CD3E antibodies bind to the epsilon (ε) chain of the CD3 complex, a multimeric protein comprising γ, δ, ε, and ζ subunits. The CD3ε chain has:
CD3E antibodies modulate T-cell activity through:
Tumor Growth Modulation:
Model | Treatment | Tumor Volume Change |
---|---|---|
Wild-type C57BL/6 | Murine CD3E Ab | +200% |
CD3E-humanized | Human CD3E Ab | +5% |
Organ-Specific Depletion:
CD3e-immunotoxin (S-CD3e-IT) showed variable efficacy:
CD3E antibodies spare CD62L<sup>lo</sup> regulatory T cells (Tregs) with low CD3ε expression, enriching them post-treatment .
Surviving Tregs exhibited minimal activation (Ki67<sup>+</sup>: <10% vs. 20–60% in CD4<sup>+</sup>Foxp3<sup>-</sup> cells) .
Bispecific Antibodies: Anti-CD3E/CD19 induced 100% B-cell depletion in humanized mice, with sustained effects for 7 days .
Checkpoint Combination: Anti-CD3E + anti-PD-1 synergistically enhanced tumor-infiltrating T-cell activation .
CD3E-targeted immunotoxins reduced pathogenic T-cell populations in collagen-induced arthritis models .
Organ-Specific Variability: CD3E expression differs across tissues, affecting antibody efficacy .
Treg Resistance: Low CD3ε levels on Tregs limit their depletion, potentially compromising tolerance .
Species Specificity: Most clinical-stage anti-human CD3E antibodies (e.g., Teplizumab) do not bind murine CD3E, necessitating humanized models .
Application | Antibody Clone | Reactivity |
---|---|---|
IHC (T-cell marking) | RBT-CD3e | Human, mouse, rat |
Flow cytometry | 145-2C11 | Mouse-specific |
Western blot | ab5690 | Human, mouse, rat |
CD3 epsilon (CD3E) is a critical component of the T-cell receptor (TCR)-CD3 complex present on T-lymphocyte cell surfaces. It plays an essential role in adaptive immune responses by transmitting signals across the cell membrane when antigen-presenting cells activate the T-cell receptor. When the TCR engages with an antigen, the immunoreceptor tyrosine-based activation motifs (ITAMs) in the CD3E cytoplasmic domain become phosphorylated by Src family protein tyrosine kinases LCK and FYN, triggering downstream signaling cascades . Beyond signal transduction, CD3E is essential for proper T-cell development and participates in the internalization and cell surface down-regulation of TCR-CD3 complexes via endocytosis mechanisms . Additionally, CD3E serves as a receptor for ITPRIPL1, with ligand recognition inhibiting T-cell activation by promoting interaction with NCK1, preventing CD3E-ZAP70 interaction and blocking the ERK-NFkB signaling cascade and calcium influx .
CD3E antibodies have been validated across multiple detection platforms with specific methodological considerations for each:
Flow Cytometry: CD3E antibodies effectively detect CD3 epsilon in human peripheral blood lymphocytes and can be used to identify T-cell subsets including CD3+, CD3+CD4+, and CD3+CD8+ populations . Optimal staining typically requires 30 minutes of incubation at 4°C protected from light, followed by washing with PBS before analysis .
Western Blot: CD3E can be detected in T-cell lines such as MOLT-4 and Jurkat cells, with a specific band appearing at approximately 21 kDa under reducing conditions . Negative controls should include cell lines lacking CD3E expression, such as Raji or THP-1 cells .
Immunocytochemistry/Immunohistochemistry: CD3E antibodies work effectively with fixed cells at concentrations of approximately 3-10 μg/ml. Detection typically employs fluorescently-conjugated secondary antibodies with nuclear counterstaining using DAPI .
The selection of detection method should align with experimental objectives, with flow cytometry being particularly suited for quantitative analysis of T-cell populations, while Western blotting and immunohistochemistry provide insights into protein expression levels and localization, respectively.
Validating CD3E antibody specificity requires a multi-faceted approach:
Knockout Controls: Comparing staining between wildtype cells (e.g., Jurkat cells) and CD3E knockout cells provides a definitive control for antibody specificity. The absence of staining in knockout cells confirms target specificity .
Cell Line Panel Testing: Test the antibody across multiple relevant cell lines known to express CD3E (T-cell lineages) and negative control cell lines (B-cell or monocytic lineages). For example, MOLT-4 and Jurkat cells should show positive staining, while Raji and THP-1 cells should be negative .
Isotype Controls: Include appropriate isotype control antibodies to assess potential non-specific binding .
Cross-Reactivity Assessment: When working with humanized models, confirm whether the antibody recognizes human CD3E, mouse CD3E, or both, as this significantly impacts experimental design and interpretation .
Proper validation ensures experimental results accurately reflect CD3E biology rather than non-specific interactions or technical artifacts.
CD3E humanized mouse models represent a significant advancement for evaluating human CD3E-targeted therapeutics in immunocompetent hosts. These models are developed through two primary approaches:
Full Gene Replacement: By replacing exons 2-7 of the murine Cd3e gene with the corresponding human CD3E sequence, researchers can generate mice that express functional human CD3E protein while maintaining normal T-cell development and function . This approach enables comprehensive evaluation of antibodies targeting the entire human CD3E protein.
Epitope Humanization: An alternative approach involves replacing only a critical epitope region of murine CD3E with the human sequence. For example, replacing a 5-residue N-terminal fragment of murine CD3-epsilon with an 11-residue stretch from the human sequence creates a model that expresses the specific epitope recognized by therapeutic anti-human CD3E antibodies .
These humanized models overcome limitations of xenograft approaches by enabling:
Evaluation of CD3E-targeted therapeutics in hosts with intact immune systems
Assessment of T-cell redirected killing by bispecific antibodies in vivo
Testing of combination therapies with immunomodulatory agents
Long-term toxicity and efficacy studies without the complications of graft-versus-host disease
The selection between full gene replacement and epitope humanization depends on research objectives, with epitope humanization being sufficient for evaluating epitope-specific antibodies while full gene replacement provides a more comprehensive model for studying human CD3E biology.
Optimizing CD3E antibody use in flow cytometry requires careful consideration of several technical parameters:
Antibody Concentration: Titration experiments are essential to determine optimal antibody concentration. While manufacturer recommendations provide a starting point (e.g., 2-10 μg/ml), optimal concentration should be determined empirically for each experimental system .
Staining Protocol: For primary lymphocytes:
Multi-Parameter Analysis: CD3E antibodies can be effectively combined with other T-cell markers (CD4, CD8, CD25, CD127) for comprehensive immunophenotyping. When designing multi-color panels, spectral overlap must be carefully considered .
Sample Preparation Variations: Fresh samples typically yield optimal results, though properly fixed samples can also be analyzed. For tissue samples, effective mechanical dissociation and enzymatic digestion protocols should be optimized to preserve CD3E epitopes .
For quantitative analysis, consistent gating strategies are critical, particularly when analyzing changes in T-cell subpopulations in response to experimental manipulations or disease progression.
CD3E antibodies are instrumental in developing and evaluating bispecific T-cell engagers (TCEs) through several methodological approaches:
Ex Vivo Cytotoxicity Assays:
Isolate splenocytes from CD3E humanized mice
Co-culture with target cells expressing the tumor antigen of interest
Add TCEs at varying concentrations
Measure T-cell activation (CD69, CD25 upregulation) and target cell killing
Include appropriate controls: isotype control antibodies and irrelevant target cells
In Vivo Efficacy Assessment:
Mechanistic Studies:
The availability of CD3E humanized mouse models has significantly enhanced the translational value of preclinical TCE evaluation by enabling assessment in immunocompetent hosts with physiologically relevant immune cell proportions and functions.
Researchers frequently encounter several challenges when working with CD3E antibodies:
Low Signal Intensity:
Potential Causes: Insufficient antibody concentration, epitope masking during fixation, low target expression
Solutions: Titrate antibody concentration, optimize fixation conditions (duration, reagent selection), enhance detection with signal amplification systems, ensure proper buffer composition to prevent non-specific binding
High Background:
Cross-Reactivity Issues:
Inconsistent Results:
Maintaining detailed records of antibody performance across experiments and implementing standardized protocols significantly improves reproducibility when working with CD3E antibodies.
CD3E detection requires tissue-specific methodological adaptations:
Peripheral Blood:
Lymphoid Tissues (Spleen, Thymus):
Tumor Tissue:
Fixed Tissue Sections:
Regardless of tissue type, validation using appropriate positive and negative controls is essential for accurate interpretation of CD3E staining patterns.
CD3E antibodies serve as versatile tools in cancer immunotherapy research through several applications:
Therapeutic Development:
Mechanistic Studies:
Biomarker Development:
Combination Therapy Optimization:
The development of humanized CD3E mouse models has particularly advanced this field by enabling the evaluation of human CD3E-targeted therapies in immunocompetent hosts with physiologically relevant immune cell populations and functions.
Recent technological innovations are expanding the research capabilities of CD3E antibodies:
Antibody Engineering Advances:
Site-specific conjugation methods for generating homogeneous antibody-drug conjugates
Affinity modulation techniques for optimizing T-cell activation thresholds
Fragment-based approaches (Fab, scFv) for enhanced tissue penetration
Conditional activation systems that restrict CD3E engagement to tumor microenvironments
Novel Detection Platforms:
Mass cytometry (CyTOF) enabling simultaneous detection of CD3E with dozens of other markers
Imaging mass cytometry for spatial resolution of CD3E-expressing cells in tissue context
High-parameter flow cytometry for comprehensive immunophenotyping of T-cell subsets
Multiphoton microscopy for real-time visualization of CD3E-mediated T-cell interactions
Humanized Model Systems:
Computational Approaches:
These technological advances collectively enhance the precision, versatility, and translational relevance of CD3E antibodies in both basic and applied immunological research.
CD3E, also known as CD3 epsilon, is a critical component of the T-cell receptor (TCR) complex. It plays a significant role in the immune response by mediating signal transduction necessary for T-cell activation and function. The mouse anti-human CD3E antibody is a monoclonal antibody that specifically targets the CD3 epsilon chain in humans. This antibody is widely used in research and clinical applications to study T-cell biology and to develop therapeutic strategies.
CD3E is a 20 kDa transmembrane protein that belongs to the immunoglobulin superfamily. It is primarily expressed on T cells, natural killer T (NKT) cells, and thymocytes at various stages of differentiation . CD3E forms a part of the TCR complex by associating with other CD3 subunits (CD3 delta, CD3 gamma, and CD3 zeta) and the TCR alpha/beta or gamma/delta chains . This complex is essential for the recognition of antigens presented by major histocompatibility complex (MHC) molecules on antigen-presenting cells.
The primary function of CD3E is to transduce activation signals from the TCR to the intracellular signaling pathways. Upon antigen recognition, the TCR-CD3 complex undergoes conformational changes that lead to the phosphorylation of immunoreceptor tyrosine-based activation motifs (ITAMs) present in the cytoplasmic domains of CD3 subunits . This phosphorylation event triggers a cascade of downstream signaling pathways, ultimately resulting in T-cell activation, proliferation, and differentiation.
The mouse anti-human CD3E antibody is a monoclonal antibody derived from Syrian hamster immunized with human T cells . This antibody specifically binds to the CD3 epsilon chain on human T cells, making it a valuable tool for various immunological assays and therapeutic applications.
The mouse anti-human CD3E antibody has significant therapeutic potential in the treatment of various immune-related disorders. For instance, it can be used to deplete T cells in conditions such as autoimmune diseases and organ transplantation . Additionally, it can be employed in adoptive T-cell therapies to enhance the efficacy of T-cell-based immunotherapies for cancer .