KAT2A antibodies are immunochemical reagents designed to detect and quantify the KAT2A protein in experimental settings. These antibodies enable researchers to investigate KAT2A's expression, localization, and interactions in diverse biological systems, ranging from cancer models to neural development studies.
Kidney Cancer: KAT2A overexpression correlates with advanced tumor stages and poor prognosis in renal cell carcinoma (RCC). Antibodies were used in Western blot and immunohistochemistry (IHC) to demonstrate elevated KAT2A levels in RCC patient samples .
Prostate Cancer: KAT2A upregulation in high-grade tumors was confirmed using IHC, linking it to biochemical recurrence .
Neural Stem Cells (NSCs): KAT2A knockdown increased PAX6 protein levels, as shown by Western blot, revealing its role in NSC differentiation .
Embryonic Stem Cells: KAT2A maintains pluripotency, with KO models showing accelerated differentiation .
Systemic Lupus Erythematosus (SLE): KAT2A modulates cGAS activity in immune cells, validated via scRNA-seq and Western blot .
Knockout Validation: Abcam’s ab217876 antibody shows no signal in KAT2A-knockout U-2 OS cells, confirming specificity .
Band Confirmation: Multiple antibodies (e.g., #3305, ABE1418) detect a single band at ~94 kDa, consistent with KAT2A’s molecular weight .
Dilution Ranges: Optimal dilutions vary by application (e.g., 1:500–1:1,000 for WB, 1:200 for IHC).
Storage: Most antibodies are stable at -20°C in glycerol-containing buffers .
Recent studies highlight KAT2A’s dual roles in acetylation:
Histone Modification: Regulates H3K9ac at gene promoters to control self-renewal pathways .
Non-Histone Targets: Acetylates transcription factors like PAX6 and MYC, influencing protein stability and transcriptional activity .
While KAT2A antibodies are widely used, cross-reactivity with paralogs (e.g., KAT2B) remains a concern. Advanced validation using CRISPR/Cas9 KO models, as seen in Abcam’s data, is critical for ensuring specificity . Future work may explore isoform-specific antibodies to dissect KAT2A’s divergent functions.
KAT2A (also known as GCN5) is a protein lysine acyltransferase with versatile activity. It functions as an acetyltransferase, glutaryltransferase, or succinyltransferase depending on the cellular context. Specifically, KAT2A acts as a histone lysine succinyltransferase, catalyzing the succinylation of histone H3 at lysine 79 (H3K79succ), predominantly near gene transcription start sites. This histone succinylation serves as an epigenetic mark for transcriptional activation. This activity requires interaction with the 2-oxoglutarate dehydrogenase complex, which provides the necessary succinyl-CoA. Within different complexes (e.g., SAGA and ATAC), KAT2A functions as either an acetyltransferase (HAT) or a succinyltransferase. It demonstrates significant histone acetyltransferase activity with core histones, although less so with nucleosome core particles. Histone acetylation, like succinylation, is an epigenetic mark promoting transcriptional activation. Furthermore, KAT2A is recruited by the XPC complex to promoters, where it acetylates histone variant H2A.Z, thereby enhancing gene expression. KAT2A plays crucial roles in several biological processes including long-term memory consolidation, synaptic plasticity (through regulation of hippocampal gene expression related to neuroactive receptor signaling), T-cell activation (by acetylating histone H3 at lysine 9 (H3K9ac) on the IL2 promoter, promoting IL2 expression), craniofacial development (by regulating H3K9ac), and embryonic stem cell pluripotency and differentiation. Beyond histones, KAT2A acetylates non-histone proteins such as C/EBPβ, PLK4, and TBX5, influencing heart and limb development (via TBX5 acetylation and nucleocytoplasmic shuttling) and acting as a negative regulator of centrosome amplification (through PLK4 acetylation). KAT2A also exhibits histone glutaryltransferase activity, catalyzing the glutarylation of histone H4 at lysine 91 (H4K91glu). This modification destabilizes nucleosomes by promoting H2A-H2B dimer dissociation. In the context of HIV-1 infection, KAT2A is recruited by the viral Tat protein, influencing Tat's transactivation activity and potentially facilitating chromatin remodeling of proviral genes.
KAT2A belongs to the nuclear A-type histone acetyltransferase (HAT) family that directly impacts gene transcription through histone modifications. Research demonstrates that KAT2A plays critical roles in cell cycle regulation, DNA replication, and DNA repair, establishing it as a key player in maintaining genome stability . Additionally, KAT2A functions as a coactivator of the c-MYC oncogene protein, highlighting its relevance in cancer biology .
Recent studies have revealed that KAT2A can regulate cellular processes through both transcriptional and post-translational mechanisms. For example, in neural stem cells (NSCs), KAT2A can acetylate PAX6 protein, facilitating its ubiquitination-mediated degradation, which affects NSC proliferation and differentiation . This demonstrates KAT2A's versatility beyond its traditional role in histone modification.
KAT2A primarily contributes to epigenetic regulation through histone acetylation, particularly H3K9 acetylation (H3K9ac), which typically promotes gene expression. In macrophages, KAT2A facilitates glycolysis reprogramming by suppressing nuclear factor-erythroid 2-related factor 2 (NRF2) activity, supporting H3K9ac and limiting NRF2-mediated transcriptional repression of proinflammatory genes .
In colorectal cancer (CRC), KAT2A-dependent cells display higher gene expression levels and enriched H3K27ac marks at gene loci linked to enterocytic differentiation . The epigenetic regulation by KAT2A appears to be context-dependent, with its activity varying across different cellular environments and developmental stages.
Based on extensive validation studies, KAT2A antibodies have been successfully employed in multiple experimental applications with specific dilution recommendations:
| Application | Validated Dilution Range | Notes |
|---|---|---|
| Western Blot (WB) | 1:2000-1:10000 | Successfully tested in multiple cell lines (HeLa, MCF-7, HSC-T6, NIH/3T3, SKOV-3) |
| Immunofluorescence (IF)/ICC | 1:200-1:800 | Validated in SKOV-3 cells |
| Immunoprecipitation (IP) | See published references | Successfully used in protein interaction studies |
| ELISA | Validated in assay-specific protocols | Used for quantitative protein measurements |
It's important to note that certain KAT2A antibodies may not be suitable for IHC applications, highlighting the necessity of selecting appropriate antibody clones for specific experimental purposes .
When designing co-immunoprecipitation experiments to study KAT2A interactions:
Begin by selecting a validated KAT2A antibody with demonstrated specificity in immunoprecipitation applications.
Include proper controls: IgG control from the same species as the KAT2A antibody and input controls to assess pull-down efficiency.
Consider crosslinking procedures for transient interactions.
Optimize lysis conditions to preserve protein-protein interactions while effectively disrupting cellular structures.
This methodology has been successfully utilized to demonstrate KAT2A interaction with PAX6 in neural stem cells. Researchers employed anti-PAX6 antibody for immunoprecipitation and detected both PAX6 and KAT2A in the precipitate by Western blot . Notably, the same approach demonstrated that KAT2A did not interact with SOX2 in neural stem cells, highlighting the specificity of the KAT2A-PAX6 interaction .
KAT2A plays a crucial role in regulating neural stem cell (NSC) differentiation through post-translational modification of PAX6, a key transcription factor in neural development. Current research indicates that KAT2A inhibition results in:
Accelerated NSC proliferation
Delayed differentiation
Potential apoptosis (context-dependent)
The effects of KAT2A on NSC differentiation appear to be mediated through a KAT2A/PAX6 axis that balances self-renewal and differentiation .
For experimental approaches, researchers have successfully employed:
Electroporation-mediated knockdown of Kat2a in NSCs followed by differentiation induction
EdU labeling to assess proliferation effects
Morphological analysis of cell processes to evaluate differentiation status
Western blotting to monitor PAX6 protein levels
After Kat2a knockdown, cells showed shorter processes after 24 hours of differentiation induction, suggesting inhibited differentiation. EdU labeling revealed increased proliferation, and Western blotting demonstrated elevated PAX6 levels .
When investigating KAT2A-mediated post-translational modifications in neural contexts, researchers should consider:
Combined transcriptional and protein analysis: KAT2A inhibition does not affect Pax6 mRNA levels in NSCs but increases PAX6 protein levels, indicating regulation at the post-translational level rather than transcriptional control .
Analysis of acetylation status: Use acetylation-specific antibodies or mass spectrometry to detect KAT2A-mediated acetylation of target proteins.
Ubiquitination assessment: KAT2A facilitates ubiquitination-mediated degradation of PAX6. Research has identified ring finger protein 8 (RNF8) as the E3 ligase responsible for PAX6 ubiquitination, working in conjunction with KAT2A .
Protein-protein interaction studies: Immunoprecipitation assays are essential to confirm direct interactions. For example, KAT2A has been shown to directly interact with PAX6 but not with SOX2 in NSCs .
Comparative HAT expression analysis: When studying KAT2A specifically, compare with other histone acetyltransferases (HATs) like CREB-binding protein (CBP) and P300/CBP-associated factor (PCAF) to establish specificity, as these showed different expression patterns during NSC differentiation .
To determine KAT2A dependency in cancer cells, researchers should employ a multi-omics approach:
CRISPR-Cas9 screening: This methodology has been successfully used to identify KAT2A-dependent cancer cell populations. In colorectal cancer (CRC), this approach revealed that KAT2A dependency is not simply correlated with KAT2A expression levels .
Genomic profiling: KAT2A dependency in CRC is associated with microsatellite stability and lower mutational burden. These genomic features can serve as potential predictive markers .
Transcriptomic analysis: Gene expression profiling can identify cancer cells with increased molecular differentiation signatures, which correlate with KAT2A dependency in CRC .
Epigenomic mapping: H3K27ac ChIP-seq analysis can reveal enrichment patterns at gene loci associated with cell differentiation states. In KAT2A-dependent CRC cells, enriched H3K27ac marks are observed at loci linked to enterocytic differentiation .
Functional validation: CRISPR-interference-mediated KAT2A knockdown experiments in both cancer cell lines and patient-derived 3D spheroid cultures can confirm dependency by assessing effects on cell growth, viability, and differentiation marker expression .
Additionally, in diffuse large B-cell lymphoma (DLBCL), KAT2A has been identified as a potential biomarker related to immune infiltration and malignant pathways .
For robust evaluation of KAT2A inhibition or depletion in cancer models, consider the following experimental design approaches:
Comprehensive in vitro models:
Gene expression analysis:
In vivo models:
Mechanistic studies:
Statistical robustness:
Research has shown that loss of KAT2A leads to decreased cell growth and viability both in vitro and in vivo, downregulation of proliferation and stem cell-associated genes, and induction of differentiation markers in colorectal cancer models .
KAT2A plays a crucial role in regulating inflammatory pathways in macrophages through multiple mechanisms:
Transcriptional regulation of inflammatory genes: KAT2A supports the transcription of proinflammatory genes such as Il1b and Nlrp3. Both pharmacological inhibition and siRNA silencing of KAT2A have been shown to suppress innate stimuli-triggered proinflammatory gene transcription .
NLRP3 inflammasome activation: KAT2A is required for proper activation of the NLRP3 inflammasome, a critical component of the innate immune response .
Metabolic reprogramming: KAT2A facilitates macrophage glycolysis reprogramming by suppressing nuclear factor-erythroid 2-related factor 2 (NRF2) activity and downstream antioxidant molecules .
Epigenetic regulation: KAT2A supports histone 3 lysine 9 acetylation (H3K9ac) and limits NRF2-mediated transcriptional repression of proinflammatory genes .
To effectively study these mechanisms, researchers should employ:
Gene expression analysis (qPCR, RNA-seq) to track inflammatory gene expression
Western blotting to assess protein levels of inflammasome components
Metabolic flux analysis to measure glycolytic activity
ChIP-seq to map H3K9ac patterns at inflammatory gene loci
In vivo models of inflammatory disease (such as rheumatoid arthritis)
Inflammasome activation assays measuring IL-1β secretion
To effectively study KAT2A's role in tumor immune microenvironments, researchers should consider:
Immune cell profiling in KAT2A-stratified tumors:
Development of a histone acetylation scoring system:
Functional validation experiments:
KAT2A knockdown in cancer cell lines followed by co-culture with immune cells
Analysis of changes in immune cell recruitment, activation, and function
Clinical correlation studies:
Stratify patient samples into high and low KAT2A expression groups
Compare immune cell infiltration patterns between groups
Correlate findings with patient outcomes
Research has demonstrated that patients with low histone acetylation scores (HAscore) have distinct tumor immune microenvironments and poorer prognoses in diffuse large B-cell lymphoma (DLBCL) . Additionally, KAT2A has been identified as a potential biomarker related to immune infiltration and malignant pathways in DLBCL .
Validating KAT2A antibody specificity is crucial for obtaining reliable experimental results. A comprehensive validation approach should include:
Knockout/knockdown controls:
Multiple antibody concordance:
Use at least two different KAT2A antibodies targeting distinct epitopes
Consistent results between antibodies increase confidence in specificity
Western blot validation:
Immunoprecipitation followed by mass spectrometry:
Perform IP with the KAT2A antibody
Analyze precipitated proteins by mass spectrometry to confirm KAT2A identity and assess off-target binding
Peptide competition assay:
Pre-incubate antibody with excess immunizing peptide
Signal elimination confirms epitope-specific binding
Cross-reactivity assessment:
Test antibody against closely related proteins (e.g., other HAT family members like PCAF)
Absence of cross-reactivity increases confidence in specificity
When using KAT2A antibodies to study multi-protein complexes:
Optimization of lysis conditions:
Different lysis buffers may preserve or disrupt specific protein-protein interactions
For nuclear protein complexes, consider nuclear extraction protocols rather than whole-cell lysis
Gentle lysis conditions (low detergent concentrations) help maintain protein complexes
Crosslinking considerations:
For transient or weak interactions, chemical crosslinking (formaldehyde, DSS, or BS3) may be necessary
Optimize crosslinking time and concentration to avoid artifactual associations
Sequential immunoprecipitation:
For specific sub-complexes, consider tandem IP approaches
First IP with KAT2A antibody followed by elution and second IP with antibody against suspected interaction partner
Mass spectrometry-based approaches:
Label-free quantitative proteomics can identify specific vs. non-specific interactors
SILAC or TMT labeling offers quantitative comparison between conditions
Consider proximity-dependent biotinylation (BioID or TurboID) as complementary approaches
Functional validation of interactions:
Control experiments:
Include IgG control from the same species as the KAT2A antibody
Include reverse IP (IP with antibody against suspected interaction partner)
Test interaction in both endogenous context and with exogenously expressed proteins
This approach has successfully revealed that KAT2A interacts with PAX6 but not with SOX2 in neural stem cells, demonstrating the specificity of protein interaction studies when properly controlled .
Based on recent findings of KAT2A's role in inflammatory pathways, researchers can design experiments to assess its therapeutic potential using:
Pharmacological inhibition studies:
Evaluate existing KAT2A inhibitors in inflammatory disease models
Monitor inflammation markers, disease progression, and clinical outcomes
Research has demonstrated that pharmacological inhibition of KAT2A suppresses proinflammatory gene transcription and impairs NLRP3 inflammasome activation both in vivo and in vitro
Genetic modulation approaches:
Disease model selection:
Rheumatoid arthritis models (as suggested by existing research)
Other inflammatory conditions with NLRP3 inflammasome involvement
Patient-derived samples for ex vivo experiments
Mechanism-focused experiments:
Combination therapy evaluation:
Test KAT2A inhibition alongside existing anti-inflammatory agents
Assess potential synergistic effects or reduced side effect profiles
To identify biomarkers of KAT2A dependency across cancer types, researchers should implement:
Multi-omics screening approaches:
Stratification criteria development:
Assess microsatellite stability status and mutational burden as potential biomarkers
Evaluate molecular differentiation signatures as indicators of KAT2A dependency
Research has shown that KAT2A dependency in colorectal cancer is associated with these features, independent of KAT2A expression levels
Epigenetic profiling:
Functional validation experiments:
Test KAT2A inhibition or depletion effects in patient-derived models
Correlate response with molecular and genetic features
Establish predictive algorithms for response likelihood
Clinical correlation studies:
These combined approaches can potentially identify patients most likely to benefit from KAT2A-targeted therapies across different cancer types.