Zeta Corporation's recombinant monoclonal antibody targets Granzyme B, a serine protease critical for cytotoxic T lymphocyte (CTL)- and natural killer (NK) cell-mediated apoptosis .
Applications:
Mechanism: Granzyme B induces apoptosis by cleaving caspases and disrupting mitochondrial integrity in target cells .
Identified as a potential biomarker for hepatocellular carcinoma (HCC), this autoantibody targets the 14-3-3 zeta protein, which is overexpressed in premalignant liver diseases .
Study Group | Prevalence of Autoantibody | Significance (vs. Controls) |
---|---|---|
Liver Cirrhosis | 16.1% (15/93) | p = 0.01 (vs. NHS) |
Chronic Hepatitis | 0% (0/75) | p = 0.001 (vs. LC) |
Normal Human Sera | 1.7% (1/60) | Baseline |
Clinical Utility: Detected 9 months before HCC diagnosis in AFP-negative patients, outperforming traditional biomarkers like alpha-fetoprotein (AFP) .
Validation Methods: ELISA, Western blotting, and immunofluorescence .
This antibody targets the CD247 antigen, a component of the T-cell receptor (TCR)-CD3 complex essential for adaptive immunity .
Applications:
IκB-zeta (NFKBIZ) regulates NF-κB transcription factor activity and is implicated in inflammatory responses .
Functional Roles:
Commercial Availability: Monoclonal antibodies from Abcam (ab221914) and Thermo Fisher (clone LK2NAP) .
A polyclonal antibody (ab155037) detects the 14-3-3 zeta isoform, involved in signal transduction and apoptosis .
Targets receptor-type tyrosine-protein phosphatase zeta, linked to cell adhesion and glioma progression .
The regulatory B' subunit of Protein Phosphatase 2A (PP2A) modulates substrate selectivity, catalytic activity, and subcellular localization of the catalytic enzyme. Specifically, the PP2AA1/PP2A4/B'ZETA holoenzyme negatively regulates plant innate immunity by controlling BAK1 phosphorylation and activation within surface-localized immune receptor complexes. This holoenzyme is also crucial for negative regulation of brassinosteroid signaling, achieved by dephosphorylating and inactivating BRI1 in the cytoplasm. B'ZETA's functions extend to growth regulation, stress signaling, and reactive oxygen species (ROS) signaling.
PP2A-B'gamma and PP2A-B'zeta regulate reactive oxygen species homeostasis and signaling, influencing acclimation strategies in response to environmental changes. This suggests a critical role in plant defense responses and light acclimation. PMID: 26012558
IKB zeta (encoded by NFKBIZ) is a critical regulator of NF-kappa-B transcription factor complexes with dual inhibitory and activating functions. IKB zeta inhibits NF-kappa-B activity without affecting its nuclear translocation upon stimulation. Specifically, it inhibits DNA-binding of RELA and NFKB1/p50, affecting both the NF-kappa-B p65-p50 heterodimer and the NF-kappa-B p50-p50 homodimer . Interestingly, IKB zeta can also activate NF-kappa-B-mediated transcription in certain contexts. When associated with NFKB1/p50, it exhibits transcriptional activation activity and is recruited to specific promoters such as LCN2 . This protein is particularly involved in the induction of inflammatory genes activated through TLR/IL-1 receptor signaling and plays a role in T helper 17 (Th17) cell differentiation following T cell receptor (TCR) activation .
CD3 zeta, encoded by the CD247 gene, is a 164-amino acid protein belonging to the CD3Z/FCER1G family with a membrane-associated cellular localization . CD3 zeta serves as a critical component of the T cell receptor (TCR) complex, playing an essential role in signal transduction following antigen recognition. The phosphorylation of tyrosine residues within CD3 zeta's immunoreceptor tyrosine-based activation motifs (ITAMs) initiates downstream signaling cascades leading to T cell activation. Specifically, phosphorylation at tyrosine positions such as Y83 and Y142 serves as docking sites for ZAP-70 and other signaling molecules, connecting antigen recognition to cellular responses .
CD20 has been validated as an effective target epitope for T cells expressing CD20-specific scFvFc:zeta chimeric receptors. Research has demonstrated that the specific interaction between CD20 molecules on target cells and scFvFc:zeta receptors on engineered T cells triggers functional responses, including cytokine production (IL-2) and cytolytic activity . This interaction is highly specific, as evidenced by the absence of responses when using mock-transfected cells or CD20-negative stimulator cells. Furthermore, the addition of soluble anti-CD20 monoclonal antibodies inhibits this interaction, confirming the specificity of the CD20-scFvFc:zeta binding . These findings have significant implications for adoptive T cell therapies targeting B-cell malignancies expressing CD20.
IKB zeta plays a critical regulatory role in the synergistic induction of interleukins by IL-17A and TNFα in human keratinocytes. Recent research has demonstrated that IKB zeta functions as a key regulator of IL-17/TNFα-inducible psoriasis-associated genes and proteins . Specifically, IKB zeta regulates the IL-17A/TNFα-mediated synergistic induction of IL-19 and IL-20, which are important members of the IL-10 cytokine family involved in inflammatory processes . This regulatory mechanism has significant implications for understanding inflammatory skin conditions like psoriasis and potentially developing targeted therapeutic approaches.
Validation of anti-CD3 zeta antibodies requires a multi-faceted approach to confirm specificity across different experimental contexts:
Western Blot Validation: When validating anti-CD3 zeta antibodies by Western blot, researchers should look for bands at the expected molecular weight (approximately 16-18 kDa for monomeric CD3 zeta). For chimeric constructs like scFvFc:zeta, validation includes confirming the expected size of the chimeric protein (approximately 66 kD under reducing conditions and 132 kD under non-reducing conditions) . Multiple cell types should be tested, including positive controls (T lymphocytes) and negative controls (non-lymphoid cells).
Flow Cytometry Validation: For surface expression analysis, researchers should compare staining patterns between T cells (positive) and non-T cells (negative). For chimeric receptors, dual staining with antibodies recognizing different components (e.g., anti-mouse Fab-specific antibody and anti-human Fc gamma-specific monoclonal antibody) can confirm proper expression and folding .
Functional Validation: Most critical is functional validation through stimulation assays, where antibody binding should trigger expected downstream events. For example, with scFvFc:zeta constructs, coculture with CD20+ target cells should result in measurable IL-2 production or cytolytic activity that is not present with CD20- cells or mock transfectants .
Phosphorylation-Specific Validation: For phospho-specific anti-CD3 zeta antibodies (like those targeting pY83 or pY142), validation should include treatment with phosphatase inhibitors versus phosphatase treatment to confirm specificity for the phosphorylated epitope .
Antibody developability assessment during early research stages significantly influences candidate selection and optimization strategies. High-throughput (HT) developability screening of 152 human or humanized monoclonal antibodies revealed critical correlations between biophysical properties and downstream process parameters .
Effective developability assessment requires:
Integrated Analysis: The implementation of integrated HT developability workflows at the beginning of antibody discovery campaigns enables researchers to evaluate candidates across multiple parameters simultaneously, including sequence attributes, functional epitope diversity, and biophysical characteristics .
Iterative Optimization: Circular optimization workflows are essential, where analytical characterization informs sequence engineering (e.g., mutagenesis to remove post-translational modifications or disrupt hydrophobic patches), followed by re-analysis to confirm improvements in biophysical properties .
Predictive Modeling: Experimental biophysical data can be used to develop quantitative structure-property relationship (QSPR) models that predict key properties like hydrophobic interaction chromatography (HIC) retention times, which correlate with developability risks .
This approach enables researchers to eliminate antibodies with suboptimal properties early in the selection process and prioritize candidates for further evaluation, ultimately reducing development risks and ensuring only robust antibody molecules progress to later development stages.
IKB zeta exhibits both inhibitory and activating functions through distinct molecular mechanisms:
Inhibitory Mechanisms:
Direct binding to NF-κB subunits, preventing their association with DNA
Competition with coactivators for binding to NF-κB complexes
Recruitment of corepressors to specific promoter regions
Activating Mechanisms:
Formation of activating complexes with NFKB1/p50 that are recruited to specific promoters like LCN2
Recruitment to IL-6 promoters to activate IL-6 expression while simultaneously decreasing TNF-alpha production in response to lipopolysaccharide stimulation
This functional duality appears to be context-dependent, influenced by:
The specific target gene's promoter architecture
Cell type-specific cofactors
The nature of the stimulating signal (e.g., TLR/IL-1 receptor vs. TCR signaling)
Post-translational modifications of IKB zeta itself
Understanding these mechanisms provides insight into how IKB zeta functions as a critical regulator in both inflammatory responses and Th17 cell differentiation.
When designing experiments to study CD3 zeta-mediated signaling in engineered T cells (such as those expressing scFvFc:zeta chimeric receptors), researchers should implement the following experimental design elements:
Cell Line Selection:
Use well-characterized T cell lines (e.g., Jurkat) for initial mechanistic studies
Validate findings in primary human T cells to confirm physiological relevance
Include appropriate control cell lines (mock-transfected or expressing irrelevant receptors)
Stimulation Protocols:
Compare antigen-specific stimulation (e.g., CD20+ target cells for CD20-specific scFvFc:zeta) with non-specific stimulation (e.g., PMA/ionomycin)
Include titration of target cells to establish dose-response relationships
Implement time-course experiments to capture both early (minutes) and late (hours) signaling events
Readout Selection:
Validation Strategies:
Use soluble blocking antibodies to confirm specificity of receptor-target interactions
Implement genetic approaches (siRNA, CRISPR) to confirm the role of specific signaling molecules
Compare wildtype and mutant CD3 zeta constructs (e.g., Y83F, Y142F) to dissect the contribution of specific phosphorylation sites
This comprehensive experimental design allows for robust characterization of CD3 zeta-mediated signaling while controlling for potential confounding factors.
To effectively study IKB zeta's role in regulating IL-17A/TNFα-induced inflammatory responses, researchers should implement the following experimental design strategies:
Cell System Selection:
Primary human keratinocytes represent an optimal model for studying inflammatory skin conditions
Compare with other cell types (fibroblasts, immune cells) to establish tissue specificity
Consider 3D skin equivalents for more physiologically relevant models
Stimulation Conditions:
Test IL-17A and TNFα both independently and in combination to identify synergistic effects
Include dose-response and time-course experiments to capture the dynamics of IKB zeta induction
Compare with other inflammatory stimuli (e.g., IL-1β, LPS) to determine pathway specificity
Gene Expression Analysis:
Employ RNA-seq or targeted qRT-PCR panels focusing on inflammatory cytokines (IL-19, IL-20)
Use chromatin immunoprecipitation (ChIP) to identify direct IKB zeta binding to target gene promoters
Implement nascent RNA analysis techniques to distinguish between transcriptional and post-transcriptional effects
Genetic Manipulation Approaches:
Use siRNA or CRISPR-Cas9 to knockdown/knockout IKB zeta
Employ inducible overexpression systems to assess IKB zeta sufficiency
Generate domain mutants to identify critical regions for different functions
Readout Selection:
Parameter | Methodology | Important Considerations |
---|---|---|
IKB zeta Expression | Western blot, qRT-PCR | Nuclear vs. cytoplasmic fractionation |
Cytokine Production | ELISA, multiplex assays | Analysis of both cell lysates and supernatants |
Transcriptional Activity | Luciferase reporter assays | Use of wild-type and mutated promoter constructs |
Signaling Pathway Activation | Phospho-Western blots | Temporal resolution of activation patterns |
This experimental design provides a comprehensive framework for dissecting the complex role of IKB zeta in inflammatory responses triggered by IL-17A and TNFα.
Non-specific binding is a common challenge when working with anti-zeta antibodies. Researchers can implement the following troubleshooting strategies:
Optimization of Blocking Conditions:
Test multiple blocking agents (BSA, milk, commercial blockers) at various concentrations
Extend blocking time to ensure complete coverage of non-specific binding sites
Consider the addition of detergents (0.05-0.1% Tween-20) to reduce hydrophobic interactions
Antibody Dilution Optimization:
Perform titration experiments to identify the optimal antibody concentration
For anti-CD3 zeta antibodies, typical dilutions range from 1:500 to 1:2000 for Western blotting
For flow cytometry applications, concentrations between 1-10 μg/ml are typically effective
Sample Preparation Refinements:
For cell lysates, ensure complete denaturation for Western blotting applications
For fixed cells (immunocytochemistry/immunohistochemistry), optimize fixation time and permeabilization methods
When possible, use phosphatase inhibitors to preserve phosphorylated epitopes
Validation with Multiple Approaches:
Confirm binding specificity using knockout/knockdown samples as negative controls
Employ peptide competition assays to verify epitope specificity
Correlate results across multiple detection methods (e.g., Western blot and immunofluorescence)
Pre-adsorption Strategies:
Pre-adsorb antibodies with cell lysates from tissues lacking the target protein
Use recombinant protein columns to purify antibodies when high specificity is required
Consider using Fc receptor blocking reagents when working with samples containing immune cells
Implementing these strategies systematically can significantly reduce non-specific binding issues and improve data quality when working with anti-zeta antibodies.
When faced with contradictory results between different anti-zeta antibody clones, researchers should systematically evaluate several key factors:
Epitope Differences:
Map the epitopes recognized by each antibody clone
For CD3 zeta, antibodies targeting different phosphorylation sites (e.g., Y83 versus Y142) may yield different results depending on the activation state of the cells
For IKB zeta, antibodies recognizing different regions (N-terminal versus C-terminal) may detect different isoforms or post-translationally modified variants
Antibody Format Variations:
Consider differences between monoclonal and polyclonal antibodies
Evaluate potential effects of conjugation (fluorophores, enzymes) on epitope recognition
Assess differences in host species and isotype that may affect non-specific binding
Application-Specific Considerations:
Application | Key Considerations | Troubleshooting Approach |
---|---|---|
Western Blot | Denaturation conditions | Compare reducing vs. non-reducing conditions |
Flow Cytometry | Fixation/permeabilization | Test multiple permeabilization protocols |
Immunoprecipitation | Binding conditions | Adjust salt/detergent concentrations |
Immunohistochemistry | Epitope accessibility | Compare different antigen retrieval methods |
Technical Validation Approaches:
Use genetic knockout/knockdown models to confirm specificity
Perform peptide competition assays with specific and non-specific peptides
Validate with orthogonal methods (e.g., mass spectrometry)
Test alternative antibody clones from different manufacturers
Biological Context Evaluation:
Consider cell type-specific differences in target protein expression and modification
Evaluate the impact of activation state on epitope accessibility
Assess potential interactions with other proteins that might mask specific epitopes