Recombinant Human Pro-epidermal growth factor (EGF), partial (Active)

Shipped with Ice Packs
In Stock

Description

The recombinant protein binds to the epidermal growth factor receptor (EGFR/ErbB1), inducing receptor dimerization and activation of downstream signaling pathways (e.g., MAPK, PI3K/AKT). Key functions include:

  • Cell proliferation: Stimulates epithelial, fibroblast, and epidermal cell growth .

  • Wound healing: Enhances re-epithelialization and granulation tissue formation .

  • Magnesium homeostasis: Activates TRPM6 channels in renal cells to regulate magnesium reabsorption .

4.1. In Vitro Studies

  • Cell culture: Used to maintain intestinal organoids and induce neurite outgrowth in neuronal models .

  • Scratch assays: Accelerates wound closure in fibroblast monolayers .

4.2. Clinical Trials (Using EGF Analogues)

IndicationRoute/DoseOutcomeStudy Design
Diabetic foot ulcersTopical (150 µg/g gel)67% reduction in healing time vs. placebo Phase III DBRCT (n=60)
Venous ulcersIntralesional (25–75 µg)89% granulation improvement Phase II trial (n=41)
Gastrointestinal repairIV infusion (100 ng/kg/h)Enhanced mucosal remodeling in neonates Randomized trial (n=8)

Limitations and Future Directions

  • Challenges: Refolding efficiency in E. coli systems affects yield .

  • Opportunities: Engineered variants with improved receptor specificity or stability are under development .

Product Specs

Buffer
0.2 mg filtered solution in PBS, pH 7.4, lyophilized
Form
Liquid or Lyophilized powder
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your order. The delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery time.
Note: All of our proteins are shipped with standard blue ice packs. If you require dry ice shipping, please inform us in advance. Additional fees will apply.
Shelf Life
The shelf life of our products is influenced by various factors, including storage conditions, buffer ingredients, 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 lyophilized form has a shelf life of 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. For multiple use, aliquoting is recommended. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
Beta urogastrone; beta-urogastrone; EGF; EGF_HUMAN; Epidermal growth factor; HOMG4; OTTHUMP00000219721; OTTHUMP00000219722; Pro epidermal growth factor; URG; Urogastrone
Datasheet & Coa
Please contact us to get it.
Expression Region
971-1023aa
Mol. Weight
6.2 kDa
Protein Length
Partial
Purity
>95% as determined by SDS-PAGE and HPLC.
Research Area
Cancer
Source
E.Coli
Species
Homo sapiens (Human)
Target Names
EGF
Uniprot No.

Target Background

Function
Epidermal growth factor (EGF) is a potent mitogen that stimulates the growth of various epidermal and epithelial tissues both in vivo and in vitro, as well as certain fibroblasts in cell culture. It acts as a magnesiotropic hormone, promoting magnesium reabsorption in the renal distal convoluted tubule through EGFR engagement and activation of the magnesium channel TRPM6. EGF can also induce neurite outgrowth in motoneurons of the pond snail Lymnaea stagnalis in vitro.
Gene References Into Functions
  1. Our research demonstrates that the chimeric EGFETA toxin is highly effective against EGFR-positive cancers, suggesting its potential for further development as a targeted therapy against EGFR-positive tumors resistant to monoclonal antibodies. PMID: 30226622
  2. These findings highlight the potential role of EGF in promoting hepatocellular carcinoma (HCC) metastasis. They also demonstrate a novel pathway for the regulation of fibronectin expression and identify potential targets for HCC prevention and treatment. PMID: 29315755
  3. Abnormally elevated expression of EGF and TGF-alpha is strongly associated with the occurrence and development of chronic pancreatitis and pancreatic cancer. PMID: 29125273
  4. ERRα positively regulates the cell proliferation, migration, and invasion of colon cancer cells. Suppression of ERRa completely eliminates EGF treatment-induced proliferation of colon cancer cells. PMID: 30185207
  5. EGF significantly upregulates RFPL3 and hTERT protein levels in non-small cell lung cancer cells. Pretreatment with AG1478 and erlotinib attenuates RFPL3 and hTERT protein upregulation induced by EGF. EGF promotes proliferation and inhibits apoptosis; PD98059 decreases RFPL3 and hTERT protein expression; and RFPL3 overexpression increases the expression of hTERT and related MEK pathway proteins. PMID: 29749533
  6. We have identified a novel N-72 residue, which plays a crucial role in EGF-induced migration by targeting MMP2 in human amnion mesenchymal stem cells (hAMSCs). PMID: 29734654
  7. The spleen regulates the functions of hematopoietic stem cells in cirrhotic hypersplenism by modulating EGF signaling. PMID: 29721775
  8. Upon blocking HIP1 expression using siRNAs, EGFR endocytosis is accelerated in a manner dependent on the EGF concentration. This endocytosis is colocalized with clathrin expression. These findings indicate that inhibiting HIP1 can accelerate the endocytosis and degradation of EGFR. PMID: 29039605
  9. The current study demonstrated that EGF induces aggressiveness of gastric cancer cells by activating epithelial to mesenchymal transition, involving the activation of the ERK1/2 pathway and subsequent uPAR expression. PMID: 28849196
  10. The EGF system serves as a mechanosensitizer in bone marrow stromal cells. PMID: 28843157
  11. EGF counteracts Tat modulation of human endogenous retroviruses of the W family in astrocytes. PMID: 28474333
  12. FTIR spectra of EGF, unconjugated, post-treatment with alpha-lipoic acid, attached to gold nanoparticle, and bound to the bifunctional nanoprobe, showed decreasing disordered structures and turns, and increasing loops, as the synthesis process progressed. There was an overall increase in beta-sheets in the final product compared to pure EGF, but this increase was not linear and fluctuated. PMID: 29122663
  13. EGF-mediated lysosome trafficking, protease secretion, and invasion are regulated by the activity of p38 mitogen-activated protein kinase (MAPK) and sodium hydrogen exchangers (NHEs). Notably, EGF stimulates anterograde lysosome trafficking through a distinct mechanism compared to that previously reported for HGF, suggesting redundant signaling pathways control lysosome positioning. PMID: 28978320
  14. Despite the hostile environment for local growth factors bioavailability in diabetic chronic wounds, EGF local infiltration overcomes the limitations of topical application, expanding its therapeutic potential. Our clinical pharmacovigilance and basic studies confirm the significance of local growth factor infiltration for chronic wound healing. PMID: 28904952
  15. These results provide the first evidence for an association between the EGF rs2298999 C/T polymorphism and gout. PMID: 27506295
  16. The increased EGFR expression observed in patients with seborrheic keratomas (SK) and concurrent type 2 diabetes mellitus (DM2) is attributed to insulin resistance and hyperinsulinemia. The dysregulation of insulin signal transmission into the cell leads to alterations in EGF synthesis and signaling pathway, which regulates cell proliferation and growth. PMID: 28791994
  17. A novel EGFR-NF-kappaB-FOXC1 signaling axis is crucial for breast lobular carcinoma (BLBC) cell function. PMID: 28629477
  18. EGFR pathway gene expression analysis indicated that DeltaNp63 alters EGFR-regulated genes involved in cell adhesion, migration, and angiogenesis. The addition of EGF or neutralizing EGFR antibodies demonstrated that EGFR activation is responsible for DeltaNp63-mediated loss of cellular adhesion. PMID: 28349272
  19. EGF upregulates CCL2 expression in head and neck squamous cell carcinoma (HNSCC) cells, recruiting monocytes and transforming them into M2-like macrophages, thus establishing a positive feedback paracrine loop. PMID: 27888616
  20. This study shows that EGF induces epithelial-mesenchymal transition through phospho-Smad2/3-Snail signaling pathway in breast cancer cells. PMID: 27829223
  21. EGF and TNFalpha cooperatively promote the motility of HCC cells primarily through NF-kappaB/p65 mediated synergistic induction of FN in vitro. These findings highlight the interplay between EGF and TNFalpha in promoting HCC and provide potential targets for HCC prevention and treatment. PMID: 28844984
  22. Data suggests that EGF induces colorectal cancer cells to undergo epithelial-mesenchymal transition, enhances their ability to invade/migrate, and promotes phosphorylation of Ezrin at Tyr353. (EGF = epidermal growth factor) PMID: 28535417
  23. Simulation results indicate that human epidermal growth factor receptor (hEGFR) soluble extracellular domains (sECD):EGF exhibit different dynamic properties between the two pHs, and the complex may have a higher tendency of activation at pH 8.5. PMID: 27179806
  24. EGF and IP-10 levels were significantly elevated, while GRO levels were lower in the tear profile of HIV patients with dry eye disease (DED) compared to immunocompetent patients with DED. PMID: 27585367
  25. Data (including data from studies using transgenic/knockout mice) suggest that surfactant protein A1 (SPA1) interferes with EGF binding to EGFR in pulmonary alveoli cell lines. SPA1 directly binds the extracellular domain of EGFR; SPA1 binding to EGFR appears to differ from SPD binding to EGFR; SPA1 binding to EGFR does not suppress EGF-induced phosphorylation of EGFR or cell proliferation. PMID: 28972165
  26. EGF-AREG interplay in airway basal cell stem/progenitor cells is one of the mechanisms mediating the interconnected pathogenesis of all major smoking-induced lesions in the human airway epithelium. PMID: 27709733
  27. Caspase-3 inhibitors also suppressed the attenuation of cell adhesion and phosphorylation of p38 MAPK by EGF-F9. Our data indicated that EGF-F9 activated signals for apoptosis and induced de-adhesion in a caspase-3 dependent manner. PMID: 27129300
  28. Evidence suggests that CDK1/2 participate in the regulation of constitutive pre-mRNA splicing by EGF stimulation in MDA-MB-468 cells. PMID: 27109354
  29. The EGF rs4444903 GG genotype is associated with a higher susceptibility to HCV-related liver cirrhosis and hepatocellular carcinoma in the Chinese Han population. PMID: 28397482
  30. TGF-beta opposes EGF-mediated sensitization to TRAIL-induced caspase-8 activation and apoptosis in non-transformed breast epithelial cells. EGF and TGF-beta finely regulate the sensitivity of human breast epithelial cells to TRAIL, which may be relevant during morphogenesis. PMID: 27208428
  31. Amplification of the EGFR gene can be maintained and modulated by varying EGF concentrations in in vitro models of glioblastoma multiforme. PMID: 28934307
  32. Our study showed that the EGF61 rs4444903GA genotype had a decreased risk of non-syndromic cleft lip with or without cleft palate. Our data provide further evidence regarding the role of EGF61 variations in the development of non-syndromic cleft lip with or without cleft palate in families of the studied populations. PMID: 28906376
  33. Interestingly, EGF rapidly downregulates LINC01089 (here renamed LncRNA Inhibiting Metastasis; LIMT) expression by enhancing histone deacetylation at the respective promoter. PMID: 27485121
  34. EGF-induced, calpain-mediated proteolysis contributes to the rapid destruction of cyclin G2, and the PEST domain is critical for EGF/calpain actions. PMID: 28640887
  35. The salivary levels of EGF are significantly increased during the acute phase of natural rotavirus infection. PMID: 28558652
  36. Findings have identified a role for members of these signaling pathways in the regulation of EGF-induced vimentin expression in the MDA-MB-468 breast cancer cell line. PMID: 27163529
  37. miR-223 downregulates the local expression of epidermal growth factor (EGF), leading to decreased activation of EGF receptor (EGFR) on target cells and ultimately dampening a positive EGF-EGFR autocrine/paracrine stimulation loop induced by the post-surgical wound-healing response. PMID: 26876200
  38. EGFR and EGF expression showed no significant difference between placentas from normal pregnancies and those complicated with preeclampsia. PMID: 27657362
  39. Atomistic molecular dynamics simulations demonstrate that N-glycosylation of the EGFR extracellular domain plays critical roles in the binding of growth factors, monoclonal antibodies, and dimeric partners to the monomeric EGFR extracellular domain. PMID: 28486782
  40. CMTM3 decreases EGFR expression, facilitates EGFR degradation, and inhibits the EGF-mediated tumorigenicity of gastric cancer cells by enhancing Rab5 activity. PMID: 27867015
  41. Findings suggest that EGF not only promotes the proliferation of adipose stem cells and delays their senescence but also maintains the differentiation potency of adipose stem cells, which is related to the EGF-induced activation of the STAT signaling pathway. PMID: 28746211
  42. The results show that the interaction between STS-1 and ShcA is regulated in response to EGF receptor activation. PMID: 28690151
  43. Insulin treatment causes sustained Akt activity, while EGF or PDGF-AA promotes transient signaling. PDGF-BB produces sustained responses at higher concentrations. Transient responses to EGF are caused by negative feedback at the receptor level, as a second treatment yields minimal responses, whereas parallel exposure to IGF-I causes full Akt activation. PMID: 27044757
  44. Our results indicate that different concentrations of bFGF and EGF supplemented during the propagation of neural rosettes are involved in altering the identity of the resultant neural cells. PMID: 27321088
  45. F25P preproinsulin effectively reduced the concentrations of EGF, VEGF, and MMP-9 in the blood of tumor-bearing mice with EGFR-mutant glioblastoma. PMID: 27317648
  46. Conformational stability of the EGFR as influenced by glycosylation, dimerization, and EGF hormone binding has been described. PMID: 28019699
  47. Differential expression patterns of EGF, EGFR, and ERBB4 are essential in epithelial restitution and remodeling in nasal epithelium. PMID: 27285994
  48. Phosphorylation and immunohistochemical assays on the EGF receptor in HeLa cells indicate that the EGF protein produced in soybean seed is bioactive and comparable to commercially available human EGF. This work demonstrates the feasibility of using soybean seeds as a biofactory to produce therapeutic agents in a soymilk delivery platform. PMID: 27314851
  49. Data suggest that activated platelets release ADAMDEC1, which hydrolyzes pro-EGF (epidermal growth factor) to soluble, active HMW-EGF. Proteolytic cleavage of pro-EGF first occurs at the C-terminal arginyl residue of the EGF domain. Proteolysis is the regulated, rate-limiting step in generating soluble EGF from activated platelets. PMID: 28455445
  50. Subgroup analysis in a Slovak population by gender showed that the genotype EGF G61G and allele G was associated with a non-significantly increased risk of MDD. PMID: 27755861

Show More

Hide All

Database Links

HGNC: 3229

OMIM: 131530

KEGG: hsa:1950

STRING: 9606.ENSP00000265171

UniGene: Hs.419815

Involvement In Disease
Hypomagnesemia 4 (HOMG4)
Subcellular Location
Membrane; Single-pass type I membrane protein.
Tissue Specificity
Expressed in kidney, salivary gland, cerebrum and prostate.

Q&A

What is the molecular structure of recombinant human EGF and how does it differ from native EGF?

Recombinant human EGF is a polypeptide containing approximately 53 amino acids with three intramolecular disulfide bonds that are critical for its biological activity. The active domain typically spans positions 971-1023 of the full pro-EGF protein, with specific sequences such as "NSDSECPLSHDGYCLHDGVCMYIEALDKYACNCVVGYIGERCQYRDLKWWELR" as observed in commercially available recombinant products . The three-dimensional structure features β-sheets stabilized by disulfide bonds, which is essential for receptor binding and biological activity.

Native EGF is produced as a large precursor protein (pro-EGF) of approximately 1207 amino acids that undergoes proteolytic processing to generate the mature, active EGF peptide. In contrast, recombinant versions are engineered to produce the active portion directly. Some recombinant forms include modifications such as histidine tags (His-tags) for purification purposes, which are not present in the native form . The isoelectric point of human EGF is approximately 4.8 , which influences its solubility and behavior in different buffer systems.

When produced in mammalian expression systems like HEK293 cells, recombinant EGF closely resembles the native structure, while bacterial expression may require additional refolding steps to ensure proper disulfide bond formation .

Through what molecular mechanisms does EGF exert its biological effects?

EGF exerts its biological effects primarily through binding to the EGF receptor (EGFR), a transmembrane receptor tyrosine kinase of the ErbB family . This binding initiates a cascade of signaling events that regulate various cellular processes:

  • Receptor Activation: When EGF binds to the extracellular domain of EGFR, it induces receptor dimerization and autophosphorylation of tyrosine residues in the cytoplasmic domain.

  • Signal Transduction: The phosphorylated receptor activates multiple signaling pathways including:

    • RAS/RAF/MEK/ERK pathway (promoting cell proliferation)

    • PI3K/AKT/mTOR pathway (regulating cell survival and metabolism)

    • PLCγ/PKC pathway (calcium signaling and cell migration)

    • JAK/STAT pathway (gene transcription regulation)

  • Cellular Responses: These signaling cascades ultimately lead to:

    • Stimulation of cell growth in various epidermal and epithelial tissues

    • Promotion of cell migration and wound healing

    • Regulation of cell differentiation

    • Modulation of magnesium reabsorption in the renal distal convoluted tubule via activation of the magnesium channel TRPM6

    • Inhibition of inflammatory responses through suppression of TLR2 and NF-κB activity

Research has also demonstrated that EGF can induce neurite outgrowth in specific neuronal cell types, including motoneurons of the pond snail Lymnaea stagnalis , indicating its diverse biological functions beyond epidermal tissues.

What are the key differences between full-length pro-EGF and partial active EGF in research applications?

The differences between full-length pro-EGF and partial active EGF have significant implications for research applications:

CharacteristicFull-length Pro-EGFPartial Active EGF
Size~130 kDa (1207 amino acids)~6.4 kDa (53 amino acids)
SolubilityLower due to size and transmembrane domainsHigher, more soluble in aqueous solutions
BioactivityRequires processing for activityDirectly active in biological assays
Half-life in cultureGenerally longer due to membrane associationShorter, requiring more frequent replenishment
Receptor bindingPrimarily through processed EGF domainDirect binding to EGFR
Research applicationsStudies of EGF processing and regulationDirect signaling studies, cellular assays
Expression systemsMore challenging to express fullyEasier to produce at high purity

For most experimental applications requiring direct biological activity, the partial active EGF is preferred due to its immediate activity, higher solubility, and ease of handling . The recombinant partial active EGF typically encompasses amino acids 971-1023 of the pro-EGF sequence, which contains the core EGF domain responsible for receptor binding and activation .

What are the optimal conditions for maintaining EGF stability and activity in experimental settings?

Maintaining the stability and activity of recombinant human EGF is critical for experimental reproducibility. Based on research findings, the following conditions are optimal:

Storage Conditions:

  • Long-term storage: -80°C in single-use aliquots to avoid freeze-thaw cycles

  • Short-term storage (up to one month): 2-8°C

  • Addition of carrier proteins (0.1% HSA or BSA) is recommended for dilute solutions to prevent adsorption to tubes

Buffer Composition:

  • Optimal pH range: 7.0-8.0

  • Common formulations include:

    • 20mM Tris, 150mM NaCl, pH 8.0, containing stabilizers such as 1mM EDTA, 1mM DTT

    • 10% glycerol, 20mM Tris-HCl (pH 8.0), 0.1M NaCl with reducing agents

    • Addition of trehalose (5%) has been shown to enhance stability during freeze-thaw cycles

Handling Recommendations:

  • Avoid repeated freeze-thaw cycles (maximum 2-3 cycles)

  • Use low-binding microcentrifuge tubes for dilute solutions

  • When reconstituting lyophilized EGF, avoid vigorous vortexing which can cause denaturation

  • Reconstitution to concentrations of 0.1-1.0 mg/mL is typically recommended

  • Sterile filtration (0.22 μm) should be performed for cell culture applications

Activity Preservation:

  • The presence of disulfide bonds in EGF makes it sensitive to reducing agents at high concentrations

  • Maintain proper disulfide bond formation for biological activity

  • For experiments requiring long incubation periods, consider periodic supplementation with fresh EGF due to its relatively short half-life in culture conditions

Research has shown that EGF activity begins to decline after multiple freeze-thaw cycles, with significant loss after 3-5 cycles. Furthermore, dilute solutions (<0.1 mg/mL) show accelerated activity loss due to adsorption to container surfaces .

How should researchers design dose-response experiments to evaluate EGF activity in different cell types?

Designing robust dose-response experiments for EGF requires careful consideration of cell type-specific responses and methodological approaches:

Experimental Design Framework:

  • Cell Selection and Preparation:

    • Choose relevant cell types (epithelial cells, fibroblasts, keratinocytes)

    • Ensure cells are responsive to EGF (express EGFR)

    • Use low-passage cells at 60-70% confluence

    • Serum-starve cells (0.1-0.5% serum) for 12-24 hours before EGF treatment to reduce background signaling

  • Dose Range Determination:

    • Typical effective concentration range: 0.1 ng/mL to 100 ng/mL

    • Include at least 6-8 concentrations in log-scale increments

    • Based on published data, the ED50 for mouse Balb/3T3 cells is ≤ 1 ng/mL

    • For human fibroblasts, the ED50 typically ranges from 0.9-3 ng/mL

  • Time-Course Considerations:

    • Short-term signaling responses: 5, 15, 30, 60 minutes

    • Proliferation assays: 24, 48, 72 hours

    • Include time-matched controls for each concentration

  • Appropriate Readouts:

    • Immediate signaling: EGFR phosphorylation (Western blot)

    • Proliferation: CCK-8, MTT, BrdU incorporation, or cell counting

    • Migration: Scratch assay or Boyden chamber

    • Gene expression: qPCR for EGF-responsive genes

Example Protocol for Cell Proliferation Assay:

  • Seed cells in 96-well plates (2,000-5,000 cells/well)

  • Allow attachment overnight

  • Replace with serum-free medium for 24 hours

  • Treat with EGF concentrations ranging from 0.1 to 100 ng/mL

  • Incubate for 72 hours

  • Add CCK-8 solution (10 μL/well)

  • Measure absorbance at 450 nm after 1-4 hours incubation

  • Calculate ED50 using appropriate curve-fitting software

Research with 3T3 fibroblasts has demonstrated significant proliferation with an ED50 of 0.9848-2.958 ng/mL , while epithelial cells may show different sensitivity thresholds. Cell type-specific responses should be carefully documented and compared to literature values to ensure EGF activity and receptor functionality.

What validated assays are available for measuring the biological activity of recombinant human EGF?

Several validated assays are available for measuring the biological activity of recombinant human EGF, each with specific advantages and applications:

1. Cell Proliferation Assays:

  • Balb/3T3 Proliferation Assay: The gold standard for EGF bioactivity testing

    • Methodology: Cells are serum-starved and treated with various concentrations of EGF for 48-72 hours

    • Readout: Cell number quantification via metabolic assays (MTT, CCK-8)

    • Expected result: ED50 typically ≤ 1 ng/mL for active EGF

    • Advantage: Well-established with reproducible results

  • Primary Human Keratinocyte Proliferation:

    • More physiologically relevant for skin applications

    • Typically requires longer incubation periods (5-7 days)

    • Used extensively in dermatological research applications

2. Receptor Activation Assays:

  • EGFR Phosphorylation Assay:

    • Methodology: Western blot or ELISA to detect phosphorylated EGFR (Tyr1068, Tyr1173)

    • Timeframe: Acute response (5-30 minutes post-stimulation)

    • Advantage: Direct measure of receptor engagement

    • Particularly useful for structure-function studies of EGF variants

  • Phospho-ERK1/2 Activation:

    • Measures downstream signaling pathway activation

    • Can be quantified by Western blot, ELISA, or flow cytometry

    • Typically assessed 10-30 minutes after EGF stimulation

    • Provides information on signal transduction efficiency

3. Functional Cellular Assays:

  • Wound Healing/Scratch Assay:

    • Methodology: Create a "wound" in cell monolayer and measure closure rate with EGF treatment

    • Readout: Wound closure percentage at 12, 24, and 48 hours

    • Advantage: Models physiological function of EGF in wound healing

    • Particularly relevant for dermatological applications

  • Cell Migration Assay (Boyden Chamber/Transwell):

    • Quantifies directional cell migration in response to EGF gradients

    • Typically performed over 4-24 hours

    • Distinguishes chemotactic from chemokinetic effects

A comprehensive validation of EGF activity should include dose-response relationship (typically 0.1-50 ng/mL), comparison to reference standard, specificity testing (inhibition by EGFR inhibitors like gefitinib), and reproducibility assessment (intra- and inter-assay CV < 15%) .

How do different expression systems affect the structure and function of recombinant human EGF?

The choice of expression system significantly impacts the structural integrity, post-translational modifications, and biological activity of recombinant human EGF:

Expression SystemAdvantagesLimitationsImpact on EGF Properties
E. coli- High yield
- Cost-effective
- Scalable production
- No glycosylation
- Potential endotoxin contamination
- Refolding often required
- May require optimization of disulfide bond formation
- Typically requires additional purification steps
- Potential for misfolding
Mammalian (HEK293, CHO)- Proper protein folding
- Authentic post-translational modifications
- Lower endotoxin
- Higher cost
- Lower yield
- More complex production
- More native-like structure
- Enhanced stability
- Potentially higher specific activity
Yeast (P. pastoris)- Higher yield than mammalian
- Some post-translational modifications
- Secretion into medium
- Hyperglycosylation
- Non-human glycan patterns
- Generally good folding
- May have altered receptor binding kinetics
Insect cells- Post-translational modifications
- High expression levels
- Complex glycosylation patterns
- Higher cost than bacterial
- Good compromise between yield and structure

Research Findings on Expression System Effects:

  • Structural Integrity: EGF expressed in E. coli systems may require additional refolding steps to ensure proper disulfide bond formation, which is critical for bioactivity. The three intramolecular disulfide bonds (positions 6-20, 14-31, and 33-42) must form correctly .

  • Specific Activity: Studies have shown that properly folded EGF from mammalian expression systems can exhibit higher specific activity in cellular assays compared to refolded bacterial EGF, although well-optimized bacterial systems can produce equally active protein.

  • Purification Strategies: Expression system choice dictates purification approach:

    • E. coli-expressed EGF often utilizes His-tags for IMAC purification

    • Mammalian-expressed EGF may be secreted with native signal sequences

  • Stability Profiles: HEK293-expressed EGF typically shows enhanced stability in solution compared to bacterial versions, potentially due to more accurate folding and disulfide bond formation .

For critical research applications requiring highest biological fidelity, mammalian expression systems may be preferred despite higher costs, particularly for studies investigating subtle signaling differences or for applications requiring extended stability .

What methodological approaches can resolve contradictory findings in EGF signaling research?

Contradictory findings in EGF signaling research are common due to the complexity of EGFR signaling networks and experimental variables. Systematic approaches to resolve these contradictions include:

1. Standardization of Experimental Systems:

  • Cell Line Authentication and Passage Number Control:

    • Document cell line sources and authentication methods

    • Use low-passage cells (typically <10-15 passages)

    • Report EGFR expression levels quantitatively

    • Consider the influence of cell culture conditions on receptor expression

  • EGF Source and Activity Verification:

    • Use standardized bioactivity assays before experiments

    • Report source, lot number, and specific activity

    • Verify activity using dose-response curves in a standard cell line

    • Account for potential differences in recombinant EGF preparations

2. Comprehensive Signaling Analysis:

  • Temporal Resolution:

    • Contradictions often arise from different time points

    • Implement time-course experiments (5 min to 24 h)

    • Example: EGFR signaling can switch from proliferative to migratory over time

    • Document receptor internalization and recycling dynamics

  • Multi-pathway Analysis:

    • Simultaneously measure multiple downstream pathways:

      • MAPK/ERK pathway (proliferation)

      • PI3K/AKT pathway (survival)

      • STAT signaling (transcriptional regulation)

      • PLCγ/PKC pathway (migration)

3. Context-Dependent Signaling Resolution:

  • Microenvironment Characterization:

    • Document matrix components and stiffness

    • Control cell density and cell-cell contact

    • Measure autocrine/paracrine factors

    • Consider the influence of extracellular matrix proteins on signaling

  • Receptor Status Analysis:

    • Quantify EGFR expression levels

    • Assess receptor dimerization patterns (EGFR homodimers vs. heterodimers with other ErbB family members)

    • Evaluate pre-existing activation states

    • Consider the role of receptor trafficking and localization

Research has demonstrated that contradictory findings in EGF responses can often be resolved by accounting for concentration-dependent effects and receptor crosstalk. For example, at low concentrations (0.1-1 ng/mL), EGF may primarily promote proliferation, while at higher concentrations (10-100 ng/mL), it may additionally induce differentiation in the same cell type .

Additionally, the cellular context significantly influences EGF signaling outcomes. For instance, in skin cells, EGF has been shown to have both pro-inflammatory and anti-inflammatory effects depending on the pre-existing inflammatory state, with studies demonstrating anti-inflammatory effects in acne and atopic dermatitis models .

How can researchers design effective EGF delivery systems for advanced research applications?

Designing effective EGF delivery systems for research applications requires careful consideration of multiple factors to ensure optimal biological activity and experimental reproducibility:

1. Stability and Bioactivity Preservation:

  • Protection Strategies:

    • Inclusion of stabilizing excipients (trehalose, albumin, poloxamers)

    • pH optimization (typically pH 6.5-7.5)

    • Protection from proteolytic degradation

    • Antioxidants to prevent oxidation of methionine residues

  • Release Kinetics Design:

    • Sustained release profiles for chronic stimulation studies

    • Pulsatile release for temporal signaling studies

    • Controlled release rates to achieve specific concentrations

    • Mathematical modeling to predict release profiles

2. Advanced Delivery Systems for Research Applications:

Delivery SystemDesign PrinciplesResearch ApplicationsMethodological Considerations
Hydrogels- Biocompatible polymers
- Tunable release kinetics
- 3D microenvironment
- Wound healing models
- Tissue engineering
- 3D cell culture
- Crosslinking density affects release
- Polymer degradation rate
- Water content and swelling
Nanoparticles- Size control (30-200 nm)
- Surface modification
- Biodegradable materials
- Intracellular trafficking studies
- Cancer research
- Targeted delivery
- Particle characterization (size, PDI, zeta)
- Loading efficiency
- Release kinetics
Microfluidic Systems- Precise concentration control
- Gradient generation
- Dynamic delivery
- Signaling dynamics
- Cell migration studies
- Single-cell responses
- Flow rate optimization
- Shear stress effects
- Material compatibility
Surface Immobilization- Covalent attachment
- Oriented presentation
- Density control
- Cell adhesion studies
- Mechanobiology
- Spatial signaling
- Surface chemistry
- Bioactivity verification
- Density quantification

3. Targeting and Localization Approaches:

  • Receptor-Specific Targeting:

    • EGFR-binding peptides for enhanced targeting

    • Antibody-conjugated systems for cell-specific delivery

    • Consideration of receptor density and distribution

  • Spatial Control Strategies:

    • Matrix-bound presentation vs. soluble delivery

    • Gradient formation for migration studies

    • Surface immobilization techniques for localized signaling

    • Micropatterns for spatial control of signaling

Research has demonstrated that the method of EGF delivery significantly impacts cellular responses. For example, matrix-bound EGF has been shown to induce sustained EGFR signaling compared to soluble EGF, which typically results in receptor internalization and signal termination . In skin applications, controlled release formulations have demonstrated enhanced wound healing properties compared to bolus administration .

For advanced research applications, the delivery system should be carefully matched to the research question, with consideration of release kinetics, spatial presentation, and maintenance of biological activity throughout the experimental timeframe.

What are the most common causes of recombinant EGF activity loss and how can researchers address them?

Loss of EGF activity is a common challenge in research settings. Understanding the mechanisms and implementing preventive measures is crucial for experimental reproducibility:

Cause of Activity LossMolecular MechanismDetection MethodPrevention/Mitigation Strategy
Improper Disulfide Bond FormationMisfolding of the three critical disulfide bondsBioactivity assay comparison to reference standard- Use non-reducing conditions during purification
- Optimize oxidative refolding if using E. coli
- Consider mammalian expression systems
Proteolytic DegradationCleavage by proteasesSDS-PAGE showing lower MW bands- Add protease inhibitors (PMSF, aprotinin)
- Store at -80°C
- Avoid repeated freeze-thaw cycles
- Use low protein-binding tubes
Adsorption to SurfacesProtein adherence to plastic/glassConcentration measurement before/after storage- Add carrier protein (0.1% BSA/HSA)
- Use low-binding tubes
- Maintain concentration >0.1 mg/mL if possible
Oxidative DamageOxidation of methionine residuesMass spectrometry analysis- Add antioxidants (e.g., 1mM DTT)
- Minimize exposure to air
- Store under nitrogen atmosphere
AggregationFormation of inactive multimersSize exclusion chromatography
Dynamic light scattering
- Optimize buffer conditions (pH, ionic strength)
- Include stabilizers (glycerol, trehalose)
- Filter before use (0.22 μm)

Methodological Approach to Troubleshooting Activity Loss:

  • Systematic Activity Assessment:

    • Perform regular activity testing using standardized assays

    • Compare to reference standards (commercial EGF with known activity)

    • Document lot-to-lot variation and storage time effects

    • Implement quality control checkpoints throughout experiments

  • Analytical Quality Control:

    • SDS-PAGE to verify molecular weight and purity

    • Circular dichroism to assess secondary structure

    • Mass spectrometry to detect modifications

    • Thermal shift assays to evaluate stability

Research has demonstrated that EGF activity is highly dependent on proper disulfide bond formation. Studies show that the ED50 of properly folded EGF in proliferation assays should be ≤1 ng/mL for Balb/3T3 cells . Significant deviations from this value may indicate structural issues requiring investigation of storage and handling conditions.

When activity loss is detected, consider filtration to remove aggregates, buffer exchange to remove accumulated degradation products, addition of fresh stabilizers, or preparation of fresh working solutions from frozen master stock.

How can researchers validate the specificity of observed EGF effects in complex experimental systems?

Validating the specificity of EGF-mediated effects is crucial for accurate interpretation of research findings, particularly in complex experimental systems:

1. Receptor-Level Validation Approaches:

  • EGFR Inhibition Studies:

    • Pharmacological inhibitors:

      • Tyrosine kinase inhibitors (gefitinib, erlotinib) at 0.1-10 μM

      • Antibody-based inhibitors (cetuximab) at 5-20 μg/mL

    • Genetic approaches:

      • EGFR siRNA/shRNA knockdown (validate 70-90% reduction)

      • CRISPR/Cas9 knockout (complete ablation)

      • Dominant-negative EGFR mutants

  • Receptor Specificity Controls:

    • Competitive binding assays with unlabeled EGF

    • Use of non-EGFR ligands (e.g., PDGF, FGF) as negative controls

    • EGFR expression correlation with response magnitude

2. Signaling Pathway Validation:

  • Pathway-Specific Inhibition:

    • MEK inhibitors (U0126, PD98059) for ERK pathway

    • PI3K inhibitors (LY294002, wortmannin) for Akt pathway

    • STAT inhibitors for JAK/STAT signaling

    • Document pathway-specific effects

  • Phosphorylation Analysis:

    • Western blot for phospho-EGFR (Tyr1068)

    • Multiplex phospho-protein analysis (Luminex, reverse phase protein array)

    • Temporal profiling (5 min to 24 h)

3. Advanced Specificity Validation Methods:

  • Receptor Binding Dynamics: Surface plasmon resonance (SPR) or biolayer interferometry to measure binding kinetics and affinity

  • Pathway Selectivity: Phospho-proteomic analysis to identify activation of canonical vs. non-canonical pathways

  • Genetic Validation: CRISPR screens of pathway components to identify essential mediators

  • Single-Cell Analysis: Flow cytometry or single-cell RNA-seq to assess population heterogeneity in responses

Research has demonstrated that EGF specifically activates the EGFR pathway, and this specificity can be confirmed through inhibition studies. Kim et al. showed that gefitinib (an EGFR inhibitor) attenuated the anti-inflammatory effects of EGF in acne models, confirming that these effects were specifically mediated through EGFR signaling . Such validation approaches are essential for attributing observed effects specifically to EGF rather than to experimental artifacts or parallel signaling pathways.

What are the critical quality control parameters for evaluating recombinant human EGF preparations for research applications?

Comprehensive quality control of recombinant human EGF is essential for research reproducibility. A systematic approach should include the following critical parameters:

1. Purity Assessment:

  • SDS-PAGE Analysis:

    • Minimum acceptable purity: ≥95%

    • Single band at approximately 6.4 kDa under reducing conditions

    • Silver staining for highest sensitivity

  • Advanced Analytical Methods:

    • RP-HPLC (reverse-phase high-performance liquid chromatography)

    • Size exclusion chromatography (SEC)

    • Mass spectrometry for exact mass confirmation

2. Identity Confirmation:

  • Primary Structure Verification:

    • N-terminal sequencing (first 10-15 amino acids)

    • Peptide mapping after enzymatic digestion

    • Mass spectrometry for molecular weight confirmation (expected MW ~6.4 kDa)

  • Immunological Methods:

    • Western blot with EGF-specific antibodies

    • ELISA with validated antibodies

3. Biological Activity Determination:

  • Cell-Based Bioassays:

    • Proliferation assay in Balb/3T3 cells (ED50 typically ≤1 ng/mL)

    • EGFR phosphorylation in A431 cells

    • Comparison to reference standard

    • Multiple assays for comprehensive activity assessment

  • Activity Specification:

    • Specific activity (units/mg)

    • Dose-response relationship

    • Lot-to-lot consistency evaluation

4. Contaminant Analysis:

Contaminant TypeTest MethodAcceptance CriteriaRelevance to Research
EndotoxinLAL test (Limulus Amebocyte Lysate)<0.1 EU/μg protein Prevents inflammatory response artifacts
Host Cell ProteinsELISA, Western blotTypically <100 ng/mgMinimizes non-specific effects
DNAqPCR<10 ng/mg proteinPrevents transfection artifacts
Microbial ContaminationSterility testingNo growthEssential for cell culture applications
AggregatesSEC, DLS<5% high molecular weight speciesEnsures consistent bioactivity

Research has shown that quality control parameters significantly impact experimental reproducibility. For example, studies have demonstrated that endotoxin contamination can confound results in inflammation studies, while aggregated protein can show reduced activity in proliferation assays .

A complete Certificate of Analysis should include lot number, production date, expression system used, purity percentage, endotoxin level, biological activity (with method and ED50), stability data, recommended storage conditions, sequence confirmation method, and formulation composition.

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.