The YWHAZ Monoclonal Antibody is a laboratory-generated immunoglobulin designed to specifically target the YWHAZ protein, also known as 14-3-3 zeta/delta. This protein belongs to the 14-3-3 family, which regulates cellular processes such as signal transduction, apoptosis, and cell cycle progression by binding to phosphorylated serine/threonine residues on partner proteins . The antibody is widely used in research to investigate YWHAZ’s role in diseases, particularly cancer, due to its overexpression in aggressive tumors .
Key Notes:
Immunohistochemistry (IHC): Effective in cancer tissues (e.g., lung, liver) with antigen retrieval using TE buffer (pH 9.0) .
Immunofluorescence (IF): Localizes YWHAZ in the cytoplasm, as shown in MCF-7 and U251 cells .
Breast Cancer (BrCa): YWHAZ overexpression correlates with DAAM1, enhancing cell migration and metastasis. Knockdown via siRNA reduces migratory capacity in MCF-7 and MDA-MB-231 cells .
Bladder Cancer: Amplification of YWHAZ at 8q22.3 is linked to muscle-invasive tumors and chemoresistance. Overexpression associates with advanced tumor stage and poor prognosis .
Gastric Cancer: High YWHAZ levels predict larger tumor size, venous invasion, and shorter survival. siRNA-mediated knockdown suppresses proliferation in MKN74 and MKN28 cell lines .
YWHAZ regulates apoptosis and survival pathways (e.g., PI3K/Akt, RhoA) by interacting with caspase-related proteins and modulating stress fibers . In pancreatic ductal adenocarcinoma (PDAC), YWHAZ overexpression drives epithelial-mesenchymal transition (EMT), promoting metastasis .
Supporting Data:
The YWHAZ monoclonal antibody specifically recognizes the YWHAZ protein and has demonstrated high specificity in human, mouse, and rat samples. It is suitable for use in a variety of applications, including ELISA, WB, IHC, IF, and FC. This YWHAZ monoclonal antibody was generated from YWHAZ antibody-secreting hybridomas, which were produced by fusing myeloma cells with spleen cells from an immunized mouse. The recombinant human 14-3-3 protein zeta/delta protein (133-212aa) was used as the immunogen. This YWHAZ monoclonal antibody undergoes protein A-mediated purification, achieving a purity level of up to 95%.
YWHAZ, also known as 14-3-3 protein zeta/delta, plays a crucial role in regulating a broad range of cellular processes, including signal transduction, cell cycle regulation, apoptosis, and gene expression through interactions with various protein partners. YWHAZ is involved in regulating multiple signaling pathways such as the PI3K/Akt, MAPK, and NF-κB pathways, and plays a significant role in mediating cell survival and growth.
YWHAZ, also known as 14-3-3 protein zeta/delta, functions as an adapter protein involved in regulating numerous cellular processes including signal transduction, cell cycle regulation, apoptosis, and gene expression through interactions with various protein partners. The protein is particularly significant due to its role in regulating multiple signaling pathways including PI3K/Akt, MAPK, and NF-κB pathways, directly influencing cell survival and growth mechanisms. In humans, the canonical YWHAZ protein consists of 245 amino acid residues with a molecular mass of approximately 27.7 kDa and primarily localizes to the cytoplasm. Its wide expression across various tissue types and conservation across species makes it a valuable research target for understanding fundamental cellular processes and disease mechanisms .
The specificity of YWHAZ monoclonal antibodies is validated through multiple cross-species reactivity tests, primarily demonstrating high specificity in human, mouse, and rat samples. Validation typically involves comparative Western blot analyses across these species to confirm consistent protein recognition patterns at the expected molecular weight (approximately 28 kDa). Additional validation may include knockout/knockdown studies where antibody signal is assessed in samples with genetically reduced YWHAZ expression to confirm signal specificity. For comprehensive specificity validation, researchers should conduct peptide competition assays where pre-incubation of the antibody with the immunizing peptide should abolish signal detection in subsequent applications. Cross-reactivity with other 14-3-3 family members should be excluded through careful epitope mapping and comparative analyses .
For optimal Western blot detection of YWHAZ protein, researchers should implement precise methodological parameters based on the 28 kDa target size. Sample preparation should include phosphatase inhibitors to preserve phosphorylation-dependent epitopes that may affect antibody recognition. Using 10-12% polyacrylamide gels provides optimal resolution for the YWHAZ protein, while PVDF membranes are preferred for their protein retention characteristics. The recommended dilution range for primary antibody incubation is 1:5000-1:60000, with overnight incubation at 4°C yielding superior results compared to shorter incubations. Blocking conditions should be optimized with either 5% non-fat milk or BSA in TBST, with milk sometimes showing reduced background for YWHAZ detection. For validation, positive control lysates from 293T or HeLa cells are recommended, as these consistently express detectable levels of YWHAZ protein .
Optimizing immunohistochemistry (IHC) protocols for YWHAZ detection requires careful consideration of fixation, antigen retrieval, and detection methods. Formalin fixation followed by paraffin embedding is generally suitable, though extended fixation beyond 24 hours may mask YWHAZ epitopes. Heat-induced epitope retrieval using citrate buffer (pH 6.0) for 20 minutes at 95-100°C significantly improves detection sensitivity compared to alternative retrieval methods. The recommended antibody dilution range for IHC applications is 1:50-1:200, with overnight incubation at 4°C generally yielding superior staining intensity and specificity compared to room temperature incubations. Background reduction can be achieved through careful titration of the primary antibody and using peroxidase blocking steps prior to antibody application. When evaluating YWHAZ expression patterns, cytoplasmic localization should be the predominant pattern, with potential additional melanosomal localization in specific cell types. Serial sections with primary antibody omission serve as essential negative controls for validating staining specificity .
For immunofluorescence applications detecting YWHAZ, subcellular localization patterns require methodological precision to distinguish authentic signals from artifacts. Fixation protocol selection significantly impacts epitope accessibility—paraformaldehyde (4%) for 15 minutes preserves YWHAZ epitopes while maintaining cellular architecture better than methanol fixation. Permeabilization should be carefully controlled, with 0.2% Triton X-100 for 10 minutes providing optimal intracellular antibody access without excessive protein extraction. The recommended dilution range (1:50-1:200) should be experimentally determined for each cell type, as expression levels vary significantly across different cellular models. When analyzing subcellular distribution, researchers should note that while YWHAZ primarily localizes to the cytoplasm, its distribution pattern may change in response to cellular signaling events, making time-course studies valuable for capturing dynamic relocalization. Co-staining with markers for specific organelles (particularly melanosomes in relevant cell types) provides critical context for interpreting localization patterns .
YWHAZ monoclonal antibodies serve as powerful tools for protein-protein interaction studies through immunoprecipitation (IP) approaches that capture physiologically relevant binding partners. For optimal IP results, researchers should use 1-4 μl of antibody per sample with magnetic protein A/G beads rather than agarose-based alternatives for reduced background. Pre-clearing lysates and implementing stringent washing protocols significantly enhances specificity. Researchers should consider that YWHAZ interactions are often phosphorylation-dependent; therefore, maintaining phosphorylation status through phosphatase inhibitors during sample preparation is critical. For confirming novel interaction partners, reciprocal co-IP experiments and proximity ligation assays provide necessary validation. Mass spectrometry analysis of immunoprecipitated complexes can reveal the complete YWHAZ interactome under specific cellular conditions. When studying stimulus-induced changes in YWHAZ interactions, time-course IP experiments following relevant treatments can reveal dynamic interaction patterns involved in signaling cascades .
Quantitative analysis of YWHAZ expression across different cellular states requires integrated methodological approaches that account for technical variability. Flow cytometry using YWHAZ monoclonal antibodies (dilution 1:50-1:200) provides single-cell resolution of expression levels, enabling identification of distinct cellular subpopulations based on YWHAZ abundance. For this application, appropriate isotype controls and permeabilization optimization are essential. When comparing YWHAZ expression across different cellular states using Western blot, normalization to multiple housekeeping proteins (rather than a single reference) significantly improves quantitative accuracy. Implementing standard curves using recombinant YWHAZ protein enables absolute quantification rather than relative comparisons. For longitudinal studies, researchers should maintain consistent antibody lots throughout the experimental timeline to eliminate batch effects. When analyzing response to treatments or stimuli, time-course sampling reveals expression dynamics that single time-point analyses might miss .
Implementation of YWHAZ monoclonal antibodies in disease-related signaling pathway studies requires integrated approaches that connect YWHAZ functional status to downstream effectors. For cancer research applications, combining YWHAZ immunodetection with phosphorylation-specific antibodies against PI3K/Akt and MAPK pathway components reveals mechanistic connections between YWHAZ expression and oncogenic signaling activation. Multiplexed immunofluorescence techniques using spectrally distinct fluorophores allow simultaneous visualization of YWHAZ and its signaling partners within the same cellular sample. For functional validation studies, YWHAZ knockdown/knockout models analyzed with the antibody can confirm specificity while revealing compensatory pathway adaptations. When studying patient-derived samples, co-staining for YWHAZ and disease-specific markers provides contextual information about its role in pathological states. Researchers should consider that altered YWHAZ subcellular localization, rather than just expression levels, may indicate pathway dysregulation in disease contexts, making compartment-specific analysis crucial .
Resolving detection issues with YWHAZ monoclonal antibodies requires systematic troubleshooting strategies addressing both technical and biological variables. For weak or absent Western blot signals, researchers should implement a step-wise approach: first, verify protein transfer efficiency using reversible staining methods; second, reduce stringency of washing steps by decreasing detergent concentration; third, extend primary antibody incubation time to 16-24 hours at 4°C. For high background issues, implement graduated BSA concentration series (1-5%) in blocking solutions to determine optimal blocking conditions. Multiple bands in Western blot may indicate post-translational modifications of YWHAZ or degradation products—validation through phosphatase treatment or freshly prepared samples can differentiate between these possibilities. Inconsistent immunohistochemistry staining often stems from variability in fixation times—standardizing this parameter across samples significantly improves reproducibility. For flow cytometry applications, titration experiments determining the optimal antibody concentration that maximizes the positive signal while minimizing background are essential for achieving reliable quantification .
Validating YWHAZ antibody specificity in complex experimental systems requires multi-parameter approaches that address potential cross-reactivity concerns. Implementing genetic validation through CRISPR/Cas9-mediated YWHAZ knockout cell lines provides the most definitive specificity control—complete signal abolishment confirms antibody specificity. Complementary RNA interference approaches using siRNA or shRNA against YWHAZ should demonstrate proportional signal reduction correlating with mRNA depletion levels. Peptide competition assays where the antibody is pre-incubated with excess immunizing peptide should eliminate specific binding in all applications. Mass spectrometry analysis of immunoprecipitated material can confirm exclusive pull-down of YWHAZ protein and its known interactors. When analyzing tissues with complex protein expression profiles, comparative detection using alternative YWHAZ antibodies recognizing different epitopes provides additional validation. Researchers should note that the 14-3-3 protein family shares sequence homology, making epitope selection critical for avoiding cross-reactivity with related family members .
When applying YWHAZ monoclonal antibodies across diverse sample types, researchers must implement systematic validation processes addressing matrix-specific challenges. For fixed tissue samples, extended antigen retrieval protocols (20-30 minutes) significantly improve epitope accessibility compared to cell line samples that typically require shorter retrieval times. Protein extraction methods substantially impact YWHAZ detection—RIPA buffer extraction maintains YWHAZ integrity while harsher lysis conditions may disrupt epitope structure. When comparing human and rodent samples, despite high sequence conservation (99% identity), subtle differences in post-translational modifications may affect antibody recognition, necessitating species-specific positive controls. For clinical specimens with variable preservation conditions, researchers should implement parallel processing of control samples to normalize for technical variation. When analyzing YWHAZ in subcellular fractions, contamination between fractions should be assessed using compartment-specific markers to ensure accurate localization claims. For all comparative studies across different sample types, standardized protocols and consistent antibody lots are essential for generating reproducible and comparable results .
Integration of YWHAZ monoclonal antibodies into multiplexed detection systems requires careful optimization of compatibility parameters across detection platforms. For multiplexed immunofluorescence applications, researchers should select YWHAZ antibodies with minimal spectral overlap with other fluorophores in the detection panel. Cross-reactivity between antibodies in multiplex panels can be assessed through sequential staining protocols compared to simultaneous application. Mass cytometry applications using metal-conjugated YWHAZ antibodies provide superior multiplexing capability with minimal signal spillover, allowing simultaneous detection of 30+ proteins including YWHAZ and its interaction partners. For tissue microarray analysis, cyclic immunofluorescence protocols with antibody stripping between rounds enable co-detection of YWHAZ with numerous other proteins on the same tissue section. Microfluidic-based detection platforms can leverage YWHAZ antibodies for sorting specific cell populations based on expression levels. When designing multiplexed experiments, researchers should validate that antibody performance remains consistent in multiplexed conditions compared to single-plex detection to ensure quantitative accuracy .
Developing functional assays with YWHAZ monoclonal antibodies requires careful consideration of antibody characteristics and cellular dynamics. For functional blocking studies, antibodies targeting specific YWHAZ domains involved in protein-protein interactions may disrupt endogenous binding events when introduced into live cells through microinjection or cell-penetrating peptide conjugation. Intracellular delivery efficiency should be quantitatively assessed before attributing phenotypic changes to specific YWHAZ function inhibition. For monitoring YWHAZ dynamics during cellular processes, antibody conjugation to quantum dots provides extended imaging capability while maintaining target specificity. When assessing phosphorylation-dependent YWHAZ interactions, phospho-specific antibodies used in combination with YWHAZ monoclonal antibodies can reveal regulatory mechanisms. Functional proximity ligation assays can detect specific YWHAZ interaction events at endogenous expression levels, providing spatial resolution of interaction events. Researchers should validate that antibody binding does not artificially stabilize or disrupt the protein complexes being studied, potentially through comparative analysis with alternative detection methods .
YWHAZ antibodies provide critical tools for dissecting post-translational regulation mechanisms when implemented in context-specific experimental designs. Phosphorylation-dependent binding is central to YWHAZ function; therefore, combining phosphatase treatments with YWHAZ immunoprecipitation reveals which interactions depend on partner protein phosphorylation status. Ubiquitination and sumoylation of YWHAZ can be assessed through sequential immunoprecipitation protocols where YWHAZ is first captured, followed by detection with modification-specific antibodies. Multiplexed Western blot analysis comparing total YWHAZ levels with specific post-translational modification states reveals the proportion of modified protein under different cellular conditions. When studying dynamic regulation, pulse-chase experiments with metabolic labeling followed by YWHAZ immunoprecipitation can reveal protein turnover rates and how modifications affect stability. For spatial regulation, subcellular fractionation followed by YWHAZ immunoblotting in each fraction reveals how modifications influence localization patterns. Researchers should consider that certain post-translational modifications may mask antibody epitopes, potentially requiring alternative antibody clones recognizing different regions of YWHAZ for comprehensive modification profiling .