The antibody demonstrates robust performance in paraffin-embedded tissues, including human breast cancer and brain specimens. Key protocols include:
Antigen Retrieval: High-pressure treatment with citrate buffer (pH 6.0) .
Blocking: 10% normal goat serum for 30 minutes at room temperature .
Detection: HRP-conjugated secondary antibodies with hematoxylin counterstaining .
Validated for ChIP applications, the antibody enables genome-wide profiling of histone H4 methylation states . Notably, histone H4K20 methylation (a related PTM) correlates with heterochromatin formation and DNA repair .
Arg35 Methylation: Associated with transcriptional repression and chromatin compaction, analogous to H4K20 methylation .
Cell Cycle Dynamics: Methylation at H4R35 peaks during S phase, suggesting roles in DNA replication .
Cancer Biomarker: Overexpression of methylated histone H4 isoforms observed in breast cancer tissues .
Neurological Disorders: Altered H4 methylation patterns linked to neurodevelopmental defects .
Specificity Testing: Peptide arrays and competitive ELISA confirm no cross-reactivity with unmodified H4 or other PTMs (e.g., acetylation at Lys12, Lys16) .
Batch Consistency: Reproducible staining patterns across multiple lots .
Table 2 compares HIST1H4A (Ab-35) with other histone H4 modification-specific antibodies.
HIST1H4A is one of multiple genes encoding histone H4, a core component of the nucleosome which serves as the basic building block of chromatin. Histone H4 is essential for packaging DNA into chromatin, thereby regulating DNA accessibility to the cellular machinery involved in transcription, replication, and repair processes. This protein plays a central role in chromatin structure and dynamics, with its functions primarily regulated through post-translational modifications .
Histone H4 is highly conserved and participates in multiple critical cellular processes including:
DNA compaction and chromatin organization
Regulation of gene expression through accessibility control
DNA replication and repair mechanisms
Maintenance of chromosomal stability
Epigenetic inheritance patterns
The protein has numerous aliases including H4/A, H4/B, H4/C, and several others, reflecting its importance across different genetic loci and conserved structure .
The HIST1H4A (Ab-35) polyclonal antibody has been validated for multiple research applications:
| Application | Recommended Dilution | Notes |
|---|---|---|
| ELISA | Varies by lot | Used for specific detection of HIST1H4A protein |
| Immunohistochemistry (IHC) | 1:10-1:100 | For tissue section analysis |
| Immunofluorescence (IF) | Application-specific | For cellular localization studies |
| Chromatin Immunoprecipitation (ChIP) | Application-specific | For studying protein-DNA interactions |
The antibody specifically recognizes the region around arginine 35 of human histone H4, making it particularly useful for studies investigating specific modifications at this site .
HIST1H4A is one of multiple genes encoding histone H4, which is among the most conserved proteins in eukaryotes. While the protein sequence of histone H4 is highly conserved across its various encoding genes (HIST1H4A through HIST1H4L, HIST2H4A, HIST2H4B, etc.), these variants differ primarily in their:
Genomic locations and organization within histone clusters
Regulatory elements controlling their expression
Expression patterns during the cell cycle
Responsiveness to cellular signals
For optimal ChIP experiments using HIST1H4A (Ab-35) antibody, the following methodology is recommended:
Cross-linking and Cell Preparation:
Fix cells with 1% formaldehyde for 10 minutes at room temperature
Quench with 125 mM glycine for 5 minutes
Wash cells twice with cold PBS
Collect 1-5×10^6 cells by centrifugation
Chromatin Preparation:
Lyse cells in appropriate buffer containing protease inhibitors
Sonicate chromatin to generate 200-500 bp fragments
Centrifuge to remove debris and transfer supernatant to new tube
Immunoprecipitation:
Pre-clear chromatin with protein A/G beads
Add HIST1H4A (Ab-35) antibody (2-5 μg per reaction)
Incubate overnight at 4°C with rotation
Add protein A/G beads and incubate for 2-3 hours
Wash beads with increasing stringency buffers
DNA Recovery and Analysis:
Reverse cross-links at 65°C overnight
Treat with RNase A and Proteinase K
Purify DNA using column purification
Analyze by qPCR, sequencing, or other appropriate methods
This protocol has been validated in studies examining histone modifications related to gene expression regulation, such as those investigating HAT1's role in coordinating histone production and acetylation .
Proper control experiments are essential for validating results obtained with HIST1H4A (Ab-35) antibody:
Positive Controls:
Use HeLa acid extract as a positive control sample, which contains abundant histone H4
Include analysis of known regions where histone H4 is expected to be present (such as actively transcribed genes for acetylated forms)
For western blots, compare samples treated with histone deacetylase inhibitors (like sodium butyrate) against untreated samples
Negative Controls:
Include isotype control antibody (rabbit IgG) matching the host species and isotype of the primary antibody
Analyze genomic regions known to lack histone H4 modifications of interest
Include samples where the target protein is depleted (RNAi knockdown when possible)
Antibody Validation Controls:
Peptide competition assay using the immunogen peptide (amino acids around Arg35 of human histone H4)
Demonstrate specificity through western blot showing the appropriate 11-15 kDa band
Compare results with other validated histone H4 antibodies targeting different epitopes
These control experiments help distinguish specific signals from background and confirm antibody specificity, which is particularly important in epigenetic studies where cross-reactivity with similar histone variants can complicate interpretation .
For optimal immunofluorescence results with HIST1H4A (Ab-35) antibody, follow this procedure:
Cell Preparation:
Culture cells on coverslips to 70-80% confluence
Fix cells with 4% paraformaldehyde for 15 minutes at room temperature
Permeabilize with 0.2% Triton X-100 in PBS for 10 minutes
Blocking and Antibody Incubation:
Block with 1-5% BSA in PBS for 1 hour at room temperature
Dilute HIST1H4A (Ab-35) antibody in blocking solution (start with 1:100 dilution and optimize)
Incubate with primary antibody overnight at 4°C
Wash 3 times with PBS, 5 minutes each
Incubate with fluorophore-conjugated secondary antibody (anti-rabbit) for 1 hour at room temperature
Wash 3 times with PBS, 5 minutes each
Nuclear Counterstaining and Mounting:
Counterstain nuclei with DAPI (0.1-1 μg/mL) for 5 minutes
Wash briefly with PBS
Mount coverslips using appropriate mounting medium
Imaging and Analysis:
Capture images using confocal or fluorescence microscopy
For co-localization studies, use appropriate filter sets for multiple fluorophores
Analyze nuclear staining patterns, noting distribution patterns
As demonstrated in reference studies, HIST1H4A should show specific nuclear localization, with distribution patterns that may vary depending on cell cycle stage and treatment conditions . For specialized applications like examining histone modifications in cancer cells, researchers may need to optimize fixation and permeabilization conditions to preserve epitope accessibility.
The HIST1H4A (Ab-35) antibody provides a powerful tool for investigating histone modifications at arginine 35 and their impact on gene expression through several advanced approaches:
ChIP-Seq Analysis:
Perform ChIP with HIST1H4A (Ab-35) antibody followed by next-generation sequencing
Map genome-wide distribution of histone H4 with modified Arg35
Correlate modification patterns with gene expression data (RNA-seq)
Identify enriched motifs in regions with specific modification patterns
Sequential ChIP (Re-ChIP):
First immunoprecipitate with HIST1H4A (Ab-35) antibody
Re-immunoprecipitate with antibodies against other histone marks or transcription factors
Identify genomic regions with co-occurrence of multiple modifications
Determine how Arg35 modifications interact with other epigenetic marks
Integration with Transcriptional Activity Assays:
Combine ChIP data with nascent transcription assays (e.g., GRO-seq)
Use reporter constructs containing promoters of interest
Mutate Arg35 to non-modifiable residues and assess transcriptional outcomes
Study effects of histone deacetylase inhibitors or other epigenetic modulators
Research has shown that histone H4 modifications are particularly important for coordinating transcriptional responses. For example, HAT1 has been found to coordinate histone production and acetylation, binding specifically to H4 gene promoters through an acetate-sensitive promoter element called the H4-box . This represents a feed-forward regulatory circuit where HAT1 captures acetyl groups on nascent histones and drives H4 production through chromatin binding, supporting proliferation and S-phase progression.
Arginine 35 methylation on histone H4 represents an important but less studied modification compared to lysine methylations. Based on current research:
Functional Significance:
Arginine methylation at position 35 can exist in different states: monomethylation, symmetric dimethylation, or asymmetric dimethylation
These modifications influence chromatin structure and recruitment of specific reader proteins
They may antagonize or synergize with nearby modifications, creating a complex "histone code"
Arg35 methylation appears to play roles in both gene activation and repression, depending on cellular context
Regulatory Mechanisms:
Specific protein arginine methyltransferases (PRMTs) target Arg35
Arginine demethylases can remove these marks in response to specific signals
The balance between methylation and demethylation creates dynamic regulation
Crosstalk with nearby modifications, particularly acetylation at lysines, affects recognition and function
Disease Relevance:
The HIST1H4A (Ab-35) antibody specifically recognizing the region around Arg35 enables researchers to investigate these mechanisms in detail, particularly when used in conjunction with modification-specific antibodies.
The HIST1H4A (Ab-35) antibody can be leveraged for sophisticated studies of cell cycle-dependent chromatin dynamics through several approaches:
Cell Cycle Synchronization Studies:
Synchronize cells at different cell cycle phases using methods like double thymidine block or nocodazole treatment
Perform ChIP or immunofluorescence at defined time points using HIST1H4A (Ab-35) antibody
Track changes in histone H4 modifications throughout the cell cycle
Correlate with DNA replication timing and transcriptional activity
Live Cell Imaging with Proximity Ligation Assays:
Combine HIST1H4A (Ab-35) antibody with antibodies against cell cycle regulators
Perform proximity ligation assays to detect interactions in situ
Use microscopy techniques as demonstrated in reference studies to capture changes in protein interactions over time
Correlate with cell cycle markers and progression indicators
Analysis of Chromatin Assembly Pathways:
Study incorporation of newly synthesized histones during S-phase
Investigate the timing of specific post-translational modifications
Examine the role of histone chaperones and assembly factors
Track arginine 35 modifications in relation to DNA replication
Research has demonstrated that HAT1 expression is critical for S-phase progression and maintenance of histone H3 lysine 9 acetylation at proliferation-associated genes, including histone genes themselves . Cells depleted of HAT1 show accumulation in G1 phase (approximately 5.2% increase compared to control), suggesting an important role in cell cycle progression. The HIST1H4A (Ab-35) antibody can help elucidate how arginine 35 modifications contribute to these regulatory networks.
When working with HIST1H4A (Ab-35) antibody, researchers may encounter several common challenges:
Cross-Reactivity Issues:
Problem: The antibody may cross-react with other histone H4 variants due to high sequence conservation.
Solution: Use peptide competition assays with the specific immunogen (peptide sequence around Arg35) to confirm specificity. Compare results with knockout or knockdown controls when available.
Epitope Masking:
Problem: Post-translational modifications near Arg35 may mask the epitope and prevent antibody binding.
Solution: Use multiple antibodies targeting different regions of histone H4. Consider native ChIP methods for certain applications to preserve natural modification states.
Fixation-Related Issues:
Problem: Overfixation can reduce epitope accessibility in IHC and IF applications.
Solution: Optimize fixation conditions (time, temperature, fixative concentration). Consider epitope retrieval methods for tissue samples, such as heat-induced or enzymatic retrieval.
Signal-to-Noise Ratio Problems:
Storage and Handling Issues:
When faced with conflicting results in HIST1H4A (Ab-35) antibody experiments, a systematic approach to data interpretation is essential:
Antibody Validation Assessment:
Confirm antibody lot consistency and validate each new lot
Verify specificity through western blot against purified histones or nuclear extracts
Use peptide competition assays to confirm epitope specificity
Compare with results from alternative antibodies targeting the same or nearby epitopes
Experimental Variables Analysis:
Document all variables between conflicting experiments (cell types, growth conditions, fixation methods)
Consider cell cycle differences, as histone modifications vary throughout cell cycle phases
Evaluate effects of culture conditions on histone modification states
Assess potential technical variations in ChIP efficiency or antibody incubation conditions
Reconciliation Strategies:
Use orthogonal methods to verify findings (e.g., compare ChIP results with mass spectrometry data)
Perform dose-response or time-course experiments to identify optimal conditions
Consider single-cell techniques to address potential heterogeneity in cell populations
Use genetic approaches (CRISPR/Cas9) to validate antibody specificity through targeted mutation of arginine 35
Data Integration Approaches:
Look for consistent patterns across different experimental systems
Weigh results based on strength of controls and methodological rigor
Consider biological context and relevance to the research question
Use computational approaches to integrate multiple datasets and identify robust signals
Cutting-edge techniques for studying histone modifications using HIST1H4A (Ab-35) antibody include:
CUT&RUN and CUT&Tag:
More sensitive alternatives to traditional ChIP
Use targeted nuclease activity to cleave DNA specifically at antibody-bound sites
Require fewer cells and less antibody than conventional ChIP
Provide improved signal-to-noise ratio for studying arginine 35 modifications
Single-Cell ChIP-Seq:
Analyze histone modifications at the single-cell level
Reveal cell-to-cell variation in histone modification patterns
Identify rare cell populations with distinct epigenetic signatures
Map changes during cellular differentiation or disease progression
Mass Spectrometry Integration:
Combine antibody-based enrichment with mass spectrometry analysis
Identify co-occurring modifications on the same histone molecule
Quantify relative abundance of different modification states
Discover novel modifications or combinations affecting Arg35 function
CRISPR-Based Epigenome Editing:
Use dCas9 fused to histone modifiers to manipulate Arg35 modifications at specific genomic loci
Compare results with antibody-based detection of natural modification patterns
Establish causal relationships between specific modifications and gene expression
Create synthetic epigenetic states to test mechanistic hypotheses
Microfluidic Approaches:
Combine antibody-based detection with microfluidic devices
Perform high-throughput analysis of histone modifications
Study dynamic changes in modification patterns over time
Integrate with other genomic and proteomic analyses
These advanced techniques allow researchers to move beyond correlation to establish causal relationships between histone modifications at Arg35 and functional outcomes, providing deeper insights into the biological roles of HIST1H4A modifications in health and disease .
HIST1H4A modifications, particularly at Arg35, undergo significant changes in cancer cells that can be studied using the HIST1H4A (Ab-35) antibody:
Cancer-Specific Modification Patterns:
Altered arginine methylation patterns correlate with specific cancer types
Changes in the balance between different histone H4 modifications affect gene expression programs
Cancer cells often show global hypoacetylation with localized hyperacetylation at oncogenes
The HIST1H4A (Ab-35) antibody can map these changes across the genome
Research Applications in Cancer Models:
Compare histone H4 modification patterns between normal and cancerous tissues
Correlate modifications with tumor stage, grade, and patient outcomes
Study effects of epigenetic drugs (HDAC inhibitors, PRMT inhibitors) on Arg35 modification state
Investigate cancer-specific epigenetic vulnerabilities through ChIP-seq profiling
Experimental Approaches:
Use the antibody in cancer cell lines treated with epigenetic modulators
Apply to patient-derived xenograft models to track modification changes during treatment
Perform IHC on tissue microarrays for biomarker discovery
Integrate with genomic mutation data to identify relationships between genetic and epigenetic alterations
Research has demonstrated that histone H4 acetylation can be detected in cancer cell lines like HeLa cells, with modifications responding to treatments such as sodium butyrate (a histone deacetylase inhibitor) . These techniques are valuable for understanding how epigenetic dysregulation contributes to cancer development and progression.
The study of HIST1H4A modifications in neurodegenerative diseases offers important insights into disease mechanisms and potential therapeutic targets:
Neurodegenerative Disease Relevance:
Histone modifications regulate neuronal gene expression programs crucial for brain function
Arginine methylation affects expression of genes involved in neuronal survival and function
Age-related changes in histone modification patterns may contribute to neurodegenerative processes
Aberrant HIST1H4A modifications have been associated with memory formation and cognitive decline
Experimental Approaches:
Use HIST1H4A (Ab-35) antibody in ChIP-seq studies of brain tissue from disease models
Compare histone modification patterns in affected vs. unaffected brain regions
Study temporal changes in modifications during disease progression
Investigate effects of neuroprotective compounds on histone H4 modification state
Therapeutic Implications:
Identify potential epigenetic targets for intervention
Develop biomarkers for disease progression based on modification patterns
Evaluate efficacy of epigenetic modulators in reversing disease-associated modifications
Design targeted approaches to restore normal histone modification patterns
Integration with Other Data Types:
Correlate histone modifications with transcriptome changes in diseased tissue
Connect genetic risk factors with epigenetic dysregulation
Combine with proteomics data to understand broader chromatin remodeling events
Relate to cellular phenotypes in patient-derived neurons or organoids
While the search results don't specifically address neurodegenerative diseases, the fundamental role of histone H4 in transcriptional regulation makes its modification patterns highly relevant to understanding neuronal gene expression changes in these conditions.
Integrating HIST1H4A (Ab-35) antibody with spatial transcriptomics represents an emerging frontier in epigenomic research:
Technical Integration Approaches:
Combine immunofluorescence using HIST1H4A (Ab-35) antibody with spatial transcriptomics platforms
Perform sequential immunostaining and RNA detection on the same tissue sections
Develop computational methods to integrate histone modification data with spatially resolved gene expression
Use multi-modal approaches to simultaneously detect histone modifications and RNA in situ
Experimental Design Considerations:
Optimize tissue preparation protocols to preserve both epitope and RNA integrity
Select anatomical regions of interest with known biological heterogeneity
Include relevant cellular markers to identify specific cell types
Design appropriate controls to account for technical variation across modalities
Analytical Frameworks:
Develop statistical methods to correlate spatial patterns of histone modifications with gene expression
Create visualization tools to represent multi-modal spatial data
Apply machine learning approaches to identify spatial domains with distinct epigenetic signatures
Integrate with single-cell approaches for comprehensive epigenetic profiling
Biological Applications:
Map tissue-specific epigenetic regulatory domains in development and disease
Identify local microenvironmental effects on histone modification patterns
Study epigenetic heterogeneity within complex tissues like tumors or brain
Investigate spatial relationships between cells with different epigenetic states
While specific examples using HIST1H4A (Ab-35) antibody in spatial genomics are not directly mentioned in the search results, the antibody's validated use in immunohistochemistry and immunofluorescence makes it a suitable candidate for such advanced spatial applications.
Targeted proteomics approaches using HIST1H4A (Ab-35) antibody offer promising future directions for histone modification research:
Antibody-Enhanced Mass Spectrometry:
Use HIST1H4A (Ab-35) antibody for immunoprecipitation before MS analysis
Enrich for histone H4 protein containing the Arg35 region
Identify co-occurring modifications and protein interaction partners
Quantify relative abundance of different modification states at and around Arg35
Proximity Labeling Applications:
Develop antibody conjugates with proximity labeling enzymes (BioID, APEX)
Map the protein interaction network around modified histones
Identify readers, writers, and erasers specific to Arg35-modified histone H4
Study dynamic changes in interaction partners under different cellular conditions
Single-Cell Proteomics Integration:
Adapt antibody-based detection for microfluidic single-cell proteomics
Profile histone modification heterogeneity at single-cell resolution
Correlate with other epigenetic marks and cellular phenotypes
Track proteome-wide responses to changes in histone modification state
Therapeutic Applications:
Develop targeted protein degradation approaches (PROTACs) directed at specifically modified histones
Use antibody-drug conjugates to target cells with aberrant modification patterns
Create screening platforms to identify compounds that modulate specific histone modifications
Monitor treatment responses through changes in histone modification profiles