Recombinant Human Epidermal Growth Factor (EGF), partial (Active) (GMP)

Shipped with Ice Packs
In Stock

Description

Biological Activity

Mechanism of Action
EGF binds to the 170 kDa EGFR tyrosine kinase receptor, initiating:

  • RAS-RAF-MEK-ERK pathway activation

  • PI3K-AKT and STAT signaling

  • NF-κB cascade modulation

Potency Metrics

Cell LineED₅₀ (Proliferation)Specific ActivitySource
Balb/3T3 fibroblasts20–100 pg/mL>8.0×10⁵ IU/mg
NIH 3T3 fibroblasts0.1–0.6 ng/mL1.0×10⁷ IU/mg
HaCaT keratinocytes0.1 ng/mL5.49×10⁶ IU/mg (typical)

Functional Outcomes

  • ↑ Collagen I/III synthesis in aged fibroblasts

  • ↓ MMP-9 expression (anti-photoaging)

  • ↑ Filaggrin/occludin production (skin barrier repair)

Clinical & Cosmetic Applications

Therapeutic Uses

  • Wound Healing: Accelerates re-epithelialization in burns and diabetic ulcers

  • Dermatology:

    • Reduces EGFR inhibitor-induced skin toxicity (Phase III trial success)

    • Improves atopic dermatitis via Th17 response modulation

  • Oncology Support: Mitigates chemotherapy-induced mucositis

Aesthetic Applications

ParameterImprovement Rate (Clinical Trials)DurationSource
Periocular wrinkles85% (20 patients)4 weeks
Skin texture/pigmentation72% (29 subjects)3 months
Hyaluronic acid synthesis3.8-fold increase (in vitro)

Emerging Research Findings

  • Fusion Protein Engineering: HaloTag-rhEGF improves solubility while retaining bioactivity (NIH 3T3 viability ↑ 2.1-fold)

  • Organoid Development: Critical for intestinal/stem cell expansion at 50–100 ng/mL

  • Biomarker Potential: Serum EGF levels correlate with renal disease progression (r=0.82, p<0.001)

Product Specs

Buffer
Lyophilized from a 0.2 μm filtered concentrated solution in phosphate-buffered saline (PBS), pH 7.4.
Form
Lyophilized powder
Lead Time
Product shipment typically occurs within 5-10 business days of order receipt. Delivery times may vary depending on the shipping method and destination. Please consult your local distributor for precise delivery estimates.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to pellet the contents. Reconstitute the protein in sterile, deionized water to a final concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and storing in aliquots at -20°C or -80°C. Our standard protocol uses 50% glycerol; this may serve as a guideline for your reconstitution.
Shelf Life
Shelf life depends on several factors, including storage conditions, buffer composition, temperature, and the inherent stability of the protein. Generally, liquid formulations have a shelf life of 6 months at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life under the same storage conditions.
Storage Condition
Upon receipt, store at -20°C or -80°C. Aliquot for multiple use to prevent 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
> 98 % by SDS-PAGE and HPLC analyses.
Research Area
Cancer
Source
E.Coli
Species
Homo sapiens (Human)
Target Names
EGF
Uniprot No.

Target Background

Function

Epidermal Growth Factor (EGF) stimulates the growth of various epidermal and epithelial tissues in vivo and in vitro, as well as some fibroblasts in cell culture. It functions as a magnesiotropic hormone, stimulating magnesium reabsorption in the renal distal convoluted tubule via engagement of the epidermal growth factor receptor (EGFR) and activation of the magnesium channel TRPM6. EGF can also induce neurite outgrowth in the motoneurons of the pond snail Lymnaea stagnalis in vitro.

Gene References Into Functions
  • Our findings demonstrate the exceptional efficacy of the chimeric EGFETA toxin against EGFR-positive cancers, suggesting its potential for further development as a targeted therapy for EGFR-positive tumors resistant to monoclonal antibodies. PMID: 30226622
  • These results indicate a potential role for EGF in promoting hepatocellular carcinoma (HCC) metastasis, revealing a novel pathway for fibronectin (FN) regulation and suggesting potential targets for HCC prevention and treatment. PMID: 29315755
  • Abnormally elevated expression of EGF and transforming growth factor-alpha (TGF-α) is strongly associated with the development and progression of chronic pancreatitis and pancreatic cancer. PMID: 29125273
  • Estrogen-related receptor α (ERRα) positively regulates cell proliferation, migration, and invasion in colon cancer cells. Suppression of ERRα completely abrogates EGF-induced proliferation in these cells. PMID: 30185207
  • EGF significantly upregulates RFPL3 and hTERT protein levels in non-small cell lung cancer cells. This upregulation is attenuated by pretreatment with AG1478 and erlotinib. EGF promotes proliferation and inhibits apoptosis; PD98059 decreases RFPL3 and hTERT protein expression; and RFPL3 overexpression increases hTERT expression and related MEK pathway proteins. PMID: 29749533
  • We have identified a novel N-glycosylation site (N-72) crucial for EGF-induced migration via matrix metalloproteinase 2 (MMP2) targeting in human amnion mesenchymal stem cells (hAMSCs). PMID: 29734654
  • The spleen regulates hematopoietic stem cell function in cirrhotic hypersplenism by modulating EGF signaling. PMID: 29721775
  • Following HIP1 silencing via siRNA, EGFR endocytosis is accelerated in an EGF concentration-dependent manner, colocalizing with clathrin expression. These data suggest that HIP1 inhibition accelerates EGFR endocytosis and degradation. PMID: 29039605
  • EGF promotes aggressiveness of gastric cancer cells by inducing epithelial-to-mesenchymal transition (EMT) through activation of the ERK1/2 pathway and subsequent upregulation of urokinase-type plasminogen activator receptor (uPAR). PMID: 28849196
  • The EGF system acts as a mechanosensitizer in bone marrow stromal cells. PMID: 28843157
  • EGF counteracts the effects of trans-activator of transcription (Tat) on the modulation of human endogenous retroviruses of the W family in astrocytes. PMID: 28474333
  • Fourier-transform infrared (FTIR) spectroscopy reveals decreasing disordered structures and turns, and increasing loops in EGF throughout the synthesis process (unconjugated, post-treatment with α-lipoic acid, attached to gold nanoparticles, and bound to the bifunctional nanoprobe). The final product shows an overall (though non-linear) increase in β-sheets compared to pure EGF. PMID: 29122663
  • EGF-mediated lysosome trafficking, protease secretion, and invasion are regulated by p38 mitogen-activated protein kinase (MAPK) and sodium hydrogen exchangers (NHEs). EGF stimulates anterograde lysosome trafficking via a distinct mechanism compared to hepatocyte growth factor (HGF), indicating redundant signaling pathways controlling lysosome positioning. PMID: 28978320
  • Local EGF infiltration overcomes limitations of topical application in the hostile microenvironment of diabetic chronic wounds, improving therapeutic prospects. Clinical and basic research support the importance of local growth factor infiltration for wound healing. PMID: 28904952
  • This study provides initial evidence linking the EGF rs2298999 C/T polymorphism to gout. PMID: 27506295
  • Increased EGFR expression in patients with seborrheic keratosis (SK) and type 2 diabetes mellitus (DM2) is linked to insulin resistance and hyperinsulinemia, where dysregulation of insulin signaling affects EGF synthesis and the cell growth-regulating pathway. PMID: 28791994
  • A novel EGFR-NF-κB-FOXC1 signaling axis is crucial for breast lobular carcinoma (BLBC) cell function. PMID: 28629477
  • EGFR pathway gene expression analysis reveals that ΔNp63 alters EGFR-regulated genes involved in cell adhesion, migration, and angiogenesis. EGF addition or EGFR antibody neutralization demonstrates that EGFR activation mediates ΔNp63-induced loss of cell adhesion. PMID: 28349272
  • EGF upregulates CCL2 expression in head and neck squamous cell carcinoma (HNSCC) cells, recruiting monocytes and polarizing them into M2-like macrophages, thereby establishing a positive feedback paracrine loop. PMID: 27888616
  • This study demonstrates that EGF induces epithelial-mesenchymal transition (EMT) via the phospho-Smad2/3-Snail signaling pathway in breast cancer cells. PMID: 27829223
  • EGF and tumor necrosis factor-alpha (TNFα) cooperatively enhance HCC cell motility primarily via NF-κB/p65-mediated synergistic induction of FN in vitro. These findings highlight the interplay between EGF and TNFα in promoting HCC and identify potential therapeutic targets. PMID: 28844984
  • EGF promotes EMT in colorectal cancer cells, enhances invasion/migration, and stimulates phosphorylation of ezrin at Tyr353. PMID: 28535417
  • Molecular dynamics simulations reveal distinct dynamic properties of the human epidermal growth factor receptor (hEGFR) soluble extracellular domains (sECD):EGF complex at different pH levels, suggesting a higher activation propensity at pH 8.5. PMID: 27179806
  • Tear fluid levels of EGF and IP-10 are significantly elevated, while GRO levels are lower, in HIV-positive patients with dry eye disease (DED) compared to immunocompetent DED patients. PMID: 27585367
  • Surfactant protein A1 (SPA1) interferes with EGF binding to EGFR in pulmonary alveoli cell lines. SPA1 directly binds the EGFR extracellular domain; this binding differs from that of surfactant protein D (SPD); and SPA1 binding does not suppress EGF-induced EGFR phosphorylation or cell proliferation. PMID: 28972165
  • The interplay between EGF and amphiregulin (AREG) in airway basal cell stem/progenitor cells is involved in the pathogenesis of smoking-related lesions in the human airway epithelium. PMID: 27709733
  • Caspase-3 inhibitors suppress the EGF-F9-induced attenuation of cell adhesion and p38 MAPK phosphorylation. EGF-F9 activates apoptotic signals and induces de-adhesion in a caspase-3-dependent manner. PMID: 27129300
  • CDK1/2 are involved in regulating constitutive pre-mRNA splicing upon EGF stimulation in MDA-MB-468 cells. PMID: 27109354
  • The EGF rs4444903 GG genotype is associated with increased susceptibility to HCV-related liver cirrhosis and hepatocellular carcinoma in the Chinese Han population. PMID: 28397482
  • TGF-β opposes EGF-mediated sensitization to TRAIL-induced caspase-8 activation and apoptosis in non-transformed breast epithelial cells. EGF and TGF-β finely regulate TRAIL sensitivity, which may be relevant during morphogenesis. PMID: 27208428
  • EGFR gene amplification can be maintained and modulated by varying EGF concentrations in in vitro glioblastoma multiforme models. PMID: 28934307
  • The EGF61 rs4444903 GA genotype is associated with a decreased risk of non-syndromic cleft lip with or without cleft palate. These data further support the role of EGF61 variations in the development of non-syndromic cleft lip with or without cleft palate. PMID: 28906376
  • EGF rapidly downregulates LINC01089 (renamed LncRNA Inhibiting Metastasis; LIMT) expression by enhancing histone deacetylation at its promoter. PMID: 27485121
  • EGF-induced, calpain-mediated proteolysis contributes to the rapid degradation of cyclin G2, with the PEST domain being critical for EGF/calpain action. PMID: 28640887
  • Salivary EGF levels are significantly increased during the acute phase of natural rotavirus infection. PMID: 28558652
  • This study identified the roles of signaling pathways in regulating EGF-induced vimentin expression in MDA-MB-468 breast cancer cells. PMID: 27163529
  • miR-223 downregulates EGF expression, decreasing EGFR activation on target cells and dampening the positive EGF-EGFR autocrine/paracrine stimulation loop during post-surgical wound healing. PMID: 26876200
  • No significant difference in EGFR and EGF expression was observed between placentas from normal pregnancies and those complicated by preeclampsia. PMID: 27657362
  • Atomistic molecular dynamics simulations indicate that N-glycosylation of the EGFR extracellular domain is critical for binding of growth factors, monoclonal antibodies, and dimeric partners to the monomeric EGFR extracellular domain. PMID: 28486782
  • CMTM3 reduces EGFR expression, promotes EGFR degradation, and inhibits EGF-mediated tumorigenicity in gastric cancer cells by enhancing Rab5 activity. PMID: 27867015
  • EGF promotes adipose stem cell proliferation, delays senescence, and maintains differentiation potential, likely through EGF-induced STAT pathway activation. PMID: 28746211
  • The interaction between STS-1 and ShcA is regulated in response to EGFR activation. PMID: 28690151
  • Insulin induces sustained Akt activity, while EGF or PDGF-AA cause transient signaling; PDGF-BB produces sustained responses at high concentrations. Transient EGF responses result from receptor-level negative feedback, while combined IGF-I exposure causes full Akt activation. PMID: 27044757
  • Varying bFGF and EGF concentrations during neural rosette propagation affect the identity of resulting neural cells. PMID: 27321088
  • F25P preproinsulin reduces blood levels of EGF, VEGF, and MMP-9 in EGFR-mutant glioblastoma-bearing mice. PMID: 27317648
  • The conformational stability of EGFR is influenced by glycosylation, dimerization, and EGF binding. PMID: 28019699
  • Differential expression of EGF, EGFR, and ERBB4 is crucial for epithelial restitution and remodeling in nasal epithelium. PMID: 27285994
  • Phosphorylation and immunohistochemical assays confirm the bioactivity of soybean-produced EGF, comparable to commercially available human EGF, demonstrating the feasibility of using soybeans as a biofactory for therapeutic protein production. PMID: 27314851
  • Activated platelets release ADAMDEC1, which cleaves pro-EGF to generate soluble, active high-molecular-weight EGF (HMW-EGF). Proteolytic cleavage at the C-terminal arginyl residue of the EGF domain is the rate-limiting step in this process. PMID: 28455445
  • In a Slovak population, the EGF G61G genotype and G allele showed a non-significantly increased risk of major depressive disorder (MDD). PMID: 27755861
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 are the structural characteristics of recombinant human EGF and how do they influence its biological activity?

Recombinant human EGF is a 53 amino acid polypeptide with a molecular weight of approximately 6.7 kDa as analyzed by SEC-MALS, functioning primarily as a monomer . The protein's biological activity critically depends on its three-dimensional structure, particularly the correct formation of three disulfide bonds that create the characteristic EGF domain responsible for receptor interaction .

Methodology for structure verification typically includes:

  • Limited proteolysis to assess folding stability

  • Light scattering to confirm monomeric state

  • Circular dichroism to analyze secondary structure elements

  • NanoDSF (Differential Scanning Fluorimetry) to evaluate thermal stability

Correctly folded recombinant EGF exhibits high stability and maintains activity at concentrations as low as 5 ng/ml in cell proliferation assays . Misfolding or incomplete disulfide bond formation significantly impairs receptor binding capacity and downstream signaling efficacy.

How is recombinant human EGF optimally produced in prokaryotic expression systems?

Production of biologically active recombinant human EGF in prokaryotic systems, particularly Escherichia coli, presents significant challenges due to the protein's disulfide bond requirements. A reliable methodology involves several critical steps:

  • Vector design with appropriate signal sequences for periplasmic expression

  • Optimization of expression conditions (temperature, IPTG concentration, induction time)

  • Harvesting procedures that preserve native conformation

  • Purification protocols typically involving:

    • Initial capture via affinity chromatography

    • Intermediate purification using ion exchange chromatography

    • Polishing steps with size exclusion chromatography

  • Refolding protocols if inclusion bodies form

  • Quality control assessments including:

    • SDS-PAGE under reducing conditions (should show a single band at ~6 kDa)

    • Bioactivity testing in established cell lines (e.g., Balb/3T3 fibroblasts)

This approach can achieve highly pure, correctly folded EGF with demonstrated bioactivity in cellular assays . The protocol must account for the frequent formation of inclusion bodies and establish conditions that maximize the yield of soluble, properly folded protein with complete disulfide bond formation.

What are the standard bioactivity assays for recombinant human EGF and how should results be interpreted?

Standard bioactivity assays for recombinant human EGF focus on its ability to stimulate cell proliferation, migration, and differentiation. The most commonly employed methods include:

Cell Proliferation Assays:

  • Balb/3T3 mouse embryonic fibroblast assay (standard reference method)

  • Effective dose (ED50) typically falls between 20-100 pg/mL

  • Quantification via direct cell counting, MTT/XTT assays, or BrdU incorporation

Wound Healing/Migration Assays:

  • Scratch assay methodology using epithelial cell monolayers

  • Time-lapse microscopy to measure gap closure rates

  • Quantification of migration distance/area over 24-72 hours

Receptor Activation Assays:

  • Phosphorylation of EGFR via Western blotting

  • ELISA-based receptor phosphorylation detection

  • Downstream signaling pathway activation (ERK1/2, AKT, etc.)

Results interpretation should consider dose-dependency, time-course effects, and comparison to reference standards. Activity is typically expressed as relative potency compared to a standard EGF preparation, with detailed statistical analysis of dose-response curves.

What strategies exist for immobilizing EGF on biomaterials while preserving its bioactivity?

Immobilization of EGF on biomaterials for tissue engineering applications requires careful consideration of orientation, flexibility, and surface accessibility. Research has identified several effective approaches:

Carrier Selection and Activation Methods:
Two carriers have demonstrated particular efficacy:

  • Cyanogen bromide (CNBr)-activated-agarose (100% immobilization yield)

  • Glyoxyl-agarose (12% immobilization yield)

Orientation Considerations:
The immobilization strategy must preserve appropriate orientation for receptor binding. Peptide mapping using LC-MS can identify which residues are involved in immobilization and assess flexibility and surface accessibility of the immobilized EGF .

Bioactivity Assessment Protocol:

  • Seed fibroblast cells with 10-100 ng/mL of immobilized EGF

  • Incubate for 24-72 hours

  • Measure proliferation compared to soluble EGF controls

  • Analyze dose-dependency and time-course effects

Research findings indicate that immobilized EGF shows significantly higher cell proliferative activity at 50-100 ng/mL compared to both control and soluble EGF. CNBr-agarose-EGF demonstrates particularly high activity at 100 ng/mL with 72 hours incubation .

This approach enables development of bioactive scaffolds that provide sustained EGF signaling for applications in wound healing and tissue regeneration.

How can EGF signaling dynamics be modeled and experimentally validated in complex cellular systems?

Modeling and experimental validation of EGF signaling dynamics in complex cellular systems requires sophisticated approaches combining computational and experimental methodologies:

Computational Modeling Approaches:

  • Differential equation models incorporating receptor trafficking and degradation

  • Models should account for the role of regulatory proteins (e.g., E3 ubiquitin ligase Cbl, GEF Cool-1, G protein Cdc42)

  • Parameter estimation using Bayesian statistical methods

Optimal Experimental Design Strategy:

  • Define measurable system components and unmeasurable components

  • Design experiments to minimize uncertainty on unmeasurable components

  • Sequential refinement of model parameters through iterative experimentation

  • Validation through prediction of system behavior under novel conditions

Key Experimental Validation Techniques:

  • Quantitative phosphoproteomics to track signaling cascade activation

  • Live-cell imaging with fluorescent reporters for spatiotemporal dynamics

  • Perturbation experiments using specific pathway inhibitors

  • Single-cell analysis to assess population heterogeneity in response

This integrated approach allows researchers to accurately reproduce experimental observations, make predictions with quantified uncertainties, and design experiments that systematically reduce uncertainty about complex system components .

What methodologies are employed for investigating EGF's role in organoid culture systems?

Organoid culture systems represent advanced models for studying EGF's role in tissue development and homeostasis. The following methodologies are employed:

Organoid Culture Protocol Components:

  • Basement membrane extracts (e.g., Cultrex UltiMatrix RGF)

  • Intestinal organoid culture medium containing:

    • Recombinant Human EGF (typically 50 ng/mL)

    • Recombinant Human Noggin

    • Recombinant Human R-Spondin 1

    • Recombinant Human Wnt-3a

Analytical Methods for Assessing EGF Function:

  • Immunohistochemical characterization:

    • Epithelial markers (E-Cadherin)

    • Cell-type specific markers (MUC2 for goblet cells, Chromogranin A for enteroendocrine cells)

    • Stromal components (Vimentin, Desmin for myofibroblasts)

  • Functional analysis:

    • Organoid formation efficiency

    • Growth rate quantification

    • Morphological complexity assessment

    • Cell type diversity analysis

    • Response to EGF withdrawal or inhibition

  • Molecular profiling:

    • Transcriptomic analysis of EGF-responsive genes

    • Signaling pathway activation status

    • Cell fate determination markers

These methods allow researchers to determine EGF's specific contributions to organoid development, cellular differentiation, and tissue-like organization in a three-dimensional context that better recapitulates in vivo conditions than traditional 2D culture systems .

How can one analyze and interpret contradictory findings regarding EGF concentration effects in different experimental systems?

Analyzing contradictory findings regarding EGF concentration effects requires systematic examination of experimental variables and careful consideration of context-dependent factors:

Methodological Approach to Resolving Contradictions:

  • Standardization of EGF Quantification:

    • Verify EGF concentration using multiple methods (ELISA, bioactivity)

    • Ensure consistent units and reporting standards

    • Consider the difference between total and bioavailable EGF

  • Context-Dependent Analysis:

    • Cell type specificity (epithelial vs. mesenchymal responses)

    • Receptor expression levels (EGFR/ErbB1-4 quantification)

    • Presence of co-factors and matrix components

    • Culture dimensionality (2D vs. 3D)

  • Temporal Dynamics Assessment:

    • Acute vs. chronic exposure effects

    • Receptor downregulation and desensitization kinetics

    • Feedback regulation mechanisms

Comparative Data Analysis Framework:

ParameterSystem ASystem BSystem CPotential Explanation for Differences
Effective EGF concentration20-100 pg/mL5-50 ng/mL50-100 ng/mLReceptor density, co-receptor availability
Cell typeBalb/3T3 fibroblastsEpithelial cellsImmobilized substrateDifferential receptor expression, signaling pathway activation
Exposure duration24-48h48-72h72h+Receptor cycling, adaptation mechanisms
Response measurementProliferationMigrationDifferentiationPathway-specific outcomes

When analyzing published literature, researchers should consider that optimal EGF concentrations can vary dramatically (from pg/mL to ng/mL ranges) depending on the experimental context. For instance, soluble EGF may be effective at concentrations as low as 20-100 pg/mL for proliferation assays in sensitive cell lines , while immobilized EGF demonstrates optimal activity at higher concentrations (50-100 ng/mL) .

What methodologies exist for evaluating EGF-based therapeutic applications in cancer research models?

Evaluation of EGF-based therapeutic approaches in cancer research requires multifaceted methodologies that address efficacy, mechanism, and potential resistance:

Experimental Design Framework:

  • Selection of appropriate cancer models:

    • Cell lines with defined EGFR expression levels

    • Patient-derived xenografts

    • Genetically engineered mouse models

    • 3D organoid cultures

  • Intervention approaches:

    • EGF-targeting vaccines (e.g., CIMAvax-EGF)

    • EGF-toxin conjugates

    • Anti-EGF antibodies

    • EGFR pathway inhibitors

  • Assessment parameters:

    • Tumor growth inhibition

    • Survival analysis using standardized statistical methods (log-rank test)

    • Biomarker evaluation (e.g., serum EGF concentration)

    • Immune response characterization for vaccine approaches

Clinical Trial Design Considerations:
The CIMAvax-EGF vaccine trial methodology provides a valuable framework:

  • Randomized phase III design

  • Post-chemotherapy (switch maintenance) timing

  • Survival analysis accounting for non-proportional hazards

  • Biomarker identification (baseline EGF concentration as predictive factor)

Results indicate median survival time was 12.43 months for vaccinated patients versus 9.43 months for controls (HR, 0.77; P = 0.036) in the per-protocol setting. Notably, patients with high baseline EGF concentration showed enhanced survival benefit (14.66 months) .

What are the critical quality attributes for GMP-grade recombinant human EGF and how are they assessed?

For GMP-grade recombinant human EGF production, critical quality attributes must be rigorously defined and assessed:

Physical and Chemical Characterization:

  • Primary structure verification:

    • Amino acid sequence analysis via mass spectrometry

    • N- and C-terminal sequencing

    • Peptide mapping

  • Higher-order structure analysis:

    • Disulfide bond mapping

    • Circular dichroism spectroscopy

    • Thermal stability assessment

  • Purity determination:

    • SDS-PAGE (>95% purity, single band at ~6 kDa)

    • Size exclusion chromatography

    • Reverse-phase HPLC

    • Host cell protein quantification (<100 ppm)

Biological Activity Assessment:

  • Potency assays:

    • Cell proliferation assay (ED50 20-100 pg/mL in reference cell line)

    • Receptor binding analysis

    • Phosphorylation of EGFR and downstream targets

  • Specificity testing:

    • Cross-reactivity assessment

    • Off-target binding evaluation

Stability and Formulation Studies:

  • Real-time and accelerated stability testing

  • Forced degradation studies

  • Formulation optimization for:

    • pH stability (typically pH 6.5-7.5)

    • Temperature sensitivity

    • Excipient compatibility

    • Freeze-thaw stability

These quality attributes ensure that GMP-grade EGF meets rigorous standards for research applications requiring high reproducibility and reliability, particularly in translational studies that may lead to clinical applications.

How can researchers optimize experimental design when studying EGF signaling pathways?

Optimizing experimental design for EGF signaling studies requires strategic planning and statistical rigor:

Mathematical Optimization Approach:
Researchers can apply optimal experimental design methods to maximize information gain while minimizing experimental effort:

  • Define the model structure (differential equations describing EGF-EGFR binding, trafficking, and signaling)

  • Identify unknown parameters and their prior distributions

  • Design experiments to minimize uncertainty on unmeasurable components

  • Apply statistical methods to suggest experimental conditions that reduce parameter uncertainty

Practical Implementation Strategy:

  • Initial characterization phase:

    • Dose-response curves across wide concentration range (pg/mL to μg/mL)

    • Time-course studies (seconds to hours)

    • Systematic inhibitor studies to isolate pathway components

  • Refinement phase:

    • Focus on identified regions of uncertainty

    • Design factorial experiments to examine interaction effects

    • Apply statistical power calculations to determine sample sizes

  • Validation phase:

    • Test model predictions under novel conditions

    • Perform perturbation experiments

    • Cross-validate findings across multiple experimental systems

This approach has demonstrated effectiveness in EGFR signaling studies, enabling accurate reproduction of experimental observations and generation of reliable predictions with quantified uncertainties .

What techniques can be employed to study the interaction between EGF and various matrix components in tissue engineering applications?

Understanding EGF-matrix interactions in tissue engineering requires specialized techniques:

Analytical Methods for EGF-Matrix Interactions:

  • Binding and release kinetics:

    • Radiolabeled EGF tracking

    • Fluorescently-labeled EGF imaging

    • ELISA-based quantification

    • Surface plasmon resonance for real-time binding analysis

  • Structural characterization:

    • Atomic force microscopy for surface topography

    • Scanning electron microscopy for morphological analysis

    • Confocal microscopy for spatial distribution

  • Functional assessment:

    • Cell attachment assays

    • Migration tracking in 3D matrices

    • Long-term stability testing

Comparative Analysis of Immobilization Strategies:
Research findings demonstrate significant differences between immobilization approaches:

Immobilization MethodImmobilization YieldBioactivity RetentionCell Proliferation at 72hAdvantages
CNBr-activated-agarose100%HighSignificant at 100 ng/mLHigh yield, maintained orientation
Glyoxyl-agarose12%ModerateModerate at 100 ng/mLSpecific attachment chemistry

LC-MS peptide mapping analysis reveals specific residues involved in immobilization, providing critical information about EGF orientation and accessibility when bound to different matrices .

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.