Sf9 cells, derived from Spodoptera frugiperda pupal ovarian tissue, are favored for recombinant protein production due to their capacity for post-translational modifications and high growth rates in suspension cultures . The baculovirus expression vector system (BEVS) is typically employed:
Workflow: Viral infection → Protein expression → Purification via His-tag chromatography .
Advantages:
Critical Parameters:
EPGN stabilizes unique EGFR dimers, distinct from those induced by EGF or epiregulin, leading to biased agonism . Key findings:
Induces phosphorylation of c-erbB-1 and MAP kinases in epithelial cells .
Acts as a partial agonist with reduced receptor dimerization strength compared to EGF .
Prolonged mitogenic activity due to delayed receptor ubiquitylation and endocytosis .
Mitogenicity: Stimulates proliferation of murine Balb/3T3 cells (ED₅₀ < 300 ng/mL) .
Cancer Research: Overexpressed in tumor cells (e.g., liver, testis), correlating with uncontrolled proliferation .
Vaccine Development: Sf9-produced EPGN is used in rabies virus-like particle (VLP) vaccines .
Epithelial mitogen, EPG, Epithelial Mitogen Homolog (Mouse), Epithelial Mitogen Homolog, ALGV3072, PRO9904, Epigen, EPGN.
Sf9, Insect cells.
ADPAAVTVTP PITAQQGNWT VNKTEADNIE GPIALKFSHL CLEDHNSYCI NGACAFHHEL EKAICRCFTG YTGERCEHLT LTSYAVDSYE KHHHHHH.
EPGN (Epigen) is an EGF-related polypeptide growth factor that signals through the ErbB receptor-1 (ErbB1). It is naturally produced in multiple tissues, including testis, liver, heart, and in certain tumor cells. EPGN functions as a mitogenic agent for fibroblasts and epithelial cells despite having relatively low affinity for its main receptor .
The enhanced mitogenic potential of EPGN is attributed to inefficient receptor ubiquitylation and endocytosis, which prolongs signaling activity. When binding to its receptor, EPGN stimulates phosphorylation of c-erbB-1 and MAP kinases in epithelial cells, activating downstream signaling cascades that promote cell proliferation .
Human EPGN is initially synthesized as a glycosylated 14.7 kDa transmembrane precursor protein, which undergoes proteolytic cleavage to produce a mature soluble sequence. The protein contains a conserved sequence of six cysteine residues and two N-linked glycosylation sites, with two hydrophobic regions comprised of a signal sequence and a transmembrane domain .
For optimal stability of EPGN Human, Sf9, researchers should follow these evidence-based storage and handling protocols:
Storage Duration | Recommended Conditions | Additional Notes |
---|---|---|
Short-term (2-4 weeks) | Store at +4°C | Only if entire vial will be used within this timeframe |
Long-term | Store at -20°C | Keep frozen for extended storage periods |
Extended storage | -20°C with carrier protein | Add 0.1% HSA or BSA as a stabilizing agent |
The recombinant protein is typically provided as a sterile filtered colorless solution at a concentration of 0.5 mg/ml in Phosphate Buffered Saline (pH 7.4) containing 10% glycerol .
Multiple freeze-thaw cycles should be strictly avoided as they can lead to protein denaturation and loss of biological activity. If working with the protein over multiple sessions, it is recommended to prepare smaller aliquots before freezing to minimize the number of freeze-thaw cycles .
The purity of commercially available EPGN Human, Sf9 is typically greater than 90% as determined by complementary analytical techniques. The primary methods used for purity assessment include:
SDS-PAGE (Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis): This technique separates proteins based on their molecular weight. For EPGN Human, Sf9, a single band should appear at approximately 13.5-18 kDa, which is higher than the calculated molecular weight (10.8 kDa) due to glycosylation .
HPLC (High-Performance Liquid Chromatography): This chromatographic method provides a quantitative assessment of protein purity based on differences in physical or chemical properties. HPLC analysis complements SDS-PAGE by detecting impurities that might not be visible on gels .
Mass Spectrometry: While not routinely used for commercial quality control, mass spectrometry can provide precise information about the molecular weight and potential modifications of the protein.
For research applications requiring exceptionally high purity, additional purification steps may be necessary. Researchers should consider the specific requirements of their experimental system when determining the acceptable purity level.
When designing experiments with EPGN Human, Sf9, researchers should consider several methodological factors to ensure robust and reproducible results:
Concentration optimization: Determine the effective dose range through dose-response experiments. Typical working concentrations range from 1-100 ng/ml, but optimal concentrations may vary depending on the cell type and the specific biological response being measured.
Receptor expression verification: Prior to experiments, confirm that your target cells express the ErbB1 receptor at levels sufficient for EPGN signaling. This can be done using Western blotting, flow cytometry, or immunocytochemistry.
Serum considerations: The presence of serum in culture media may contain growth factors that could interfere with EPGN activity. Consider using serum-free or serum-reduced conditions during the treatment period.
Positive controls: Include EGF or other well-characterized ErbB1 ligands as positive controls to validate receptor functionality and experimental conditions.
Treatment duration: EPGN has been shown to induce prolonged signaling due to inefficient receptor ubiquitylation and endocytosis. Time-course experiments are recommended to determine optimal treatment duration for your specific experimental endpoints .
Signaling readouts: Common downstream readouts for EPGN activity include phosphorylation of ERK1/2, AKT, and STAT proteins, which can be measured by Western blotting or phospho-specific ELISAs.
The use of Sf9 insect cells for EPGN expression has several important implications for the protein's structure and function:
Glycosylation patterns: Sf9 cells produce proteins with simpler glycosylation patterns compared to mammalian cells. This can affect receptor binding, stability, and immunogenicity. While the core biological activity is usually preserved, subtle differences in potency or receptor interaction kinetics may exist .
Post-translational modifications: Beyond glycosylation, other post-translational modifications might differ between insect and mammalian expression systems. This could potentially impact protein folding, stability, or specific functional aspects.
Protein folding and disulfide bonds: Sf9 cells are capable of forming disulfide bonds, which are critical for the proper folding and function of EPGN. The six conserved cysteine residues in EPGN form disulfide bonds that maintain the protein's tertiary structure .
Scale and yield advantages: The Sf9 baculovirus expression system offers advantages in terms of production scale and yield. Recent advances in stable Sf9 cell lines have improved the efficiency of recombinant protein production by 10-fold compared to traditional methods .
His-tag considerations: The C-terminal histidine tag used for purification may potentially influence certain binding or functional properties. Control experiments comparing tagged versus untagged protein may be necessary for certain applications.
When designing experiments with Sf9-produced EPGN, researchers should consider these factors and include appropriate controls to account for potential system-specific effects.
EPGN activates several signaling pathways through its interaction with the ErbB1 receptor. Key pathways and their measurement methods include:
Signaling Pathway | Key Phosphorylation Events | Measurement Methods | Typical Timeframe |
---|---|---|---|
MAPK/ERK | ERK1/2 (Thr202/Tyr204) | Western blot, phospho-ELISA, cell-based ELISA | 5-30 minutes (rapid, transient) |
PI3K/AKT | AKT (Ser473, Thr308) | Western blot, phospho-ELISA | 15-60 minutes |
STAT | STAT1 (Tyr701), STAT3 (Tyr705) | Western blot, phospho-flow cytometry | 15-60 minutes |
PLCγ/PKC | PLCγ (Tyr783), PKC isoforms | Western blot, calcium flux assays | 2-15 minutes (very rapid) |
For comprehensive signaling analysis, consider these methodological approaches:
Phosphoproteomics: Mass spectrometry-based phosphoproteomics can provide an unbiased view of all phosphorylation changes induced by EPGN.
Multiplexed assays: Phospho-protein arrays or bead-based multiplex assays allow simultaneous measurement of multiple phosphorylation events.
Live-cell imaging: Fluorescent reporters or FRET-based biosensors can be used to monitor signaling dynamics in real-time.
Inhibitor studies: Pathway-specific inhibitors can help delineate the contribution of individual pathways to biological responses.
RNA-seq: Transcriptional profiling at various time points following EPGN treatment can reveal the downstream effects of signaling pathway activation .
When studying EPGN signaling, it's important to note that the protein's enhanced mitogenic potential is attributed to inefficient receptor ubiquitylation and endocytosis, which may result in prolonged signaling compared to other EGF family members .
When designing receptor binding studies with EPGN Human, Sf9, consider the following methodological approaches:
Direct binding assays:
Radiolabeled ligand binding using 125I-labeled EPGN
Fluorescently labeled EPGN (with careful validation that labeling doesn't interfere with binding)
Surface Plasmon Resonance (SPR) using purified ErbB1 receptor
Competition binding assays:
Using a well-characterized ligand (e.g., EGF) as the labeled ligand
Determining IC50 values for EPGN by competition
Buffer conditions:
pH: Typically 7.4 (physiological)
Salt concentration: 150 mM NaCl (physiological)
Presence of divalent cations: Include Ca2+ and Mg2+ (1-2 mM)
Detergents: Low concentrations (0.01-0.05%) of non-ionic detergents may reduce non-specific binding
Blocking agents: BSA (0.1-1%) to minimize non-specific binding
Experimental controls:
Positive control: EGF binding
Negative control: Irrelevant growth factor or heat-denatured EPGN
Non-specific binding: Determined by adding excess unlabeled ligand
Kinetic parameters to determine:
Kon (association rate constant)
Koff (dissociation rate constant)
KD (equilibrium dissociation constant)
Bmax (maximum binding capacity)
When analyzing binding data, consider that EPGN has been reported to have lower affinity for ErbB1 compared to EGF, but its enhanced mitogenic potential is attributed to inefficient receptor ubiquitylation and endocytosis, which extends signaling duration rather than initial binding strength .
EPGN has been found in certain tumor cells and functions as a mitogen, making it relevant for cancer research. When designing experiments to study cancer cell proliferation using EPGN Human, Sf9, consider these methodological approaches:
Proliferation assay selection:
MTT/MTS/WST-1 assays: Measure metabolic activity as a proxy for cell number
BrdU incorporation: Directly measures DNA synthesis
Ki-67 immunostaining: Identifies actively proliferating cells
Colony formation assays: Assess long-term proliferative capacity
Real-time cell analysis: Monitors proliferation continuously
Experimental design considerations:
Cell density optimization: Seed cells at subconfluent densities to allow room for proliferation
Serum conditions: Reduce serum concentration (0-2%) to minimize background proliferation
Treatment duration: 24-72 hours depending on cell doubling time
Dose-response: Test EPGN concentrations ranging from 0.1-100 ng/ml
Essential controls:
Positive control: EGF or serum stimulation
Negative control: Serum-free or low-serum media alone
Receptor blocking: ErbB1 blocking antibodies or small molecule inhibitors
Downstream inhibition: MEK inhibitors (U0126, PD98059) or PI3K inhibitors (LY294002, wortmannin)
Advanced analyses:
Cell cycle analysis by flow cytometry to determine G1/S transition effects
Combination treatments with chemotherapeutic agents to assess synergy or antagonism
Comparison of effects across cell lines with varying ErbB1 expression levels
RNA interference or CRISPR-based approaches to validate receptor dependency
Researchers should note that cancer cells often have altered expression or regulation of ErbB receptors, which may influence their response to EPGN. Characterizing the receptor expression profile of the cell lines being studied is essential for proper interpretation of results.
Researchers working with EPGN Human, Sf9 may encounter several challenges. Here are common issues and evidence-based solutions:
Issue | Possible Causes | Solutions |
---|---|---|
Low or no biological activity | Protein denaturation during storage/handling | Avoid freeze-thaw cycles; add carrier protein (0.1% BSA) |
Receptor absence or dysfunction in target cells | Verify ErbB1 receptor expression and functionality | |
Interference from media components | Use serum-free or reduced-serum conditions during treatment | |
Inconsistent results across experiments | Variability in protein aliquots | Prepare single-use aliquots; standardize freeze-thaw protocols |
Cell density variations | Standardize cell seeding protocols; optimize cell density | |
Passage number effects | Use cells within a defined passage range | |
Poor solubility or aggregation | Improper reconstitution | Follow recommended reconstitution protocols; avoid vortexing |
Protein concentration too high | Dilute in appropriate buffer containing carrier protein | |
Buffer incompatibility | Test different buffer systems if compatibility issues arise | |
Non-specific effects | Endotoxin contamination | Use endotoxin-tested preparations; include polymyxin B controls |
His-tag interference | Compare with untagged protein for critical applications |
When troubleshooting EPGN activity, it's important to include proper positive controls (such as EGF) and to verify that your experimental system can detect the expected biological responses. For receptor activation studies, phospho-specific antibodies against ErbB1 and downstream signaling molecules can help confirm that the protein is biologically active.
To ensure the recombinant EPGN is functionally active before use in critical experiments, consider these validation approaches:
Receptor phosphorylation assay:
Treat ErbB1-expressing cells (e.g., A431, MCF-7) with EPGN
Analyze ErbB1 phosphorylation by Western blotting using phospho-specific antibodies
Expected result: Dose-dependent increase in receptor phosphorylation
Proliferation validation:
Perform proliferation assays using responsive cell lines (fibroblasts, epithelial cells)
Include dose-response analysis to determine EC50
Compare potency with commercially available standards
Expected result: Dose-dependent increase in proliferation
Signaling cascade activation:
Assess ERK1/2 and AKT phosphorylation at various time points
Compare activation kinetics with EGF
Expected result: Activation of both pathways, potentially with different kinetics than EGF
Receptor specificity confirmation:
Pre-treat cells with ErbB1-specific inhibitors (gefitinib, erlotinib) or blocking antibodies
Determine if EPGN effects are abolished
Expected result: Inhibition of EPGN-induced effects by ErbB1 blockade
Structural integrity assessment:
Circular dichroism (CD) spectroscopy to assess secondary structure
Size exclusion chromatography to detect aggregation
Expected result: Properly folded protein with minimal aggregation
When validating EPGN activity, remember that its binding affinity to ErbB1 is reported to be lower than that of EGF, but it induces prolonged signaling due to inefficient receptor internalization mechanisms .
EPGN is expressed in various tissues during development, making it relevant for developmental biology studies. Methodological approaches for such research include:
Tissue-specific expression analysis:
Immunohistochemistry or in situ hybridization to map EPGN expression patterns
qPCR analysis of EPGN expression during developmental stages
Compare with other EGF family members to identify unique developmental roles
Ex vivo tissue culture systems:
Organ explant cultures treated with EPGN
Assess morphological changes, proliferation patterns, and differentiation markers
Use time-lapse imaging to track developmental processes in real-time
Stem cell differentiation studies:
Investigate EPGN's role in stem cell maintenance or differentiation
Combine with other growth factors in defined differentiation protocols
Analyze lineage-specific marker expression following EPGN treatment
Conditional expression/knockout models:
Generate inducible EPGN expression systems in relevant cell lines
Create conditional knockout models to study tissue-specific requirements
Use CRISPR/Cas9 to introduce mutations in EPGN or its receptor
When designing developmental biology experiments with EPGN Human, Sf9, consider that the insect cell-produced protein may have different glycosylation patterns compared to endogenous EPGN, which could potentially affect certain developmental processes that are sensitive to these modifications.
When conducting comparative studies between EPGN and other EGF family members, consider these methodological approaches:
Receptor binding profile comparison:
Compare binding affinities to ErbB1 and other ErbB family receptors
Assess competition binding between EPGN and other family members
Evaluate receptor dimerization patterns induced by different ligands
Signaling dynamics analysis:
Compare temporal phosphorylation patterns of shared downstream targets
Assess receptor internalization and degradation kinetics
Identify signaling pathways uniquely activated by EPGN versus other family members
Biological response profiling:
Compare dose-response relationships for proliferation, migration, and differentiation
Assess cell type-specific responses to different family members
Evaluate combinatorial effects of multiple EGF family ligands
Structural and functional relationship studies:
Analyze structure-function relationships through chimeric proteins
Identify domains responsible for specific activities through mutagenesis
Use computational modeling to predict and test binding interactions
When conducting comparative studies, it's important to standardize experimental conditions, including:
Using the same cell lines and passage numbers
Employing equimolar concentrations of ligands rather than mass-based concentrations
Standardizing storage and handling procedures across all proteins being compared
Including appropriate controls for each family member
The unique feature of EPGN compared to other EGF family members is its combination of lower receptor binding affinity but enhanced mitogenic potential due to reduced receptor downregulation following activation .
Based on current research trends, several emerging applications for EPGN Human, Sf9 show particular promise:
Cancer research: Given EPGN's expression in certain tumor cells and its mitogenic properties, further investigation into its role in cancer progression and potential as a therapeutic target is warranted. Particular interest lies in understanding how EPGN's unique signaling properties might contribute to tumor growth and therapy resistance.
Regenerative medicine: EPGN's mitogenic effects on epithelial cells suggest potential applications in wound healing and tissue regeneration. Researchers might explore EPGN in combination with biomaterials or other growth factors for enhanced tissue repair.
Stem cell technologies: The role of EPGN in regulating proliferation makes it relevant for stem cell expansion protocols. Further research could optimize EPGN concentrations and combinations with other factors for efficient ex vivo expansion of stem cells.
Comparative signaling studies: Understanding the unique aspects of EPGN signaling compared to other EGF family members could reveal new insights into receptor biology and signal transduction mechanisms.
Structure-function studies: The availability of recombinant EPGN enables detailed structure-function analyses through protein engineering and mutagenesis, potentially revealing new therapeutic approaches targeting ErbB receptor signaling.
Epigen is originally synthesized as a glycosylated transmembrane precursor protein with a molecular mass of approximately 14.7 kDa. This precursor undergoes proteolytic cleavage to produce a mature, soluble sequence . The recombinant form of Epigen produced in Sf9 insect cells is a single, glycosylated polypeptide chain containing 97 amino acids and has a molecular mass of 10.8 kDa . It is expressed with a 6-amino acid His tag at the C-terminus and is purified using proprietary chromatographic techniques .
Epigen is a potent mitogen for fibroblasts and epithelial cells . It is produced in various tissues, including the testis, liver, heart, and certain tumor cells . The biological activity of Epigen is determined by its ability to stimulate the proliferation of cells, such as murine Balb/3T3 cells, in a dose-dependent manner .
Epigen (Human Recombinant, Sf9) is primarily used for laboratory research purposes. It is not intended for use as a drug, agricultural or pesticidal product, food additive, or household chemical . The protein is supplied as a sterile filtered colorless solution, formulated in phosphate-buffered saline (PBS) with 10% glycerol . For long-term storage, it is recommended to add a carrier protein such as human serum albumin (HSA) or bovine serum albumin (BSA) to prevent multiple freeze-thaw cycles .