CD3D Human

CD3D Human Recombinant
Shipped with Ice Packs
In Stock

Description

Biological Function and Mechanism

The protein binds GM-CSF with low affinity and forms a heterodimeric receptor with the beta subunit (CSF2RB) to activate downstream signaling pathways, including JAK-STAT and MAPK. Key roles include:

  • Regulating granulocyte and macrophage differentiation .

  • Modulating surfactant metabolism in pulmonary alveoli .

  • Influencing inflammatory responses via NF-κB and TNF pathways .

3.1. In Vitro Studies

  • Functional Assays: Inhibits GM-CSF-dependent proliferation of TF1 erythroleukemic cells (ED₅₀ ≤ 10 µg/mL with 0.5 ng/mL GM-CSF) .

  • Disease Modeling: Used to study pulmonary alveolar proteinosis (PAP) and surfactant metabolism disorders linked to CSF2RA mutations .

3.2. Preclinical Findings

Study ModelKey Results
Csf2ra⁻/⁻ Mice- Attenuated acute lung injury (ALI) from ricin exposure .
- Improved survival post-inhalation injury due to reduced neutrophil chemotaxis .
Gene-PMT Therapy- Restored GM-CSF signaling and surfactant homeostasis in hereditary PAP .
- Long-term macrophage engraftment with no adverse effects observed .

Associated Diseases

  • Surfactant Metabolism Dysfunction, Pulmonary 4: Caused by CSF2RA mutations impairing alveolar macrophage function .

  • Hereditary PAP: Linked to defective GM-CSF receptor signaling .

Product Specs

Introduction

CD3D, alternatively known as Delta Polypeptide (TiT3 Complex), is a single-pass transmembrane protein classified as type 1. It forms a crucial part of the T-cell receptor-CD3 complex by associating with CD3-zeta, CD3-gamma, CD3-epsilon, and the T-cell receptor heterodimers (alpha/beta or gamma/delta). This complex is essential for T-cell activation. Upon activation of the T-cell receptor (TCR) by antigen-presenting cells (APCs), CD3 chains, including CD3D, CD3E, CD3G, and CD3Z, mediate the transmission of TCR-mediated signals across the cell membrane. CD3D plays a vital role in the adaptive immune response and contributes to the differentiation of thymocytes.

Description

Produced in Sf9 Baculovirus cells, the recombinant CD3D protein consists of a single glycosylated polypeptide chain encompassing amino acids 22 to 105. It is fused at its C-terminus to a 239 amino acid hIgG-His-tag, resulting in a protein with a total of 323 amino acids and a molecular weight of 36.5 kDa. Under reducing conditions on SDS-PAGE, CD3D exhibits multiple bands ranging from 40 to 57 kDa. The protein undergoes purification using proprietary chromatographic techniques.

Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation

The CD3D protein solution is provided at a concentration of 0.5 mg/ml. It is formulated in a buffer containing 10% glycerol and Phosphate-buffered saline (pH 7.4).

Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To ensure optimal long-term stability, the addition of a carrier protein such as HSA or BSA (0.1%) is advisable. It is crucial to avoid repeated freeze-thaw cycles to maintain protein integrity.
Purity

The purity of the CD3D protein is determined to be greater than 90.0% based on SDS-PAGE analysis.

Synonyms

CD3d Molecule, CD3d Antigen, Delta Polypeptide (TiT3 Complex), CD3d Molecule, Delta (CD3-TCR Complex), T-Cell Receptor T3 Delta Chain, T3D, T-Cell Surface Glycoprotein CD3 Delta Chain, CD3 Antigen, Delta Subunit, OKT3, Delta Chain, CD3d Antigen, CD3 Delta, CD3-DELTA, IMD19, CD3D.

Source
Sf9, Baculovirus cells.
Amino Acid Sequence

FKIPIEELED RVFVNCNTSI TWVEGTVGTL LSDITRLDLG KRILDPRGIY RCNGTDIYKD KESTVQVHYR MCQSCVELDP ATVALEPKSC DKTHTCPPCP APELLGGPSV FLFPPKPKDT LMISRTPEVT CVVVDVSHED PEVKFNWYVD GVEVHNAKTK PREEQYNSTY RVVSVLTVLH QDWLNGKEYK CKVSNKALPA PIEKTISKAK GQPREPQVYT LPPSRDELTK NQVSLTCLVK GFYPSDIAVE WESNGQPENN YKTTPPVLDS DGSFFLYSKL TVDKSRWQQG NVFSCSVMHE ALHNHYTQKS LSLSPGKHHH HHH

Q&A

What is CD3D and what is its role in the T-cell receptor complex?

CD3D (Cluster of Differentiation 3d) is one of the three invariant chains (along with CD3γ and CD3ε) that form part of the human αβ T-cell receptor (TCR) complex. The complete TCR complex consists of a variable heterodimer (TCRαβ) and three invariant dimers: CD3γε, CD3δε, and ζζ/CD247 . CD3D forms a specific dimer with CD3ε (CD3δε) and plays a critical role in TCR assembly, transport to the cell surface, and signal transduction following antigen recognition. CD3D is indispensable for proper TCR expression on the cell surface and subsequent T cell function .

How does CD3D deficiency clinically differ from other CD3 chain deficiencies?

Despite high sequence homology between CD3D and CD3G, their deficiencies result in markedly different clinical presentations:

CD3D deficiency:

  • Early diagnosis due to severe symptoms

  • Severe T lymphopenia

  • No detectable TCR expression

  • Complete block in thymic T-cell differentiation

  • Requires urgent hematopoietic stem cell transplantation

CD3G deficiency:

  • Late diagnosis (patients can live into their 30s without stem cell replacement)

  • Mild T lymphopenia

  • Low but significant TCR expression (>30% vs. normal controls)

  • Autoimmune features are common

These clinical differences suggest fundamental differences in how each chain contributes to T cell development and function.

Why do mouse models of CD3D deficiency not recapitulate human findings?

Mouse knockout (KO) models show that CD3γ, but not CD3δ, is critical for early thymic development, which contradicts observations in humans where CD3D deficiency has more severe consequences than CD3G deficiency . This species-specific difference highlights the limitations of using mouse models for studying human CD3 deficiencies. Research suggests these differences may arise from species-specific variations in TCR assembly, thymic selection processes, or compensatory mechanisms. For this reason, humanized mouse models have been developed where mouse Cd3ε, Cd3δ, and Cd3γ are replaced with human CD3E, CD3D, and CD3G to better evaluate human CD3-mediated therapies and biology .

What are the most effective experimental approaches for studying CD3D function in human T cells?

Several experimental approaches have proven effective for studying CD3D function:

  • Knockdown (KD) strategies:

    • ShRNA-mediated knockdown using lentiviral vectors (shCD3δ-3 showed most efficient inhibition)

    • Validation via Western blot analysis normalized to loading controls like alpha tubulin

  • Cellular models:

    • Jurkat T-cell lines (E6-1, CH7C17)

    • HPB-ALL T-cell line

    • Primary T cells

    • HVS-transformed T cell lines derived from CD3-deficient patients

  • Developmental models:

    • Fetal thymus organ cultures (FTOC) with CD3D-knockdown human T-cell progenitors

    • Transduction of early CD34+ T-cell progenitors followed by FTOC to assess both development and TCR expression

  • Analysis methods:

    • Flow cytometry with multiple antibodies against TCR/CD3 components

    • Co-immunoprecipitation to study TCR assembly

    • Confocal microscopy for intracellular versus surface TCR expression

    • Western blot for protein expression analysis

How can researchers distinguish between effects of CD3D on TCR assembly versus TCR signaling?

Distinguishing these effects requires specific experimental designs:

  • For TCR assembly analysis:

    • Co-immunoprecipitation experiments to track associations between TCR components

    • Western blot analysis of TCR subunit interactions under non-reducing conditions

    • Subcellular fractionation to determine where TCR assembly is blocked (e.g., endoplasmic reticulum)

  • For TCR signaling analysis:

    • Measure calcium flux following TCR stimulation

    • Assess phosphorylation of downstream signaling molecules

    • Evaluate T cell activation markers and functional outcomes

Research has demonstrated that CD3D knockdown in mature T cells leads to TCR complexes that begin to form but are unable to incorporate ζζ/CD247 dimers and are retained in the endoplasmic reticulum, indicating a primary effect on assembly rather than signaling .

What experimental factors account for differences between CD3D knockdown effects in mature versus developing T cells?

Several key factors explain the differential effects observed:

  • Cellular context differences:

    • Mature T cells (like Jurkat) represent a single clone with no selection pressure or diversity

    • Developing T cells in the thymus represent polyclonal populations under selective pressure

  • Experimental evidence:

    • CD3D knockdown in mature Jurkat T cells results in severely reduced surface TCR (<11% of controls)

    • CD3D knockdown in human thymic progenitors followed by FTOC shows that the few T cells that emerge express relatively normal TCR levels (~80% of controls)

  • Interpretive framework:

    • Intrathymic plasticity allows selection of T cells that can express sufficient TCR despite CD3D deficiency

    • In clonal cell lines, this selection pressure and diversity doesn't exist

This explains why CD3D deficiency in patients is characterized by severe T cell development blockade rather than abundant T cells with low TCR expression.

What are the current methodologies for detecting and quantifying CD3D protein in research samples?

Multiple complementary techniques are available for CD3D detection:

  • Western blot:

    • Using antibodies specific for CD3D (such as from Santa Cruz Biotechnology)

    • Typically loading 40 μg of total protein extracted with NP-40 buffer

    • Normalization to loading controls like alpha tubulin

  • Flow cytometry:

    • Multiple antibodies against different epitopes of CD3 complex

    • Enables quantification of surface versus intracellular expression

    • Allows analysis at single-cell level

  • Confocal microscopy:

    • Visualization of both intracellular and extracellular CD3D/TCR

    • Detection of TCR patching and distribution patterns

    • Requires cell permeabilization for intracellular detection

  • ELISA:

    • Sandwich enzyme immunoassay with pre-coated antibody specific to Human CD3d

    • Detection range: 0.32-20 ng/mL

    • Sensitivity: 0.116 ng/mL

    • Suitable for serum, plasma, and other biological fluids

  • Co-immunoprecipitation:

    • For studying CD3D interactions with other TCR components

    • Requires careful lysis conditions to maintain protein-protein interactions

How should researchers optimize experimental conditions when studying CD3D in primary human T cells?

Optimization suggestions for CD3D studies in primary human T cells:

  • Sample preparation:

    • Fresh isolation is preferable to frozen samples

    • Careful activation status monitoring as this affects CD3 expression

    • Standardize isolation protocols to reduce variability

  • CD3D knockdown considerations:

    • Transduction efficiency must be optimized for primary cells

    • GFP or other selection markers should be used to identify transduced cells

    • Account for shorter lifespan of primary cells compared to cell lines

  • Controls:

    • Include both non-target shRNA controls and untransduced controls

    • Compare multiple donor samples to account for genetic variability

    • Include both CD3D and CD3G knockdown for comparative analysis

  • Analysis parameters:

    • Use multiple detection antibodies targeting different epitopes

    • Consider both surface and intracellular staining

    • Validate findings with multiple methodologies (flow cytometry, Western blot, etc.)

How can researchers reconcile differences between in vitro CD3D knockdown studies and observations in CD3D-deficient patients?

The reconciliation requires understanding several key factors:

  • Identified discrepancies:

    • CD3D knockdown in mature T cell lines shows <11% surface TCR expression

    • CD3D-deficient patient T cells show severe lymphopenia but those T cells that do develop can express TCR

  • Explanatory model:

    • Emerging evidence supports "intrathymic TCR expression plasticity" in developing T cells

    • Polyclonal T cell progenitors with varied TCR rearrangements allow for selection of those capable of expressing TCR despite CD3D deficiency

    • In contrast, established T cell lines represent a single clone with no selection pressure

  • Supporting evidence:

    • FTOC experiments with CD3D-knockdown progenitors showed that the few emerging T cells expressed considerable TCR levels

    • This suggests selective survival of those T cells capable of expressing TCR

  • Implication:

    • CD3D is absolutely required for normal T cell development, but rare T cells may develop with alternative TCR configurations in CD3D deficiency

    • These rare cells are insufficient to provide normal immunity in patients

What considerations should researchers keep in mind when comparing human versus mouse CD3D study results?

When comparing across species, researchers should consider:

  • Established differences:

    • In mice, CD3G (not CD3D) is critical for early thymic development

    • In humans, CD3D deficiency has more severe consequences than CD3G deficiency

  • Model selection:

    • Traditional mouse models do not recapitulate human CD3 deficiency phenotypes

    • Humanized mice with human CD3E, CD3D, and CD3G replacement provide better models for human CD3 biology

  • Experimental validation:

    • Findings in mouse models should be validated in human cells when possible

    • Parallel experiments in both species can identify species-specific differences

    • Humanized mouse models provide an intermediate system to bridge species differences

  • Therapeutic implications:

    • Human CD3-targeting therapies require testing in humanized mouse models

    • Humanized CD3 EDG mice are immune competent and allow evaluation of bispecific antibodies targeting human CD3

What research directions might resolve current contradictions in CD3D functional studies?

Several promising research directions could help resolve existing contradictions:

  • Developmental studies:

    • Further investigation of CD3D function at different stages of T cell development

    • Single-cell analysis of TCR assembly in CD3D-deficient thymocytes

    • Deeper investigation of selection processes that allow survival of rare T cells in CD3D deficiency

  • Structural approaches:

    • Detailed structural analysis of CD3D versus CD3G interactions with other TCR components

    • Investigation of alternative TCR configurations in CD3D-deficient cells

    • Identification of compensatory mechanisms in surviving T cells

  • Therapeutic explorations:

    • Development of transient CD3D suppression as a novel immunosuppressive approach

    • Testing in models of autoimmune or alloimmune disorders

    • Comparative analysis of CD3D versus CD3G targeting for therapeutic applications

  • Advanced models:

    • Development of inducible CD3D deficiency models to separate developmental from functional effects

    • Patient-derived induced pluripotent stem cells differentiated to T cells

    • Organoid models of human thymic development with CD3D modification

How might insights from CD3D research translate to novel immunotherapeutic approaches?

CD3D research suggests several promising therapeutic directions:

  • Immunosuppressive applications:

    • Transient reduction of CD3D expression as a novel immunosuppressive therapy

    • Particularly valuable for diseases with overactive T cells (autoimmune disorders)

    • May offer more selective immunosuppression than current approaches

  • Cancer immunotherapy:

    • Bispecific antibodies linking CD3 (including CD3D epitopes) to tumor-associated antigens

    • Evaluation in humanized CD3 EDG mouse models engrafted with tumors

    • Potential for improved T cell redirection strategies

  • Genetic intervention for CD3D deficiency:

    • Gene therapy approaches to restore CD3D expression

    • Modified T cell precursors with corrected CD3D for autologous transplantation

    • Gene editing of patient stem cells

What are the most promising tools and models for future CD3D research?

Emerging tools and models with significant potential include:

  • Advanced genetic models:

    • Complete CD3E, CD3D, and CD3G humanized mice that maintain immune competence

    • These models allow evaluation of human CD3-mediated therapy in vivo

    • CRISPR-engineered primary human T cells with CD3D modifications

  • Advanced analytical techniques:

    • Single-cell proteomics to track TCR assembly in individual cells

    • Live-cell imaging of TCR assembly and trafficking

    • Cryo-electron microscopy of TCR-CD3 complexes with various CD3D mutations

  • Clinical resources:

    • Expanded registry and biobanking of samples from CD3D-deficient patients

    • Longitudinal studies of CD3D-deficient patients after various interventions

    • Humanized patient-derived xenograft models

These tools will enable more precise understanding of CD3D functions and more effective translation of findings into therapeutic strategies.

Product Science Overview

Structure and Function

CD3D is a single-pass type I membrane protein that, along with CD3-gamma, CD3-epsilon, and CD3-zeta, forms the TCR-CD3 complex . This complex is composed of a variable heterodimer (either TCRαβ or TCRγδ) responsible for ligand recognition and three invariant dimers (CD3γε, CD3δε, and ζζ/CD247) that participate in the assembly and surface expression of the TCR complex . The binding of the peptide-MHC complex by the TCR results in conformational changes in CD3, leading to the phosphorylation of tyrosine residues within the immunoreceptor tyrosine-based activation motifs (ITAM) of the cytoplasmic tails of the CD3 and ζζ/CD247 chains . This phosphorylation event triggers several signaling cascades that result in T-cell activation and the induction of a cellular immune response .

Recombinant CD3D

Recombinant human CD3D is produced using DNA sequences encoding the human CD3D extracellular domain. This recombinant protein is typically expressed in HEK293 cells and is used in various research applications . The recombinant human CD3D protein comprises 95 amino acids with a predicted molecular mass of 11 kDa. Due to glycosylation, it migrates as an approximately 20-23 kDa band in SDS-PAGE under reducing conditions .

Applications and Importance

Recombinant CD3D is used in research to study T-cell development, function, and signaling pathways. It is also utilized in the development of immunotherapies and diagnostic tools. Understanding the role of CD3D in the TCR complex and its impact on T-cell activation is crucial for developing targeted therapies for various immune-related disorders and diseases .

Storage and Stability

Recombinant CD3D protein is typically provided as a lyophilized powder and should be stored under sterile conditions at -20°C to -80°C. It is recommended to aliquot the protein for optimal storage and avoid repeated freeze-thaw cycles .

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.