CD3E is a vital part of the TCR-CD3 complex, essential for adaptive immune responses . The TCR-CD3 complex initiates the transmission of TCR-mediated signals across the cell membrane through CD3 chains (CD3D, CD3E, CD3G, and CD3Z) when antigen-presenting cells (APCs) activate the T-cell receptor (TCR) . CD3E also facilitates the assembly of the TCR-CD3 complex and participates in its internalization and downregulation .
Recombinant Pig T-cell surface glycoprotein CD3 epsilon chain (CD3E), partial is primarily intended for research use . Potential applications include:
Positive Control: Can be used as a positive control in experiments involving pig T-cells and CD3E .
Immunogen: It can serve as an immunogen to generate antibodies against pig CD3E .
SDS-PAGE and Western Blotting: Useful in SDS-PAGE and Western blotting experiments for protein identification and characterization .
T-cell depletion: Complete T-cell depletion (TCD) during the induction period is essential for tolerance induction .
Immunotoxins: Anti-CD3 immunotoxins (ITs) are potential targeted therapy options for T-cell disorders .
T-cell depletion studies: Studies involving anti-CD3 recombinant immunotoxins have demonstrated the possibility of T-cell depletion in animal models . In one study, an anti-monkey CD3 recombinant immunotoxin (anti-CD3 rIT) was shown to induce a marked decrease in CD3+ T-cells in baboons, followed by a return to pre-injection levels within a few weeks .
Vascular Leakage: Research indicates the importance of the saporin molecule in inducing vascular leakage in mice .
| Property | Value |
|---|---|
| Target Name | T-cell surface glycoprotein CD3 epsilon chain (CD3E) |
| Species | Pig (Sus scrofa); Porcine |
| Host | E. coli |
| Expression Region | 22-116aa |
| Tag Info | N-terminal 6xHis-tagged |
| Theoretical MW | 15.2 kDa |
| Purity | >90% as determined by SDS-PAGE |
| Accession Number | Q7YRN2 |
| Form | Liquid or Lyophilized powder |
| Storage (Lyophilized) | 12 months at -20°C/-80°C |
| Storage (Liquid) | 6 months at -20°C/-80°C |
| Applications | Positive Control, Immunogen, SDS-PAGE, WB |
| Biological Activity | n/a |
| Restrictions | For Research Use Only. Not for use in diagnostic procedures. |
CD3 epsilon (CD3E) is an essential invariant component of the T-cell receptor (TCR)/CD3 complex in pigs, playing a crucial role in signal transduction following antigen recognition. The CD3 complex consists of CD3 epsilon, gamma, delta, and zeta chains, with CD3E being particularly important for TCR-dependent signal transduction across the lymphocyte membrane .
Structurally, CD3E contains:
An extracellular domain that associates with CD3 delta or gamma to form heterodimers
A transmembrane domain involved in complex assembly
A cytoplasmic domain containing immunoreceptor tyrosine-based activation motifs (ITAMs) that participate in signaling
The cytoplasmic domains of CD3E show a higher degree of conservation across species compared to the extracellular domains, reflecting their critical role in signaling functions . In pigs, CD3E is found on all T lymphocytes and a subpopulation of thymocytes .
Comparative analysis of porcine CD3E with other species reveals both conserved and divergent elements:
| Domain | Conservation Level | Notable Features |
|---|---|---|
| Extracellular | Lower conservation | More species-specific variations |
| Transmembrane | High conservation | Particularly between closely related species |
| Cytoplasmic | Highest conservation | Contains signaling motifs essential for T-cell function |
The short extracellular domain of the TCR zeta-chain shows 100% conservation between different species, while CD3E's extracellular domains show more variation . Importantly, antibodies directed against the intracytoplasmic domain of human CD3E have been shown to recognize bovine CD3E produced in E. coli, suggesting cross-reactivity in conserved regions . This conservation pattern is significant for developing cross-reactive reagents and understanding evolutionary preservation of T-cell signaling mechanisms.
Several expression systems have been documented for the production of recombinant pig CD3E with varying advantages for different research applications:
Bacterial systems (E. coli):
Mammalian cell expression systems:
Yeast expression systems:
Baculovirus expression system:
The choice of expression system should be guided by the intended application, with functional studies typically requiring mammalian expression systems that preserve native conformation and modifications.
Recombinant pig CD3E serves as a valuable tool for developing and testing T-cell-based immunotherapies using porcine models, which offer advantages over rodent models due to their physiological similarities to humans. Key applications include:
CD3E-targeting bispecific antibodies development:
CD3E-based CAR-T cell research:
CD3E immunotoxins (CD3E-IT) testing:
This translational research helps bridge the gap between rodent studies and human clinical applications, particularly for transplantation and cancer immunotherapy.
Researchers face several challenges when producing functional recombinant pig CD3E:
Heterodimer formation requirements:
CD3E naturally forms heterodimers with CD3 delta or gamma in the TCR complex
Refolding CD3E/δ heterodimers from E. coli-expressed inclusion bodies has proven difficult, unlike CD3E/γ
Stabilization methods include refolding in the presence of antibody fragments (e.g., single-chain variable domain fragments like UCHT1-scFv)
Disulfide bond formation:
Conformational validation methods:
Post-translational modifications:
Glycosylation patterns affect antibody recognition and protein stability
Bacterial systems lack glycosylation capabilities, potentially affecting conformation and function
These challenges highlight the importance of selecting appropriate expression systems and validation methods based on the intended application of the recombinant protein.
Investigating CD3E-mediated signaling in porcine T-cells requires multiple complementary approaches:
Monoclonal antibody-based activation studies:
Antibodies like PPT3 and BB23-8E6 recognize porcine CD3E and can activate T-cells
Activation can be demonstrated through calcium mobilization, increases in protein tyrosine phosphorylation, and proliferation assays
Flow cytometry with double-color analysis confirms binding specificity to swine lymphocyte populations
Phosphorylation analysis of signaling components:
Fluorescence resonance energy transfer (FRET) approaches:
Genetic modification studies:
CRISPR/Cas9 engineering of porcine T-cells to introduce mutations or tags in CD3E
Allows for tracking of signaling dynamics in real-time when combined with fluorescent reporters
Reconstitution experiments:
Expression of recombinant pig CD3E in cell lines lacking endogenous CD3E
Enables structure-function analysis of specific domains or mutations
These methods collectively provide insights into how CD3E contributes to T-cell receptor signaling pathways in porcine systems, with implications for both veterinary medicine and translational research.
CD3E expression exhibits notable patterns in porcine immunodeficiency models, particularly in SCID (Severe Combined Immunodeficiency) pigs:
ARTEMIS-deficient SCID pigs:
Despite severe lymphopenia, ART16/16, ART12/12, and ART12/16 SCID pigs unexpectedly maintain small populations of CD3ε+ cells in circulation and lymph nodes
CD3ε+ cells are detectable in newborn pigs (0 days of age) within lymph nodes prior to environmental exposure
These CD3ε+ cells show a skewed CD4α+CD8α+CD8β− T helper memory phenotype
Functional evidence of residual T-cell development:
Organ-specific distribution:
This "leaky" CD3E phenotype in porcine SCID models has important implications for using these animals in biomedical research, as it represents a variation from the classical SCID phenotype that may affect experimental outcomes.
Surface expression of CD3E shows significant variation across porcine T-cell subpopulations, with important functional implications:
Expression level differences between T-cell subsets:
Quantitative representation in different tissues:
Response to CD3E-targeting agents:
CD3E-immunotoxin (CD3E-IT) treatment preferentially depletes T cells with high CD3E expression (CD3Ehi)
This selective depletion results in enrichment of CD3Edim cells, particularly CD62Llo Tregs
The CD4/CD8 ratios typically increase following CD3E-IT treatment, indicating preferential depletion of CD8+ T cells over CD4+ T cells
This heterogeneity in CD3E expression creates opportunities for selective targeting of specific T-cell populations in therapeutic applications, particularly for transplantation tolerance induction.
Comprehensive validation of recombinant pig CD3E requires multiple complementary approaches:
Biochemical and biophysical characterization:
Structural validation:
Functional binding assays:
Signaling capacity assessment:
Heterodimer formation:
Co-expression with CD3 delta or gamma to validate heterodimer formation capacity
Co-immunoprecipitation studies to confirm interactions with other TCR components
These validation methods ensure that recombinant pig CD3E preparations maintain both structural integrity and functional activity for reliable research applications.
Developing antibodies that effectively target porcine CD3E presents several specific challenges:
Domain conservation variations:
Epitope accessibility issues:
Validation methodologies:
Clone selection considerations:
Clones like BB23-8E6-2B3C (an immunoglobulin isotype switch variant of BB23-8E6) have demonstrated utility in immunoprecipitation, immunocytochemistry, and costimulation
The PPT3 monoclonal antibody has been validated for activation capabilities through calcium mobilization, protein tyrosine phosphorylation, and proliferation assays
For researchers seeking to develop new antibodies, targeting the conserved regions of the CD3E molecule while considering species-specific variations in the extracellular domain is crucial for achieving both specificity and potential cross-reactivity.
Recombinant pig CD3E has significant potential for advancing xenotransplantation research:
T-cell depletion strategies optimization:
CD3E-immunotoxins (CD3E-IT) have demonstrated effectiveness in inducing long-term allograft acceptance in swine models
Understanding the differential depletion of T-cell subsets based on CD3E expression levels can improve transplant protocols
CD3E-IT treatment enriches Foxp3+ regulatory T cells (Tregs) in tissue-resident pools, potentially contributing to tolerance induction
Chimeric receptor engineering:
Tolerance induction mechanisms:
T-cell monitoring tools:
Recombinant CD3E can be used to develop improved flow cytometry reagents for monitoring T-cell responses post-transplantation
Multiparameter analysis can track the emergence of donor-reactive T cells or regulatory T cells
These applications could collectively advance our understanding of the porcine immune system's role in xenograft rejection and facilitate the development of more effective strategies for inducing transplantation tolerance.
Several cutting-edge methodologies are advancing our understanding of CD3E biology in porcine systems:
Cryo-electron microscopy (Cryo-EM):
Single-cell transcriptomics and proteomics:
CRISPR/Cas9 genome editing in primary porcine T-cells:
Enables precise modification of CD3E domains to assess functional consequences
Can introduce reporter tags for live-cell imaging of CD3E dynamics
Facilitates structure-function studies by creating domain deletions or point mutations
Advanced imaging techniques:
Molecular dynamics simulations:
Computational modeling of CD3E interactions with other TCR components
Insights into how specific domains contribute to complex stability and signaling
Prediction of critical residues for targeted mutagenesis studies
These methodologies collectively provide unprecedented resolution for understanding how CD3E structure relates to its function in porcine T-cells, with implications for both basic immunology and translational applications.
Researchers can employ several strategies to improve the production of correctly folded recombinant pig CD3E in bacterial expression systems:
Co-expression with chaperones:
Co-expression with molecular chaperones like GroEL/GroES, DnaK/DnaJ/GrpE, or trigger factor
Reduces inclusion body formation and enhances proper folding
Optimization of induction conditions:
Lower growth temperatures (16-25°C) to slow protein synthesis and improve folding
Reduced IPTG concentrations (0.1-0.5 mM) for more gradual induction
Use of auto-induction media for gradual protein expression
Fusion tag selection:
Refolding with stabilizing partners:
Oxidative refolding optimization:
Careful control of redox conditions with optimal glutathione ratios
Step-wise dialysis with decreasing denaturant concentrations
Addition of stabilizing agents like L-arginine or glycerol
Construct design considerations:
Expression of functional domains rather than full-length protein
Removal of hydrophobic transmembrane regions
Codon optimization for E. coli expression
These approaches have been successfully applied to various CD3E expression challenges, with the choice of method depending on the specific experimental requirements and downstream applications.
Accurately differentiating tissue-resident from circulating T-cells based on CD3E expression requires specialized techniques:
Intravascular staining approach:
Multi-parameter flow cytometry panels:
Combining CD3E with tissue-residency markers:
CD69 (early activation marker retained on tissue-resident cells)
CD103 (αE integrin, expressed on many tissue-resident T cells)
CD62L (L-selectin, low on tissue-resident effector memory cells)
CD3E+ cells in SCID pigs were found to be primarily CD8α+CD8β+CD4α− and CD8α+CD8β−CD4α+ phenotypes
Immunohistochemistry with spatial analysis:
Transcriptional profiling:
Tissue-resident T cells have characteristic transcriptional signatures
Single-cell RNA sequencing can classify cells based on residence markers
TCR sequencing can identify clonal relationships between populations
Parabiosis experiments:
Surgical joining of circulation between animals
Allows distinction between cells that equilibrate between partners (circulating) versus those that remain host-derived (resident)
These approaches collectively provide robust methods for distinguishing CD3E expression in tissue-resident versus circulating T-cells, critical for understanding T-cell biology in porcine models.
Rigorous experimental design for recombinant pig CD3E studies requires comprehensive controls:
Positive and negative expression controls:
Isotype controls for antibody experiments:
Specificity controls:
Competitive binding with known anti-CD3E antibodies
Binding to CD3E-knockout or CD3E-depleted cells
Cross-species reactivity assessment with human and mouse T-cells
Functional validation controls:
Positive control for T-cell activation: PMA/ionomycin or concanavalin A
Inhibition control: Cyclosporin A to block TCR-mediated activation
Dose-response curves to establish optimal concentrations
Reagent quality controls:
Endotoxin testing to ensure preparations are free from bacterial contaminants
Protein concentration verification through multiple methods (BCA, Bradford)
Storage stability testing with repeat analysis after freeze-thaw cycles
Technical replicates and biological replicates:
Minimum of three technical replicates per experiment
Independent biological samples from different animals
Statistical analysis appropriate for sample size and distribution