Recombinant Mouse Epidermal Growth Factor-Like Protein 8 (Egfl8) is a protein produced through the manipulation of the Egfl8 gene in Escherichia coli (E. coli) cells. This process involves techniques such as strain selection, codon optimization, fusion systems, co-expression, mutagenesis, and isotope labeling to achieve high levels of protein expression . Egfl8 is a member of the epidermal growth factor-like (EGFL) family and plays significant roles in cellular processes, including thymopoiesis, cell migration, and neurogenesis .
The production of recombinant Egfl8 involves several steps:
Expression in E. coli: The Egfl8 gene is introduced into E. coli cells, which are then manipulated to express large amounts of the protein.
Isolation: The protein is isolated from cell lysates or culture medium.
Purification: Techniques such as protein refolding, cleavage of fusion moieties, and chromatography are used to purify the protein .
Egfl8 has been identified as a negative regulator in mouse thymic epithelial cells (TECs) and thymocytes. It influences various cellular processes, including:
Cell Proliferation: Overexpression of Egfl8 inhibits the proliferation of cortical thymic epithelial cells (cTECs) .
Immune Response: Egfl8 affects the expression of genes involved in immune responses .
Neurogenesis: It acts as a neuritogen, influencing neurogenesis by activating kinases .
Egfl8 is downregulated in gastric and colorectal cancers, and its reduced expression is associated with a higher tumor-node-metastasis stage and poor prognosis . This suggests that Egfl8 could serve as a potential biomarker for these cancers.
Microarray analysis of Egfl8-overexpressing and -silenced cTECs revealed significant changes in gene expression profiles. In Egfl8-overexpressing cells, 43 genes were differentially expressed, with 10 upregulated and 33 downregulated . In contrast, Egfl8 silencing resulted in the differential expression of 390 genes, with 192 upregulated and 198 downregulated .
Pathway analysis using Pathway Studio identified that Egfl8-associated genes are involved in several biological processes, including cell proliferation, apoptosis, differentiation, and immune responses . Vascular endothelial growth factor-A (VEGF-A) acts as a pivotal hub among genes connected with Egfl8 .
| Condition | Upregulated Genes | Downregulated Genes |
|---|---|---|
| Egfl8 Overexpression | 10 genes | 33 genes |
| Egfl8 Silencing | 192 genes | 198 genes |
| Biological Process | Percentage of Associated Genes |
|---|---|
| Cell Proliferation | 20% |
| Apoptosis | 19% |
| Differentiation | 15% |
| Growth | 12% |
| Migration | 10% |
| Immune Response | 7% |
| Cell Cycle | 7% |
| ROS Generation | 7% |
| Chemotaxis | 3% |
Note: We will prioritize shipping the format currently in stock. If you require a specific format, please specify this in your order notes, and we will fulfill your request to the best of our ability.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement and will incur additional charges.
The specific tag will be determined during production. If you require a particular tag type, please inform us, and we will prioritize its use.
Epidermal growth factor-like protein 8 (Egfl8) is an evolutionarily conserved protein that belongs to the EGFL family. It was originally identified as a paralog of EGFL7 through a BLAST search of the mouse genome . Egfl8 is a secretory protein containing EGF-like repeats located in its extracellular domains, which are structurally similar to those found in Notch receptors . These EGF-like domains play a central role in controlling the Notch signaling pathway, suggesting that Egfl8 may function as a regulator of Notch signaling. The protein has been demonstrated to inhibit the survival and proliferation of mouse thymocytes, indicating its crucial role in T-cell development and immune regulation .
Mouse Egfl8 protein exhibits a diverse expression pattern across various tissue types. Western blot analysis has revealed high expression levels in several tissues including the thymus, lymph nodes, testis, and ovaries . The expression pattern suggests that Egfl8 may have tissue-specific functions, particularly in immune and reproductive systems. Within the thymus specifically, Egfl8 demonstrates equal expression levels in both freshly isolated thymocytes and thymic stromal cells, indicating its potential role in both cell populations during T-cell development .
The following table summarizes the expression pattern of mouse Egfl8 across various tissues:
| Tissue Type | Relative Expression Level |
|---|---|
| Thymus | High |
| Lymph nodes | High |
| Testis | High |
| Ovaries | High |
| Lung | Moderate |
| Ileum | Moderate |
| Skin | Low to moderate |
| Other tissues | Variable |
Research has identified an alternative splicing variant of Egfl8 in specific mouse organs. This 296-bp EGFL8 isoform, designated as EGFL8-2 (GenBank accession no. AB613266), contains an open reading frame (ORF) resulting from alternative splicing of the mouse full-length EGFL8 gene transcript . Analysis using the NCBI Conserved Domain Database (CDD) revealed that EGFL8-2 contains an EMI domain, which is also present in the full-length EGFL8 protein .
This alternative splicing variant is specifically found in the ileum, lung, and thymus, suggesting potential tissue-specific functions different from the full-length Egfl8 . The presence of this variant underscores the complexity of Egfl8 regulation and function across different tissues.
Production of recombinant mouse Egfl8 (rEgfl8) for research applications typically involves an E. coli expression system, as demonstrated in previous studies . The methodology includes:
Cloning and Expression Vector Construction: The mouse Egfl8 coding sequence is cloned into an appropriate expression vector containing a strong promoter (commonly T7) and a tag for purification (such as His-tag).
Protein Expression: The construct is transformed into an E. coli strain optimized for protein expression (e.g., BL21(DE3)). Expression is induced using IPTG (Isopropyl β-D-1-thiogalactopyranoside) under optimized conditions.
Protein Purification: The recombinant protein is purified using affinity chromatography (e.g., Ni-NTA for His-tagged proteins), followed by additional purification steps like ion-exchange or size-exclusion chromatography to achieve high purity.
Protein Verification: The purified rEgfl8 is verified using techniques such as SDS-PAGE, Western blotting, and mass spectrometry to confirm identity, purity, and integrity.
Biological Activity Testing: The functional activity of the recombinant protein is validated through appropriate bioassays, such as assessing its effects on thymocyte survival and proliferation.
Researchers should carefully optimize each step for their specific experimental requirements, as expression conditions and purification strategies may significantly impact the yield and activity of the recombinant protein.
When designing experiments to study Egfl8 function in vivo, researchers should consider:
Animal Model Selection: Choose appropriate mouse strains based on research objectives. Consider age, sex, and genetic background, as these factors may influence Egfl8 expression and function.
Delivery Method: For administration of recombinant Egfl8, determine the optimal route (intravenous, intraperitoneal, or direct thymic injection) based on target tissues and experimental endpoints.
Dosage Optimization: Establish dose-response relationships by testing multiple concentrations of rEgfl8. Previous studies have demonstrated that injection of rEgfl8 in mice resulted in decreased thymus weight and thymocyte numbers .
Timing Considerations: Determine appropriate time points for sample collection, as the effects of Egfl8 may be time-dependent. Both acute and chronic administration should be considered.
Control Groups: Include appropriate controls, such as vehicle-only injections or heat-inactivated protein controls, to distinguish specific Egfl8 effects from non-specific responses.
Outcome Measurements: Plan for comprehensive outcome assessments, including:
Statistical Design: Ensure adequate sample sizes for statistical power while minimizing animal use, following ethical guidelines for animal research.
Egfl8 has been demonstrated to negatively regulate the Notch signaling pathway in mouse thymocytes and thymic epithelial cells (TECs). Experimental evidence shows that recombinant Egfl8 (rEgfl8) suppresses the expression of Notch downstream targets, specifically Hes1 and Hey1, in both thymocytes and TECs .
The mechanistic interaction likely involves:
Direct or Indirect Interaction with Notch Receptors: The EGF-like repeats in Egfl8, which are structurally similar to those in Notch receptors, may facilitate direct interaction with Notch receptors or their ligands.
Competitive Inhibition: Egfl8 may competitively inhibit the binding of canonical Notch ligands to Notch receptors.
Downstream Signaling Modulation: The suppression of Hes1 and Hey1 expression suggests that Egfl8 interferes with the nuclear translocation of the Notch intracellular domain (NICD) or the formation of the transcriptional activation complex.
These interactions are particularly significant in the context of T-cell development, where Notch signaling plays a critical role in thymocyte differentiation, proliferation, and survival. The negative regulation of Notch signaling by Egfl8 may contribute to the observed inhibition of thymocyte survival and proliferation .
Recombinant Egfl8 (rEgfl8) has several significant physiological effects on mouse thymus and thymocytes:
Thymic Involution: In vivo injection of rEgfl8 in mice results in a decrease in thymus weight . This suggests that Egfl8 may play a role in thymic involution or atrophy.
Reduced Thymocyte Numbers: Administration of rEgfl8 leads to a significant reduction in the total number of thymocytes . This effect may be due to a combination of inhibited proliferation and increased apoptosis.
Inhibition of Thymocyte Proliferation: rEgfl8 has been shown to inhibit thymocyte proliferation, potentially by interfering with cell cycle progression .
Induction of Thymocyte Apoptosis: Treatment with rEgfl8 induces apoptosis in thymocytes, suggesting a role in regulating thymocyte survival .
Suppression of Notch Signaling: As mentioned earlier, rEgfl8 suppresses the expression of Notch downstream targets (Hes1 and Hey1) in thymocytes and TECs .
These findings collectively suggest that Egfl8 functions as a negative regulator of T-cell development by inhibiting thymocyte proliferation, inducing apoptosis, and suppressing Notch signaling. The physiological implications of these effects may include a role in regulating thymus size, controlling thymocyte numbers, and potentially participating in thymic selection processes.
Research has indicated that Egfl8 expression patterns may be altered in certain pathological conditions:
Cancer: Studies have demonstrated that EGFL8 expression is significantly decreased in patients with colorectal and gastric cancer . This suggests that EGFL8 may have a distinct expression pattern and mechanism of action in cancer progression, potentially functioning as a tumor suppressor.
Immune Disorders: Given its role in T-cell development and the Notch signaling pathway, altered Egfl8 expression may be implicated in immune disorders, particularly those involving T-cell dysfunction.
Developmental Abnormalities: As a regulator of cell survival and proliferation, abnormal Egfl8 expression might contribute to developmental abnormalities, especially in tissues where it is highly expressed.
Research investigating Egfl8 expression in pathological conditions should:
Utilize multiple detection methods (qRT-PCR, Western blotting, immunohistochemistry) to validate expression changes
Consider tissue-specific and cell-type-specific expression patterns
Correlate expression levels with disease progression and clinical outcomes
Investigate potential regulatory mechanisms (genetic, epigenetic, post-transcriptional) that may alter Egfl8 expression
Understanding the structure-function relationship of Egfl8 requires a multifaceted experimental approach:
Domain Deletion/Mutation Analysis: Generate recombinant Egfl8 variants with specific domain deletions or point mutations, particularly targeting the EGF-like repeats and the EMI domain. Functional assays can then determine which domains are essential for different activities.
Protein-Protein Interaction Studies:
Co-immunoprecipitation to identify binding partners
Surface plasmon resonance (SPR) to measure binding kinetics
Yeast two-hybrid screening to identify novel interactors
Proximity ligation assays to visualize protein interactions in situ
Structural Biology Techniques:
X-ray crystallography to determine high-resolution 3D structure
Nuclear magnetic resonance (NMR) spectroscopy for solution structure
Cryo-electron microscopy for larger complexes
Molecular dynamics simulations to predict domain movements and interactions
Cell-Based Functional Assays:
Reporter assays to measure effects on Notch signaling
Cell proliferation and apoptosis assays with structure-modified variants
Subcellular localization studies using fluorescently tagged variants
Comparative Analysis with EGFL7:
These approaches, when combined, can provide comprehensive insights into how the structural elements of Egfl8 contribute to its biological functions, particularly in the context of T-cell development and Notch signaling regulation.
When designing experiments to investigate Egfl8 function, researchers should implement the following critical controls:
Protein-Related Controls:
Heat-inactivated rEgfl8: To distinguish between specific biological activity and non-specific protein effects
Purification tag control: If using tagged recombinant protein, include the tag alone as a control
Structure-modified variants: Include functionally inactive mutants as negative controls
Dose-response experiments: Test multiple concentrations to establish dose-dependent effects
Cell/Tissue-Related Controls:
Vehicle controls: For in vivo experiments, include vehicle-only injections
Cell type controls: Test effects on different cell populations (e.g., thymocytes vs. thymic epithelial cells)
Time-course controls: Sample at multiple time points to establish temporal dynamics
Molecular Pathway Controls:
Pathway inhibitors: Include Notch pathway inhibitors (e.g., γ-secretase inhibitors) to compare with Egfl8 effects
Pathway activators: Include Notch pathway activators to test if Egfl8 can counteract activation
Gene expression controls: Include housekeeping genes for normalization in gene expression studies
Genetic Controls:
Gene knockdown/knockout controls: Include EGFL8 siRNA or CRISPR-Cas9 knockout controls
Overexpression controls: Include EGFL8 overexpression vectors with appropriate empty vector controls
Methodological Controls:
Positive and negative technical controls: For each assay (flow cytometry, Western blot, qPCR, etc.)
Inter-assay controls: To ensure reproducibility across experimental replicates
Blinding procedures: To minimize experimenter bias in subjective assessments
When faced with contradictory findings regarding Egfl8 function, researchers should:
Systematically Evaluate Experimental Differences:
Protein source and quality: Different recombinant protein preparation methods can yield proteins with varying activities
Experimental models: Results may differ between in vitro and in vivo systems or between different cell lines
Genetic background: Mouse strain differences can influence results
Developmental timing: Effects may vary depending on age or developmental stage
Dose and duration: Contradictory findings may result from differences in concentration or exposure time
Conduct Comprehensive Validation Studies:
Multiple methodological approaches: Verify findings using different techniques
Independent replication: Have different researchers or laboratories replicate key experiments
Cross-validation: Use both gain-of-function and loss-of-function approaches
Investigate Context-Dependent Effects:
Microenvironment factors: Examine if differences in cellular microenvironment explain contradictions
Signaling crosstalk: Investigate interactions with other signaling pathways that may modify Egfl8 effects
Cell state dependency: Determine if effects depend on cell activation state or cell cycle phase
Employ Advanced Analytical Methods:
Meta-analysis: Systematically analyze published data to identify patterns and sources of variability
Systems biology approaches: Develop computational models integrating contradictory findings
Single-cell analysis: Investigate if population heterogeneity explains seemingly contradictory results
Resolve Technical Considerations:
Antibody specificity: Validate antibodies against positive and negative controls
Splice variant specificity: Design experiments to distinguish between effects of different splice variants
Post-translational modifications: Investigate if PTMs affect function and contribute to contradictory findings