Histone H3.1 is a replication-dependent variant expressed during S phase, integral to nucleosome assembly and chromatin structure . Acetylation at K4 (H3K4ac) disrupts histone-DNA interactions, promoting chromatin accessibility and active transcription. This modification is associated with gene activation, particularly at promoter regions, and is dynamically regulated by histone acetyltransferases (HATs) and deacetylases (HDACs) .
The recombinant monoclonal antibody is engineered to target the acetylated K4 site on H3.1. Key features include:
Specificity: Recognizes acetylated K4 with minimal cross-reactivity to unmodified H3, methylated K4, or other acetylated lysines (e.g., K9, K14) .
Recombinant Production: Expressed in HEK293 or suspension cells via cloned antibody genes, ensuring lot-to-lot consistency and avoiding animal-derived components .
Epitope Recognition: Binds to the acetylated peptide sequence surrounding K4, validated via peptide array and dot blot assays .
Antibodies are rigorously tested for specificity:
ab176799: Shows strong binding to H3K4ac peptides but no cross-reactivity with formylated, methylated, or unmodified K4 .
ab300641: Targets multiple acetylated lysines (K4, K9, K14, etc.), but this broad specificity contrasts with H3.1 K4-focused antibodies .
4H6 clone: Demonstrates high affinity for acetylated K4 on H3.1, confirmed via peptide arrays .
ab176799: Enrichment at active promoters (e.g., GAPDH, EIF4A2) in HeLa cells, validated via qPCR and sequencing .
RM149: Used in ChIP-seq to map H3K4ac sites, critical for studying transcriptional regulation .
Supplier | Clone/ID | Key Features | Applications | Price Range |
---|---|---|---|---|
Abcam | ab176799, ab232931 | ChIP-grade, validated in HeLa cells; reacts with human/mouse. | ChIP, WB, ICC | $200–$500 |
Sigma-Aldrich | 4H6 clone (SAB6010027) | Targets H3.1 K4ac; recombinant, HEK293F-expressed. | ELISA, ICC, IHC | $300–$600 |
Cusabio | CSB-RA010418A0HU | Limited data; targets H3.1 K4ac. | WB, ICC | ~$210 |
Revmab | RM149 | No cross-reactivity; validated in neurite outgrowth studies. | ChIP, WB, ICC | N/A |
H3K4ac is a hallmark of active chromatin. Antibodies targeting this modification enable studies on:
Gene Regulation: Mapping transcriptional start sites and enhancer regions .
Cancer Biology: Investigating acetylation patterns in tumor microenvironments .
Developmental Biology: Analyzing chromatin dynamics during differentiation .
This acetyl-histone H3.1 (K4) recombinant monoclonal antibody is produced through a multi-step process. Initially, genes encoding the HIST1H3A antibody are extracted from rabbits immunized with a synthesized peptide derived from the human HIST1H3A protein acetylated at K4. These genes are then integrated into expression vectors and introduced into host suspension cells. These cells are cultured to promote the expression and secretion of the antibody. Subsequently, the HIST1H3A recombinant monoclonal antibody undergoes a rigorous purification process using affinity chromatography, isolating it from the cell culture supernatant. Its functionality is then validated through a range of assays, including ELISA, WB, ICC, and IF tests, confirming its ability to bind specifically to the human HIST1H3A protein acetylated at K4.
Acetylation of HIST1H3A at K4 is a crucial epigenetic modification that promotes an open chromatin structure and activates gene expression. It plays a critical role in transcriptional regulation, cellular differentiation, and the maintenance of gene expression patterns across generations of cells. Dysregulation of this modification can have significant consequences for health and disease.
Histone H3.1 is a core component of nucleosomes. Nucleosomes wrap and compact DNA into chromatin, limiting DNA accessibility to cellular machineries that require DNA as a template. As such, histones play a central role in transcription regulation, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is regulated through a complex set of post-translational modifications of histones, known as the histone code, and nucleosome remodeling.
H3K4 acetylation occurs on histone H3, a core component of nucleosomes that wrap and compact DNA into chromatin. As a post-translational modification, H3K4 acetylation contributes to the "histone code" that regulates DNA accessibility for cellular machineries involved in transcription, DNA repair, and replication .
Research has revealed significant cross-talk between H3K4 acetylation and H3K4 trimethylation (H3K4me3), with H3K4me3 being a well-established hallmark of transcription initiation . Notably, all K4-trimethylated histone H3 in human, mouse, and Drosophila cells undergoes dynamic acetylation mediated by the same lysine acetyltransferase, p300/CBP . This dynamic acetylation plays an essential role in the activation of immediate-early genes through facilitating RNA polymerase II association .
Genome-wide analyses have demonstrated that H3K4 acetylation, similar to H3K4me3, is enriched at the 5' ends of active genes along with other histone H3 acetylation marks . This positioning at transcription start sites further supports its role in gene activation.
Validation of antibody specificity is critical for reliable experimental outcomes. For Acetyl-Histone H3.1 (K4) antibodies, several complementary approaches should be employed:
Dot blot analysis: Test the antibody against a panel of modified and unmodified histone peptides. A specific antibody should:
Western blot validation:
ChIP validation:
Perform ChIP-qPCR targeting genes known to be enriched for active histone marks (e.g., GAPDH, EIF4A2) as positive controls
Include regions known to lack the modification (e.g., HBB promoter in non-erythroid cells, Sat2 satellite repeats) as negative controls
Compare enrichment patterns with other active marks like H3K4me3
ChIP-seq validation:
A properly validated antibody should consistently demonstrate specificity across multiple validation methods.
Acetyl-Histone H3.1 (K4) antibodies have been validated for several important research applications:
Chromatin Immunoprecipitation (ChIP):
ChIP-sequencing (ChIP-seq):
Western blotting:
Dot blot analysis:
Immunocytochemistry/Immunofluorescence (ICC/IF):
Each application requires specific optimization of antibody concentration, incubation conditions, and appropriate controls to achieve reliable results.
Optimizing ChIP experiments with Acetyl-Histone H3.1 (K4) antibodies requires careful consideration of several parameters:
Antibody amount: Titrate the antibody to determine optimal concentration. Based on published protocols, 1-10 μg per ChIP experiment is typically effective. For example, ChIP assays have been successfully performed using 1, 2, 5, and 10 μg of antibody against H3K4ac per experiment .
Chromatin preparation:
Essential controls:
Data analysis:
Express results as percent of input (the relative amount of immunoprecipitated DNA compared to input DNA after qPCR analysis)
For ChIP-seq, align reads to the reference genome using appropriate algorithms (e.g., BWA)
Examine peak distribution along the genome, particularly at transcription start sites
Technical considerations:
Pre-clear chromatin with protein A/G beads to reduce background
Use optimized wash buffers with appropriate salt concentrations
Consider that H3K4ac is dynamically regulated and may have rapid turnover
Successful ChIP experiments should show enrichment at active gene promoters compared to negative control regions and negative control IP samples.
H3K4 acetylation is dynamically regulated and sensitive to various experimental conditions:
HDAC inhibition: Treatment with HDAC inhibitors (e.g., Trichostatin A) causes rapid accumulation of acetylated histones, including H3K4ac, due to blocked deacetylation .
HAT inhibition: The p300/CBP inhibitor C646 blocks dynamic acetylation of H3K4me3, disrupting RNA polymerase II association and activation of immediate-early genes .
Cell cycle effects: Histone modifications can vary throughout the cell cycle, potentially affecting H3K4ac levels. Cell synchronization may be necessary for certain experiments.
Metabolic state: Acetylation requires acetyl-CoA as a substrate, so cellular metabolic changes can affect acetylation levels. Nutrient availability and metabolic inhibitors may alter global H3K4ac.
Gene activation: Induction of immediate-early genes is associated with dynamic changes in H3K4 acetylation. Serum stimulation after starvation can be used to study these dynamics .
Cross-talk with other modifications: H3K14 acetylation has been shown to protect H3K4me3 from demethylation, suggesting that manipulating one histone modification can affect others .
When designing experiments to study H3K4ac, these factors should be controlled or accounted for to ensure reproducible and interpretable results.
Several complementary methods can be employed to quantify changes in H3K4 acetylation:
Western blot analysis:
Use acid-urea (AU) gels to separate histones based on charge, resolving acetylated from non-acetylated forms
Normalize H3K4ac signal to total H3 to account for loading differences
Use dilution series of standards for semi-quantitative analysis
Particularly useful for assessing global changes
ChIP-qPCR:
ChIP-seq:
Dot blot quantification:
Use dilution series of modified peptides as standards
Apply equal amounts of histone extract from different conditions
Quantify signal intensity with image analysis software
Useful for comparing relative levels between conditions
Each method has advantages and limitations regarding sensitivity, specificity, and throughput. Combining multiple approaches provides more robust quantification and interpretation of H3K4ac changes.
The relationship between H3K4 acetylation and methylation reveals complex regulatory mechanisms:
Co-occurrence and cross-talk:
Enzymatic regulation:
Impact on gene expression:
Protection mechanisms:
Evolutionary conservation:
These findings indicate that H3K4 acetylation and methylation operate together in an intricate regulatory network controlling gene expression, with acetylation potentially serving as a mechanism to preserve or regulate methylation states.
Investigating the dynamic nature of H3K4 acetylation requires specialized techniques:
HDAC inhibitor time courses:
HAT inhibitor studies:
Pulse-chase experiments:
Label cellular acetyl-CoA pools with isotope-labeled acetate
Chase with non-labeled acetate
Track labeled acetyl groups on histones using mass spectrometry
Calculate half-life of acetyl marks on specific residues
Time-resolved ChIP:
Perform ChIP at multiple time points after stimulus
Measure changes in H3K4ac at target genes
Correlate with transcriptional changes
This can reveal kinetics of acetylation changes during gene activation
Sequential ChIP (Re-ChIP):
First ChIP with anti-H3K4me3 antibody
Second ChIP on the eluted material with anti-H3K4ac antibody
This identifies genomic regions where both modifications co-occur
Can reveal temporal relationships between modifications during gene activation
These methods can provide valuable insights into the highly dynamic nature of H3K4 acetylation and its role in transcriptional regulation.
H3K4 acetylation plays multifaceted roles in transcriptional regulation:
Promoter activation:
Immediate-early gene regulation:
Interplay with histone methylation:
HAT involvement:
Evolutionary conservation:
The data collectively indicate that H3K4 acetylation serves as both a direct regulator of transcription through affecting chromatin structure and RNA polymerase II recruitment, and as an indirect regulator by protecting other activating modifications like H3K4me3.
When encountering challenges with Acetyl-Histone H3.1 (K4) ChIP experiments, consider these troubleshooting strategies:
For weak signals:
Optimize chromatin fragmentation (200-500 bp fragments are ideal)
Increase cell number (use 4-10 million cells per ChIP experiment)
Reduce wash stringency (lower salt concentration in wash buffers)
Consider that H3K4ac might be dynamically regulated with rapid turnover
Try adding HDAC inhibitors to cell culture prior to harvest
For high background:
For inconsistent results between replicates:
For unexpected distribution patterns:
A properly optimized ChIP protocol should yield enrichment at active gene promoters compared to negative control regions, with recovery values typically ranging from 0.2-1.5% of input for active regions versus <0.05% for inactive regions .
ChIP-seq data analysis for H3K4ac requires several analytical steps and interpretative frameworks:
Primary data processing:
Distribution pattern analysis:
Comparative analysis:
Functional interpretation:
Visualization examples:
Properly analyzed ChIP-seq data should reveal that H3K4ac is primarily enriched at active gene promoters and correlates well with transcriptional activity and the presence of other active histone marks.
Robust validation of H3K4 acetylation findings requires multiple types of controls:
Antibody specificity controls:
ChIP-qPCR controls:
Genetic and chemical validation:
Cross-validation with other techniques:
Western blot to confirm global changes
Immunofluorescence to visualize nuclear distribution
Mass spectrometry for absolute quantification
RNA-seq to correlate with transcriptional outcomes
Biological replicates:
Multiple independent experiments
Different cell lines or tissues where appropriate
Different time points to capture dynamics
These controls collectively ensure that observations about H3K4 acetylation are specific, reproducible, and biologically meaningful, rather than artifacts of the experimental system or antibody properties.
Single-cell approaches represent the frontier for understanding H3K4 acetylation heterogeneity:
Single-cell ChIP-seq adaptations:
Current technical challenges in sensitivity and throughput
Modified protocols to improve antibody efficiency with limited material
Computational methods to address technical noise
Potential to reveal cell-to-cell variation in H3K4ac patterns
CUT&Tag and CUT&RUN single-cell approaches:
Higher sensitivity than traditional ChIP for limited material
More direct approach with fewer washing steps
Potential for multiplexing with other histone modifications
Could reveal correlation between H3K4ac and other marks at single-cell resolution
Integrated multi-omics approaches:
Combining H3K4ac profiling with RNA-seq in the same cells
Correlating with chromatin accessibility (ATAC-seq)
Establishing causal relationships between H3K4ac dynamics and gene expression heterogeneity
Understanding cell-to-cell variability in immediate-early gene responses
Spatial epigenomics:
In situ approaches to visualize H3K4ac in tissue contexts
Correlating with cell positioning and microenvironment
Tracking H3K4ac dynamics during tissue development
Understanding the role of H3K4ac in maintaining tissue-specific gene expression programs
These emerging approaches will likely reveal how H3K4 acetylation heterogeneity contributes to cell fate decisions, developmental processes, and disease progression at unprecedented resolution.
H3K4 acetylation has emerging implications in various disease contexts:
Cancer:
Altered H3K4ac patterns may contribute to oncogene activation
Mutations in p300/CBP (mediators of H3K4ac) are found in multiple cancer types
Dynamic H3K4 acetylation may be involved in cancer cell response to environmental changes
Potential therapeutic target through modulation of acetylation/deacetylation balance
Neurodegenerative diseases:
H3K4ac dynamics may be disrupted in conditions like Alzheimer's disease
The role of H3K4ac in immediate-early gene activation suggests potential importance in neuronal plasticity and memory formation
HDAC inhibitors show neuroprotective effects in multiple models, potentially through restoring acetylation patterns
Metabolic disruptions in neurodegenerative diseases may affect acetyl-CoA availability for histone acetylation
Inflammatory disorders:
H3K4ac likely plays a role in the rapid activation of inflammatory genes
The involvement of H3K4ac in immediate-early gene response suggests importance in cytokine production
Targeting the dynamic acetylation of H3K4me3 could provide new anti-inflammatory approaches
Differential H3K4ac patterns may contribute to chronic inflammatory states
Therapeutic implications:
p300/CBP inhibitors like C646 could provide targeted approaches to modulate H3K4ac
Understanding the interplay between H3K4 acetylation and methylation could inform combination therapies
Biomarkers based on H3K4ac patterns might help stratify patients for epigenetic therapies
Metabolic interventions affecting acetyl-CoA pools could indirectly modulate H3K4ac levels
Future research in these areas may establish H3K4 acetylation as both a biomarker and therapeutic target in multiple disease contexts.
Emerging technologies promise to revolutionize H3K4 acetylation research:
Advanced genomic approaches:
CUT&RUN and CUT&Tag for higher resolution and lower background
Long-read sequencing to link H3K4ac with distant regulatory elements
Targeted epigenome editing (using CRISPR-dCas9 fused to acetyltransferases or deacetylases) to establish causality
Single-molecule real-time techniques to observe dynamic changes
Proteomics innovations:
Enhanced mass spectrometry for absolute quantification of H3K4ac
Targeted proteomics to quantify combinatorial histone modifications
Identification of proteins specifically binding to H3K4ac versus H3K4me3
Proximity labeling to identify the complete interactome of H3K4ac-containing chromatin
Imaging technologies:
Super-resolution microscopy to visualize H3K4ac distribution in the nucleus
Live-cell imaging using engineered reader domains to track H3K4ac dynamics
Multiplexed imaging to simultaneously monitor multiple histone modifications
Correlative light and electron microscopy to link H3K4ac with chromatin ultrastructure
Computational approaches:
Machine learning to predict H3K4ac sites from DNA sequence and other epigenetic features
Integrative multi-omics analysis to understand the relationship between H3K4ac and other cellular processes
Modeling approaches to simulate dynamic acetylation/deacetylation cycling
Network analysis to position H3K4ac within broader epigenetic regulatory networks
These technological advances will likely provide deeper insights into the mechanistic roles of H3K4 acetylation in chromatin dynamics and gene regulation, with implications for both basic science and clinical applications.