CD3 Anti-Human

CD3, Mouse Anti-Human
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Targets

CD3 is a multi-subunit complex (γ, δ, ε, ζ) associated with the TCR on T lymphocytes. Anti-human CD3 antibodies primarily bind the ε chain (CD3ε), a 20 kDa transmembrane protein essential for TCR signaling .

Key Antibody ClonesIsotypeSpecificityApplications
UCHT1 Mouse IgG1κCD3εFlow cytometry, IHC, T-cell activation
OKT3 Mouse IgG2aCD3εImmunosuppression, allograft rejection
CD3-2 Mouse IgG2aCD3εELISpot, T-cell stimulation
HIT3a Mouse IgG2aCD3εMitogenic assays, immunohistochemistry

Mechanisms of Action

  • TCR Modulation: Anti-CD3 antibodies induce internalization or modulation of the TCR-CD3 complex, impairing antigen recognition .

  • Activation-Induced Apoptosis: Non-Fc receptor-binding variants (e.g., Fc-mutated OKT3) promote sustained TCR signaling, leading to apoptosis of activated T cells .

  • Regulatory T-Cell (Treg) Induction: Oral or modified anti-CD3 antibodies enhance IL-10 and TGF-β production, expanding CD4+CD25+FOXP3+ Tregs .

Therapeutic Uses

  • Type 1 Diabetes: Fc-non-binding anti-CD3 (e.g., teplizumab) preserves β-cell function by dampening autoimmune T-cell responses .

  • Transplant Rejection: OKT3 prevents acute graft rejection via T-cell depletion and TCR modulation .

  • Autoimmunity: Oral anti-CD3 suppresses inflammation in lupus and multiple sclerosis models by inducing systemic tolerance .

Experimental Findings

  • In hu-SPL-SCID mice, Fc-modified anti-CD3 reduced cytokine release (e.g., TNF-α) while maintaining immunosuppressive efficacy .

  • Intravenous teplizumab decreased C-peptide loss in recent-onset type 1 diabetes patients over 2 years .

Comparative Efficacy

ParameterFc-Binding Anti-CD3Non-Fc-Binding Anti-CD3
T-cell activationHigh (via FcγR cross-linking)Minimal
Apoptosis inductionModerateHigh (sustained ERK signaling)
Clinical tolerancePoor (CRS risk)Improved

Ongoing Research Directions

  • Oral Administration: Preclinical studies show oral anti-CD3 increases intestinal Tregs without systemic absorption, suggesting utility in gut-specific autoimmunity .

  • Humanized Mouse Models: NOD-SCID-IL2γc−/− mice reconstituted with human T cells enable mechanistic studies of anti-CD3 in vivo .

Product Specs

Introduction
CD3 is a complex found on T cells, consisting of four peptide chains: gamma, delta, epsilon, and zeta. This complex plays a crucial role in signal transduction within T cells. When defects occur within CD3, it can lead to T cell immunodeficiency, highlighting its importance in immune system function.
Formulation
This product is provided as a lyophilized powder. When reconstituted with the appropriate amount of solvent, the final concentration will be 1 mg/ml in a PBS solution.
Storage Procedures
For long-term storage, it is recommended to keep the lyophilized product at a temperature of 4 degrees Celsius. Upon reconstitution, if the antibody solution is not intended for immediate use (within one month), it is advisable to aliquot the solution into smaller volumes and store them at -20 degrees Celsius to maintain stability and prevent degradation.
Solubility
To reconstitute the lyophilized antibody, add the recommended volume of sterile H₂O to the vial. Gently mix the solution by swirling or inverting the vial to ensure complete dissolution. Avoid vigorous shaking. Rinse the sides of the vial with the solution to recover any residual powder. Allow the reconstituted antibody to sit at room temperature for 30-60 seconds before use.
Applications
This antibody is a versatile tool suitable for various applications, including blocking, staining, and activating experiments. For staining procedures, the recommended concentration is 10 µl of antibody per 1,000,000 cells. The optimal titers for blocking and T cell activation may vary depending on the specific experimental conditions and should be determined by the investigator.
Available Conjugates
In addition to the unconjugated form, this antibody is also offered with biotin and FITC conjugations, providing researchers with flexibility in their experimental design and detection methods.
Purification Method
Ion exchange column.
Type
Mouse Anti Human Monoclonal.
Clone
hCD3.
Immunogen
Purified human PBL T cells.
Ig Subclass
Mouse IgG2a.

Q&A

What is the molecular composition of the human CD3 complex and its significance in T cell function?

The human CD3 complex is a critical component of the T cell antigen receptor (TCR) complex. It consists of four distinct subunits: gamma (γ), delta (δ), epsilon (ε), and zeta (ζ). The CD3ε chain is a 20-kDa subunit that plays a central role in signal transduction during antigen recognition. CD3 is expressed on 70-80% of normal human peripheral blood lymphocytes and 60-85% of thymocytes, serving as a pan T cell marker on over 95% of circulating human peripheral T cells. The complex is also present on Purkinje cells and neurons in the cerebellar cortex but not on B cells or NK cells .

The TCR complex consists of TCR heterodimers (either α/β or γ/δ) that associate with the CD3 complex for effective cell surface expression and signaling. Each CD3 subunit contains one or multiple immunoreceptor tyrosine-based activation motifs (ITAMs) in their cytoplasmic domains that are essential for signal transduction following TCR activation .

How do different anti-CD3 antibody clones differ in their epitope recognition and functional properties?

Different anti-CD3 antibody clones recognize distinct epitopes on the CD3 complex and exhibit varying functional properties:

  • UCHT1: Specifically binds to the human CD3ε-chain and can stain both surface and intracellular CD3ε. It has mitogenic properties when used with costimulatory agents like pokeweed mitogen or anti-CD28 antibody .

  • MEM-57: Recognizes an extracellular epitope of the human CD3 complex containing either δ-ε or γ-ε subunit complexes. It requires the presence of the CD3ε subunit extracellular domain and demonstrates mitogenic effects on peripheral blood T cells and the Jurkat cell line .

  • HIT3a: Unlike UCHT1, this clone stains only extracellular CD3ε, providing researchers with options for specific experimental needs .

  • OKT3: A mouse anti-human CD3 mAb that has been used clinically as an immunosuppressive agent. It activates T cells through cross-linking between T cells and Fcγ receptor-bearing cells, leading to cytokine release .

These differences are crucial for selecting the appropriate antibody clone based on specific experimental objectives, whether for phenotyping, functional studies, or potential therapeutic applications.

What are the key considerations when selecting anti-CD3 antibodies for different experimental applications?

When selecting anti-CD3 antibodies, researchers should consider:

  • Epitope specificity: Different clones recognize distinct epitopes on the CD3 complex, which affects their binding characteristics and functional outcomes .

  • Fc receptor binding: Some anti-CD3 antibodies are engineered with mutations in their Fc portion to decrease binding to Fc receptors, reducing T cell activation and cytokine release. This is particularly important for therapeutic applications .

  • Species specificity: Anti-human CD3 antibodies are highly "species-specific" and typically do not cross-react with lymphocytes from other species except chimpanzees. For preclinical studies, researchers have developed models like NOD mice expressing human CD3ε as a transgene .

  • Application requirements: For immunohistochemistry, flow cytometry, or functional assays, different formats and clones may be optimal. For instance, for optimal indirect immunohistochemical staining, the UCHT1 antibody should be titrated (1:10 to 1:50 dilution) and visualized via a three-step staining procedure .

  • Isotype control: An isotype control should be used at the same concentration as the antibody of interest to account for non-specific binding .

What is the optimal protocol for using anti-CD3 antibodies in immunohistochemistry applications?

For optimal immunohistochemical staining with anti-CD3 antibodies, particularly the UCHT1 clone, researchers should follow this methodological approach:

  • Antibody titration: Titrate the UCHT1 antibody at dilutions ranging from 1:10 to 1:50 to determine optimal concentration for your specific tissue sample .

  • Three-step staining procedure:

    • Primary staining with the titrated anti-CD3 antibody

    • Secondary staining with polyclonal, biotin-conjugated anti-mouse Igs (multiple adsorbed) (e.g., Cat. No. 550337)

    • Tertiary detection with Streptavidin-HRP (e.g., Cat. No. 550946)

  • Visualization: Use the DAB detection system (e.g., Cat. No. 550880) for optimal visualization of the bound antibody complex .

  • Controls: Include appropriate isotype controls at the same concentration as the primary antibody to distinguish specific from non-specific staining .

  • Optimization note: Since applications vary, each investigator should titrate the reagent to obtain optimal results. Not all lots undergo routine immunohistochemistry testing, so researchers are encouraged to optimize protocols for their specific experimental conditions .

How can researchers accurately assess T cell depletion following anti-CD3 antibody administration in experimental models?

Accurate assessment of T cell depletion following anti-CD3 antibody administration requires comprehensive analysis beyond peripheral blood sampling:

  • Multiple tissue analysis: Examine both spleen and lymph nodes in experimental animal models, as peripheral blood analysis alone can yield biased results due to cytokine-induced effects on lymphocyte trafficking .

  • Timing considerations: The standard picture observed after administration of antibodies like OKT3 is the disappearance of all T cells from circulation within 30-60 minutes after the first injection. This is primarily caused by cytokine release that increases endothelial cell adhesiveness, leading to temporary lymphocyte sequestration rather than true depletion .

  • Quantitative assessment: Mitogenic CD3 antibodies (like hamster 145 2C11) can induce partial T cell depletion affecting approximately 50% of CD3/TCR+ cells, while non-mitogenic antibodies typically affect 20-30% of T cells .

  • Advanced models: For testing anti-human CD3 antibodies, specialized models such as NOD mice expressing the human CD3ε chain as a transgene can provide more relevant data, as conventional mouse models would not react with human-specific antibodies .

  • Flow cytometric analysis: Comprehensive multi-parameter flow cytometry should be employed to distinguish between true depletion, receptor downregulation, and trafficking effects on different T cell subpopulations.

What are the critical parameters for optimizing anti-CD3 antibody-mediated T cell activation protocols?

Optimizing anti-CD3 antibody-mediated T cell activation requires careful consideration of several parameters:

  • Antibody selection: Different clones (UCHT1, MEM-57, OKT3) have varying mitogenic potentials. Some are engineered to be non-activating through Fc mutations while retaining other functional properties .

  • Cross-linking requirements: The degree of T cell activation depends on cross-linking between T cells and Fcγ receptor-bearing cells. Non-Fc receptor-binding variants produce more sustainable phosphorylation of extracellular signal-regulated kinase-2 and greater release of IFN-γ than wild-type counterparts .

  • Presentation format: Solid phase-bound anti-CD3 (immobilized) versus soluble antibody can yield different outcomes. Human T cells prestimulated with solid phase-bound anti-CD3 for 24 hours may not enter cell cycle and may respond differently to subsequent stimulation .

  • Co-stimulation: Optimal T cell activation often requires co-stimulatory signals. Studies from the HLDA Workshop showed that antibodies like UCHT1 are mitogenic for CD3ε-positive cells when used in conjunction with costimulatory agents such as pokeweed mitogen or anti-CD28 antibody .

  • TCR internalization dynamics: Non-Fc receptor-binding variants dissociate more quickly from the T cell surface and cause less internalization of the TCR, which remains available in greater abundance on the cell surface for signaling. Cross-linking of these variants by antiglobulin enhances TCR internalization and minimizes induction of T cell apoptosis .

How do engineered modifications in anti-CD3 antibodies affect their functional properties and potential therapeutic applications?

Engineered modifications in anti-CD3 antibodies significantly alter their functional properties:

  • Humanization and Fc mutations: Humanized anti-CD3 antibodies with mutations in their Fc portion decrease binding to Fc receptors, which reduces cross-linking of T cell receptors. This minimizes T cell activation and cytokine release, addressing a major concern in clinical applications .

  • Signaling dynamics: Non-Fc receptor-binding variants produce more sustainable phosphorylation of extracellular signal-regulated kinase-2 and greater release of IFN-γ, more effectively causing activation-dependent T cell apoptosis compared to wild-type antibodies .

  • TCR modulation patterns: Non-Fc receptor-binding variants dissociate more quickly from the T cell surface and cause less internalization of the TCR. This leaves the receptor available in greater abundance on the cell surface for signaling, enabling sustained signaling pathways that promote apoptosis of activated T cells .

  • Therapeutic implications: Modified antibodies with reduced activating properties still retain significant immunosuppressive properties in vivo. Experimental models have shown that non-activating anti-CD3 mAbs can provide similar prolongation of human allograft survival as their activating counterparts, but with potentially fewer side effects .

  • Epitope-specific effects: Anti-CD3 antibodies that recognize different epitopes within the CD3 complex can elicit distinct cellular responses, providing opportunities for fine-tuning therapeutic interventions .

What are the mechanisms by which different anti-CD3 antibodies induce T cell apoptosis versus T cell activation?

The mechanisms distinguishing T cell apoptosis from activation by anti-CD3 antibodies involve complex signaling pathways:

How can researchers distinguish between the immunosuppressive effects of T cell depletion versus functional modulation when evaluating anti-CD3 antibody therapeutics?

Distinguishing between T cell depletion and functional modulation requires sophisticated experimental approaches:

  • Comprehensive immune monitoring: Beyond simple cell counts, researchers should analyze multiple parameters including:

    • T cell receptor expression levels

    • Activation markers (CD25, CD69, HLA-DR)

    • Functional responses to antigen stimulation

    • Cytokine production profiles

    • Regulatory T cell (Treg) frequency and function

  • Specialized experimental models: Models like NOD mice expressing human CD3ε as a transgene allow researchers to test anti-human CD3 antibodies and obtain insights into their mechanisms of action. This model helps differentiate between depletion and functional changes by providing access to multiple immune compartments beyond peripheral blood .

  • Comparative studies: Comparing non-Fc receptor-binding variants with their wild-type counterparts can help isolate mechanisms. Studies have shown that both types can provide similar immunosuppression (e.g., prolongation of human allograft survival), despite differences in T cell activation and cytokine release .

  • Temporal analysis: Differentiate immediate effects (often dominated by redistribution of T cells) from long-term effects (which may involve genuine depletion or functional reprogramming). CD3 antibody administration induces partial T cell depletion affecting about 50% of CD3/TCR+ cells with mitogenic antibodies, but only 20-30% with non-mitogenic variants .

  • Functional readouts: In transplantation studies, examining graft survival alongside immunological parameters helps determine whether observed effects result primarily from depletion or from altered T cell functionality .

What factors might explain contradictory results when using anti-CD3 antibodies across different experimental systems?

Several factors can contribute to contradictory results when using anti-CD3 antibodies:

How should researchers address variability in TCR/CD3 complex downregulation when interpreting flow cytometry data?

When interpreting flow cytometry data involving TCR/CD3 complex downregulation:

  • Multi-epitope analysis: Use antibodies targeting different epitopes on the CD3 complex or other T cell markers (CD2, CD5, CD7) to confirm T cell identification when CD3 expression is downregulated .

  • Kinetic studies: Perform time-course experiments to distinguish between temporary and persistent downregulation. Non-Fc receptor-binding variants dissociate more quickly from the T cell surface and cause less internalization of the TCR than wild-type antibodies .

  • Internalization vs. shedding: Distinguish between receptor internalization and shedding by comparing surface and intracellular staining. UCHT1 can stain both surface and intracellular CD3ε, making it useful for this purpose .

  • Control for technical artifacts: Include isotype controls at the same concentration as the primary antibody to account for non-specific binding. This is particularly important when analyzing samples with low CD3 expression .

  • Standardization approaches: Implement standardized protocols for sample processing, antibody titration, and instrument calibration to minimize technical variability. Since applications vary, each investigator should titrate reagents to obtain optimal results for their specific experimental conditions .

What are the critical considerations for interpreting T cell functional assays following anti-CD3 antibody treatment?

When interpreting T cell functional assays after anti-CD3 antibody treatment:

  • Pre-existing activation state: The response to anti-CD3 antibodies depends on the T cell's prior activation state. Human T cells prestimulated with solid phase-bound anti-CD3 for 24 hours respond differently than resting T cells .

  • Antibody properties: Different anti-CD3 antibodies (mitogenic vs. non-mitogenic, Fc receptor-binding vs. non-binding) elicit distinct functional responses. Non-Fc receptor-binding variants can induce more robust apoptosis of activated T cells while causing less general T cell activation .

  • Recovery kinetics: Monitor functional recovery over time, as effects may be transient or persistent depending on the mechanism of action. Some antibodies induce temporary anergy while others may cause lasting functional reprogramming or depletion of specific T cell subsets .

  • Subset-specific effects: Different T cell subpopulations may respond differently to anti-CD3 treatment. Comprehensive analysis should include assessment of CD4+ and CD8+ subsets, naive vs. memory cells, and regulatory T cells .

  • Context-dependent interpretation: Functional data should be interpreted in the context of the specific experimental system and therapeutic goal. For example, in transplantation models, non-activating humanized anti-CD3 mAbs retained significant immunosuppressive properties in vivo despite reduced T cell activation, suggesting different mechanisms of action compared to activating antibodies .

How do preclinical findings with anti-CD3 antibodies in animal models translate to human clinical applications?

Translating preclinical findings with anti-CD3 antibodies to human applications presents several challenges:

  • Species specificity barriers: Anti-human CD3 antibodies are highly "species-specific" and do not cross-react with lymphocytes from most non-human species (except chimpanzees). This necessitates specialized models like NOD mice expressing the human CD3ε chain as a transgene to test anti-human CD3 antibodies preclinically .

  • Mechanistic differences: Preclinical murine studies have indicated that non-FcR-binding anti-CD3 can induce apoptosis of antigen-activated T cells, which may be an important mechanism of immunosuppression. These findings helped guide the development of second-generation humanized anti-CD3 mAbs with reduced Fc binding avidity to minimize cytokine-induced toxicity while maintaining therapeutic efficacy .

  • Translational success: Humanized, non-activating anti-CD3 antibodies designed based on preclinical insights have shown promise in clinical settings. For example, humanized Fc variants demonstrated significant immunosuppressive properties in vivo comparable to OKT3 but with reduced side effects in human transplant models .

  • Pharmacodynamic considerations: Effects observed in peripheral blood may not accurately reflect what's happening in tissues. Comprehensive immune monitoring across multiple compartments in preclinical models provides more reliable predictions of clinical responses .

  • Model validation: The NOD mouse expressing human CD3ε as a transgene has proven to be a powerful tool for gaining insights into the mode of action of anti-human CD3 antibodies and developing suitable treatment protocols, particularly for combination therapies. This model bridges the gap between conventional animal studies and human clinical trials .

What are the key differences between first-generation and newer engineered anti-CD3 antibodies for immunotherapy applications?

First-generation and newer engineered anti-CD3 antibodies differ in several important aspects:

  • Origin and structure: First-generation antibodies like OKT3 were murine anti-human CD3 mAbs, while newer generations include humanized antibodies with CDRs of murine anti-CD3 grafted onto human IgG frameworks. This reduces immunogenicity in human patients .

  • Fc receptor binding: Newer antibodies often contain point mutations that reduce affinity for Fc receptors. This engineering approach minimizes cross-linking between T cells and Fcγ receptor-bearing cells, thereby reducing cytokine release syndrome—a major limitation of first-generation antibodies like OKT3 .

  • Activation potential: First-generation antibodies like OKT3 are potent T cell activators, causing systemic cytokine release and associated adverse effects. Newer humanized Fc variants with reduced activating properties maintain immunosuppressive efficacy while minimizing these side effects .

  • Signaling dynamics: Non-FcR-binding variants produce more sustainable phosphorylation of extracellular signal-regulated kinase-2, greater release of IFN-γ, and more effectively cause activation-dependent T cell apoptosis compared to wild-type antibodies. They also dissociate more quickly from the T cell surface and cause less TCR internalization .

  • Clinical application focus: While first-generation antibodies like OKT3 were primarily used to prevent or treat allograft rejection, newer engineered antibodies are being explored for broader applications including autoimmune conditions like type 1 diabetes, based on their improved safety profiles and mechanistic advantages .

How can researchers optimize anti-CD3 antibody protocols for inducing immune tolerance versus immune suppression?

Optimizing anti-CD3 antibody protocols for specific immunological outcomes requires careful consideration of several factors:

  • Antibody engineering: Select appropriately engineered antibodies based on the desired outcome. Non-FcR-binding variants may be preferable for inducing tolerance through specific effects on activated T cells while minimizing general immunosuppression .

  • Dosing strategies: The dose and schedule can significantly impact outcomes. Lower doses or short-course treatments may favor tolerance induction over prolonged immunosuppression. Titration studies are essential to determine optimal concentrations for specific applications .

  • Combination approaches: For complex therapeutic goals like operational tolerance in transplantation, consider combining anti-CD3 antibodies with complementary agents. The human CD3ε NOD mouse model has proven valuable for testing such combination therapies .

  • Target population selection: The timing of intervention relative to disease or transplantation can determine outcomes. Evidence suggests that anti-CD3 antibodies may be particularly effective when administered at disease onset when targeting recently activated pathogenic T cells .

  • Monitoring strategies: Implement comprehensive immune monitoring to distinguish tolerance from temporary immunosuppression. This should include assessment of regulatory T cell induction, functional responses to relevant antigens, and long-term follow-up after treatment discontinuation .

How are recent advances in understanding T cell signaling pathways influencing the development of next-generation anti-CD3 therapeutics?

Recent advances in T cell signaling research are driving innovation in anti-CD3 therapeutics:

  • Pathway-selective modulation: Deeper understanding of CD3 signaling reveals that distinct downstream pathways can be differentially modulated. Non-FcR-binding variants produce more sustainable phosphorylation of extracellular signal-regulated kinase-2 (ERK-2) compared to wild-type antibodies, suggesting targeted pathway modulation is possible .

  • Signal duration optimization: Research shows that the temporal dynamics of TCR/CD3 signaling significantly impact outcomes. Non-FcR-binding variants that dissociate more quickly from the T cell surface while causing less TCR internalization maintain the receptor available for sustained signaling, which appears advantageous for inducing apoptosis in activated T cells .

  • Epitope-specific engineering: Different antibody clones recognize distinct epitopes on the CD3 complex (e.g., UCHT1 binds CD3ε while MEM-57 recognizes complexes containing either δ-ε or γ-ε subunits). This knowledge enables the development of antibodies with epitope-specific effects tailored to particular therapeutic goals .

  • Cross-linking modulation: Understanding how cross-linking affects signaling outcomes allows for rational design approaches. Studies show that artificially enhancing cross-linking of non-FcR-binding variants with antiglobulin increases TCR internalization and reduces T cell apoptosis, suggesting that controlled modulation of cross-linking could fine-tune therapeutic effects .

  • Subset-specific targeting: Advances in understanding differential expression and signaling requirements across T cell subsets may enable the development of therapeutics that preferentially target pathogenic populations while sparing regulatory or memory compartments.

What novel experimental models are being developed to better predict human responses to anti-CD3 therapeutics?

Innovative experimental models are advancing predictive capabilities for anti-CD3 therapeutics:

  • Humanized CD3ε transgenic models: NOD mice expressing the human CD3ε chain as a transgene have been developed to overcome species specificity barriers. These mice have T cells that are sensitive to anti-human CD3 antibodies both in vitro and in vivo, providing a powerful tool to obtain further insight into antibody mechanisms of action and to implement suitable treatment protocols, particularly for combination therapies .

  • Human immune system mice: More comprehensive humanized mouse models incorporating multiple components of the human immune system are being developed to better recapitulate complex immune interactions and predict therapeutic responses.

  • Hu-SPL-SCID mice: Experimental models in which human splenocytes from cadaveric organ donors are inoculated into severe combined immunodeficient mice (hu-SPL-SCID mice) have been used to test the activating and immunosuppressive properties of anti-human CD3 mAbs in vivo, providing insights into their mechanisms of action and potential clinical efficacy .

  • Patient-derived xenografts: Models incorporating patient-derived immune cells can help assess individual variability in responses to anti-CD3 therapeutics, potentially enabling more personalized treatment approaches.

  • In vitro organoid systems: Advanced three-dimensional culture systems that better mimic tissue microenvironments may provide more physiologically relevant platforms for studying anti-CD3 antibody effects on tissue-resident T cells.

How might single-cell analysis technologies enhance our understanding of heterogeneous T cell responses to anti-CD3 antibodies?

Single-cell technologies offer unprecedented insights into T cell responses to anti-CD3 antibodies:

  • Response heterogeneity characterization: Single-cell RNA sequencing can reveal diverse transcriptional programs activated in different T cell subsets following anti-CD3 stimulation, potentially identifying previously unrecognized response patterns and cellular states.

  • Clonal fate tracking: Combined TCR sequencing and phenotypic analysis at the single-cell level can track the fate of specific T cell clones after anti-CD3 treatment, distinguishing between depletion, anergy, functional reprogramming, or expansion of particular clonal populations.

  • Signaling pathway resolution: Single-cell phosphoproteomic and proteomic approaches can resolve differences in signaling pathway activation between responding and non-responding T cells, potentially identifying biomarkers predictive of therapeutic outcomes.

  • Spatial context integration: Emerging spatial transcriptomic and proteomic technologies can map T cell responses to anti-CD3 antibodies within tissue microenvironments, providing context for how local factors influence treatment efficacy.

  • Temporal dynamics analysis: Single-cell technologies applied across multiple timepoints can reveal the kinetics of T cell responses, potentially identifying optimal therapeutic windows and explaining variability in clinical outcomes.

Product Science Overview

Mouse Anti-Human CD3 Antibody

Mouse Anti-Human CD3 antibodies are monoclonal antibodies developed in mice that specifically target the human CD3 complex. These antibodies are widely used in various scientific applications, including:

  • Flow Cytometry: For identifying and quantifying T cells in a sample.
  • Immunohistochemistry (IHC): For detecting CD3 in tissue sections, aiding in the classification of T-cell neoplasms .
  • Western Blot: For analyzing protein expression levels.
  • Immunocytochemistry: For visualizing CD3 in cell cultures.
  • Immunoprecipitation: For isolating CD3 proteins from a mixture.
Applications and Usage

The OKT3 clone is a well-known monoclonal antibody that reacts with an epitope on the epsilon-subunit within the human CD3 complex. It is capable of inducing T cell activation and is used in various functional assays . The antibody is typically purified by affinity chromatography and is available in different formulations, including phosphate-buffered solutions with sodium azide for preservation .

Storage and Handling

The antibody solution should be stored undiluted between 2°C and 8°C and protected from prolonged exposure to light. It is important not to freeze the solution to maintain its efficacy .

Significance in Research

Mouse Anti-Human CD3 antibodies are essential tools in immunology research. They help in understanding T cell behavior, immune responses, and the mechanisms underlying various diseases. These antibodies also play a crucial role in the development of immunotherapies and diagnostic techniques.

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.