The HIST1H3A recombinant monoclonal antibody is a rabbit-derived antibody engineered to target the human histone H3.1 protein. Key features include:
Attribute | Details |
---|---|
Immunogen | Synthetic peptide derived from human HIST1H3A (e.g., acetylated Lys18) |
Host | Rabbit |
Purification | Affinity chromatography |
Isotype | IgG |
Gene Cloning: Antibody genes (heavy/light chains) are cloned from immunized rabbits .
Vector Integration: Genes are inserted into expression vectors for transfection into host cells (e.g., suspension cells) .
Expression and Purification: Host cells secrete antibodies, which are purified via affinity chromatography .
Validation: Functionality is confirmed via ELISA, Western blot (WB), immunofluorescence (IF), immunohistochemistry (IHC), and flow cytometry (FC) .
This antibody is versatile for studying histone H3.1 dynamics in epigenetics, cell cycle regulation, and cancer research.
DNA Damage Response: Phosphorylated H3.1 (S1) detection during mitosis .
Epigenetic Markers: Acetylated HIST1H3A (K18) detection for gene activation studies .
Cancer Research: Identifying histone variants in tumor samples .
Code | CSB-RA010418A10phHU |
---|---|
Target | H3.1 phosphorylated at Serine 10 |
Applications | WB, ICC, IF |
Biological Role | Chromatin condensation during mitosis; DNA repair regulation . |
Code | CSB-RA547773A0HU |
---|---|
Target | H3.1 acetylated at Lysine 18 |
Applications | IHC, IF, FC |
Biological Role | Gene activation via chromatin remodeling . |
Epigenetic modifiers enhance recombinant antibody expression in CHO cells:
HDAC/LSD1 Inhibitors: Increase histone acetylation/methylation, boosting mRNA levels of HIST1H3A antibodies .
The HIST1H3A recombinant monoclonal antibody preparation involves obtaining the HIST1H3A antibody genes, introducing them into suitable host cells, and synthesizing HIST1H3A antibodies using a cell expression and translation system. This method significantly enhances the purity and stability of the synthesized HIST1H3A recombinant monoclonal antibodies while also increasing their affinity and specificity. This HIST1H3A recombinant monoclonal antibody undergoes affinity chromatography purification and has been tested in ELISA, WB, IHC, and IF assays. It exhibits the ability to recognize both human and mouse HIST1H3A protein.
HIST1H3A, also known as histone H3.1, plays a crucial role as a structural protein within chromatin. It aids in packaging DNA and regulating gene expression. The dynamic modifications and interactions of HIST1H3A with other proteins are essential for the intricate regulation of various cellular processes, including gene transcription, DNA replication, DNA repair, and cell division.
HIST1H3A encodes histone H3.1, one of the primary histone H3 variants found in metazoans. Histone H3.1 functions as a core component of nucleosomes, the fundamental units of chromatin structure that package DNA. Unlike variant H3.3, histone H3.1 is primarily expressed during S phase and incorporated into chromatin through replication-coupled pathways .
Epigenetic research has demonstrated that histone H3.1 plays crucial roles in chromatin dynamics, DNA accessibility regulation, and gene expression control. The post-translational modifications on histone H3.1 (including acetylation and methylation) serve as epigenetic marks that influence chromatin state and transcriptional activity. These modifications can be regulated by epigenetic modulators, making HIST1H3A a central target for understanding gene regulation mechanisms .
To study HIST1H3A effectively, researchers should consider:
Different expression patterns between histone variants
Incorporation mechanisms (replication-dependent vs independent)
Post-translational modification profiles
Interaction with specific histone chaperones
Recombinant monoclonal antibodies against HIST1H3A offer several methodological advantages over polyclonal alternatives in chromatin research:
Specificity advantages:
Target single epitopes with higher precision
Demonstrate reduced batch-to-batch variation
Allow for better discrimination between histone H3 variants that differ by only a few amino acids
Provide more consistent results in experiments requiring quantitative analysis
For experimental validation, researchers should perform Western blot analysis across multiple cell lines with known histone variant expression patterns, as demonstrated in search result where validation was conducted using A549, C6, AML-12, and HepG2 cell lysates to confirm antibody specificity .
Based on the search results, HIST1H3A antibodies demonstrate utility across multiple experimental applications. The methodological approaches for each application include:
Chromatin Immunoprecipitation (ChIP):
Fixation protocol: Formaldehyde fixation (10 minutes optimal)
Chromatin amount: ~25 mg recommended
Antibody quantity: 3 μg antibody with 20 ml Protein A/G sepharose beads
Controls: Include non-specific antibody controls
Quantification: Real-time PCR (Taqman approach) for reliable quantification
Western Blot:
Protein loading: 20-40 μg total protein (cell-type dependent)
Conditions: Reducing conditions on 4-12% Bis-tris gels
Expected band size: 15-17 kDa
Blocking: 2-5% BSA solution
Immunohistochemistry (IHC):
Fixation: Paraformaldehyde fixation
Permeabilization: 0.05% Triton X-100 (30 minutes)
Blocking: 5% BSA (1 hour)
Antigen retrieval: Heat mediation in sodium citrate pH 6
Primary antibody dilution: 1:100-1:500
Immunocytochemistry/Immunofluorescence (ICC/IF):
Fixation options: 100% methanol (5 min) or paraformaldehyde
Permeabilization: 0.1% PBS-Triton X-100 (5 minutes)
Blocking: 0.2% fish scale gelatin or 1% BSA with 10% normal goat serum
Antibody dilution: 1:300 in PBS with 0.2% gelatin
Effective ChIP experimental design with HIST1H3A antibodies requires careful optimization of several parameters:
Critical methodological factors:
Crosslinking optimization: Formaldehyde crosslinking for 10 minutes has been validated, but time should be optimized for specific cell types
Chromatin fragmentation: Sonication parameters should be optimized to achieve fragment sizes of 200-500bp
Antibody concentration: Begin with 3 μg antibody per 25 mg chromatin and titrate as needed
Control selection: Include both input control and IgG negative control (as demonstrated in search result )
Quantification approach: Implement real-time PCR with appropriate normalization to input
For cell-type specific optimization, researchers should consider:
Crosslinking efficiency varies by cell type (adherent vs suspension)
Nuclear accessibility differs between cell types
Chromatin compaction state affects antibody accessibility
Cell cycle status influences histone variant distribution
Advanced ChIP applications like ChIP-seq require additional considerations including library preparation quality control, sequencing depth, and bioinformatic analysis pipelines optimized for histone modification data.
Distinguishing between highly similar histone H3 variants (like H3.1 encoded by HIST1H3A versus H3.3) requires specialized methodological approaches:
Epitope selection strategy:
Target variant-specific amino acid differences (H3.1 and H3.3 differ by only 4-5 amino acids)
Focus on regions containing S31 (present in H3.3 but not H3.1/H3.2)
Consider antibodies recognizing specific post-translational modifications unique to each variant
Validate specificity using recombinant proteins of each variant
Experimental validation approaches:
Competitive binding assays: Use peptides corresponding to different H3 variants
Immunoprecipitation-mass spectrometry: Verify precise variant recognition
Cell line panel testing: As demonstrated in search result , test across multiple cell lines with different variant expression
Genetic models: Test in cell lines with knockout/knockdown of specific variants
Sequential ChIP: For distinguishing variant-specific modification patterns
When interpreting results, researchers must account for:
Cell cycle phase effects on variant abundance
Tissue-specific expression patterns
Post-translational modification interference with epitope recognition
Cross-reactivity potential with other histone family members
Optimizing Western blot protocols for HIST1H3A detection requires attention to several technical parameters:
Sample preparation optimization:
Protein extraction: Use specialized histone extraction protocols with acid extraction
Loading amount: 10-40 μg total protein depending on cell type (higher amounts recommended for yeast samples)
Reduction conditions: Use fresh reducing agents for consistent results
Size separation: 4-12% Bis-tris gel under MES buffer system
Transfer and detection optimization:
Transfer parameters: 30V for 70 minutes to nitrocellulose membrane
Blocking solution: 2-5% BSA superior to milk-based blockers
Primary antibody: Overnight incubation at 4°C at 1:500 dilution
Secondary antibody: HRP-conjugated anti-rabbit IgG
Detection system: ECL technique with exposure times of 7-30 seconds
Critical control recommendations:
Include positive control lysates from cells with known HIST1H3A expression
Run molecular weight markers to confirm expected 15-17 kDa band size
Consider loading controls appropriate for nuclear proteins
Include validation across multiple cell lines as demonstrated in search result
Implementing HIST1H3A antibodies for studying epigenetic modifications requires integrated methodological approaches:
Experimental design strategies:
Sequential ChIP (Re-ChIP): First immunoprecipitate with HIST1H3A antibody, then with modification-specific antibodies
Comparative ChIP-seq: Compare HIST1H3A distribution with specific histone modifications
Mass spectrometry integration: Combine immunoprecipitation with mass spectrometry to identify HIST1H3A-associated modification patterns
Epigenetic modulator treatment: Test how epigenetic drugs affect HIST1H3A modifications
Research has demonstrated that epigenetic modulators significantly impact histone H3 modifications. For example, the dual-HDAC/LSD1 inhibitor I-4 increased histone H3 acetylation and methylation levels, correlating with increased recombinant protein expression. Specifically, treatment with 2 μM I-4 resulted in a 1.94-fold increase in monoclonal antibody titer and a 2.43-fold increase in specific monoclonal antibody production .
The mechanistic pathway involves:
HDAC inhibition leading to increased histone acetylation
LSD1 inhibition affecting histone methylation status
Downstream effects on chromatin accessibility
Altered gene expression patterns, including transgene expression
Investigating HIST1H3A interactions with histone chaperones requires specialized techniques addressing both physical interactions and functional relationships:
Interaction mapping approaches:
Co-immunoprecipitation (Co-IP): Use HIST1H3A antibodies to pull down associated chaperone proteins
Proximity ligation assay (PLA): Visualize in situ interactions between HIST1H3A and candidate chaperones
FRET/BRET analysis: Monitor real-time interactions in living cells
Protein fragment complementation: Assess direct physical interactions
Cross-linking mass spectrometry: Map interaction interfaces at amino acid resolution
Based on search result , histone H3 variants interact with specific chaperone proteins that facilitate their incorporation into chromatin. While H3.3 interacts with HIRA or DAXX for replication-independent deposition, H3.1 (encoded by HIST1H3A) has distinct chaperone interactions for replication-coupled incorporation .
Functional analysis methods include:
Chaperone depletion studies to assess effects on HIST1H3A incorporation
Chromatin assembly assays with purified components
Cell cycle synchronization to study temporal dynamics of interactions
Domain mapping to identify critical interaction regions
Competitive binding assays with different histone variants
Researchers should consider controls including:
Interaction studies with other H3 variants for comparison
Mutational analysis of interaction interfaces
Antibody epitope accessibility verification
HIST1H3A antibodies enable sophisticated investigation of histone variant roles in disease and development through several methodological approaches:
Disease-specific applications:
Cancer research: Analyze HIST1H3A distribution in tumor vs. normal tissues (search result mentions H3.3 mutations in cancer)
Neurodevelopmental disorders: Map histone variant transitions during neural development
Aging research: Track age-associated changes in variant distribution
Inflammatory conditions: Correlate variant patterns with inflammatory signatures
Developmental biology applications:
Lineage specification: Track HIST1H3A distribution during differentiation
Cellular reprogramming: Map variant switching during induced pluripotency
Embryonic development: Analyze spatial-temporal dynamics of histone variants
Tissue regeneration: Investigate variant roles in regenerative processes
Methodological implementation requires:
Tissue-specific immunohistochemistry optimization (as shown in search result )
Development stage-specific sampling strategies
Integration with other epigenetic profiling methods
Correlation with gene expression changes
Technical considerations when studying HIST1H3A in disease contexts include:
Preservation of native chromatin architecture in clinical samples
Appropriate control tissue selection
Consideration of heterogeneity within diseased tissues
Integration of genetic and epigenetic data
HIST1H3A antibody research faces several technical challenges that require systematic troubleshooting approaches:
Common specificity issues:
Cross-reactivity with other H3 variants: H3.1 and H3.3 differ by only 4-5 amino acids, making specific recognition challenging
Post-translational modification interference: Modifications near antibody epitopes can block recognition
Batch-to-batch variation: Particularly problematic with polyclonal antibodies
Fixation-dependent epitope masking: Critical for immunohistochemistry applications
Non-specific nuclear protein binding: Can create background in nuclear preparations
Methodological solutions:
Antibody validation strategy:
Protocol optimization approaches:
Verification techniques:
Mass spectrometry validation of immunoprecipitated proteins
Orthogonal detection methods alongside antibody-based approaches
Sequential epitope mapping to confirm specific recognition sites
Pre-adsorption tests to eliminate cross-reactivity
When designing experiments to study histone H3 variant dynamics, researchers should implement a comprehensive methodological framework:
Experimental design principles:
Cell cycle considerations:
Technical controls for variant-specific studies:
Include pan-H3 antibodies alongside variant-specific antibodies
Implement genetic controls (overexpression or knockdown)
Use recombinant proteins as standards for quantification
Include spike-in controls for quantitative comparisons
Multi-technique validation strategy:
Combine ChIP-seq with RNA-seq to correlate variant localization with transcription
Implement imaging approaches alongside biochemical methods
Use nascent chromatin capture for temporal dynamics
Apply quantitative proteomics to measure variant stoichiometry
Biological replication requirements:
Minimum three biological replicates recommended
Account for cell type-specific variant distributions
Consider developmental stage effects on variant patterns
Address inter-individual variation in primary samples
Analysis framework:
Normalize for total histone H3 levels when quantifying variants
Implement appropriate statistical tests for variant ratio comparisons
Consider genomic distribution patterns alongside absolute levels
Correlate with functional genomic features (enhancers, promoters, etc.)
Rigorous quality control measures are essential for reliable results with HIST1H3A recombinant monoclonal antibodies:
Pre-experimental quality control:
Antibody validation requirements:
Storage and handling protocols:
Experimental quality control:
Control inclusion requirements:
Positive controls: Cell lines with known HIST1H3A expression
Negative controls: Isotype controls or pre-immune serum
Technical controls: Secondary-only controls for background assessment
Blocking peptide controls: To confirm epitope specificity
Quantitative assessment metrics:
Signal-to-noise ratio measurement
Antibody titration to determine optimal concentration
Reproducibility across technical and biological replicates
Cross-platform validation when possible
Documentation requirements:
Record antibody catalog number, lot number, and concentration
Document all experimental conditions in detail
Maintain validation data for each antibody lot
Report all quality control measures in publications
Following these quality control measures will ensure robust, reproducible results when working with HIST1H3A recombinant monoclonal antibodies in research applications.