCarrier protein (BSA) interference, Buffer components effectsUse carrier-free preparations, Perform buffer exchange if necessary

Additional quality control measures should include:

  • SDS-PAGE analysis to confirm protein integrity (should show a single band at approximately 5-6 kDa)

  • Endotoxin testing for sensitive applications

  • Biological activity assessment in a well-characterized assay system

How can researchers distinguish between EREG-specific effects and other EGF family ligand effects in experimental systems?

To distinguish EREG-specific effects from those of other EGF family ligands:

  • Receptor profiling and manipulation:

    • Characterize the expression profile of ErbB receptors in your experimental system

    • Use receptor-specific siRNAs or CRISPR-based approaches to selectively modulate individual receptors

    • Employ receptor-selective inhibitors when available

  • Comparative studies:

    • Perform parallel experiments with multiple EGF family ligands at equivalent molar concentrations

    • Analyze temporal dynamics of signaling activation, as different ligands may induce different kinetic profiles

    • Examine differences in biological outcomes and dose-response relationships

  • Molecular approaches:

    • Utilize EREG-specific neutralizing antibodies

    • Generate EREG knockout cell lines using CRISPR/Cas9 technologies

    • Employ receptor mutants with altered binding specificity

  • Downstream signaling analysis:

    • Comprehensive phosphoproteomic analysis to identify EREG-specific signaling nodes

    • Transcriptomic profiling to identify EREG-specific gene expression signatures

    • Analysis of receptor trafficking and degradation patterns

Studies have demonstrated that combining these approaches can effectively separate EREG-specific signaling from that of other family members, revealing unique biological functions in contexts such as wound healing and tumor progression .

How can EREG be utilized in cancer research models and what methodological considerations are important?

Epiregulin has emerged as an important factor in various cancer types, with several methodological approaches proving valuable:

  • Expression correlation studies:

    • Analysis of EREG expression in patient samples correlated with clinical outcomes

    • Investigation of EREG as a biomarker for therapy response, particularly for EGFR-targeted therapies

    • Meta-analysis of public cancer genomics datasets for EREG expression patterns

  • Functional studies in cancer models:

    • EREG overexpression or knockdown in cancer cell lines to assess effects on proliferation, migration, and invasion

    • Use of recombinant EREG to stimulate cancer cells in combination with therapeutic agents to assess modulation of drug sensitivity

    • Investigation of EREG-blocking antibodies as potential therapeutic strategies

  • Targeting approaches:

    • Development of antibody-drug conjugates targeting EREG for selective delivery of cytotoxic agents to cancer cells

    • Testing of combination approaches with established therapies

    • Evaluation of EREG as an immunotherapy target

Research has shown particularly promising applications in colorectal cancer, where antibody-drug conjugates targeting EREG have demonstrated robust anti-tumor activity . Additionally, EREG has been implicated in Head and Neck Squamous Cell Carcinoma, where its expression correlates with sensitivity to EGFR inhibitors like Erlotinib .

What are the methodological approaches for studying EREG's role in inflammatory and wound healing processes?

To investigate Epiregulin's functions in inflammation and wound healing:

  • In vitro wound healing models:

    • Scratch assays with primary keratinocytes or fibroblasts with EREG treatment

    • Transwell migration assays to assess cellular motility responses

    • 3D organotypic culture systems to evaluate tissue architecture and repair mechanisms

  • Inflammation models:

    • Macrophage polarization studies in response to EREG stimulation

    • Assessment of inflammatory cytokine production in relevant cell types

    • Neutrophil recruitment and activation analyses

  • In vivo approaches:

    • Cutaneous wound healing models in wild-type and EREG-deficient animals

    • Inflammatory disease models (e.g., colitis, arthritis) with EREG neutralization or supplementation

    • Tissue-specific conditional knockout models to dissect cell type-specific contributions

  • Mechanistic investigations:

    • Analysis of angiogenesis markers and vascular remodeling in response to EREG

    • Assessment of extracellular matrix production and remodeling

    • Investigation of epithelial-mesenchymal interactions during the repair process

These methodological approaches have revealed that EREG contributes significantly to wound healing by regulating angiogenesis and vascular remodeling while stimulating cell proliferation essential for tissue repair .

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 2025 TheBiotek. All Rights Reserved.