To create the phospho-histone H3.1 (S1) recombinant monoclonal antibody, genes encoding the HIST1H3A antibody are first cloned, encompassing both heavy and light chains. These cloned genes are then inserted into expression vectors, which are introduced into host cells via transfection. The host cells are responsible for the production and secretion of the antibody. Affinity chromatography is employed to ensure the antibody's purity, after which it undergoes rigorous functionality testing in various applications, including ELISA, WB, ICC, and IF, enabling accurate detection of the human HIST1H3A protein phosphorylated at S1.
Phosphorylation at H3.1 S1 can promote chromatin condensation thus leading to gene repression. During mitosis, histone H3.1 S1 phosphorylation plays a role in chromosome condensation and segregation. It helps ensure accurate cell division by regulating chromosomal architecture. Phosphorylated H3.1 S1 is also associated with DNA damage response and repair processes. H3.1 S1 phosphorylation is also linked to cell cycle regulation, epigenetic signaling, and coordinated gene regulation.
The phospho-histone H3.1 (S1) recombinant monoclonal antibody is meticulously crafted through a process involving gene cloning, expression vector insertion, and cell transfection. This antibody is specifically designed to recognize the human HIST1H3A protein when phosphorylated at serine 1. The genes encoding the HIST1H3A antibody, encompassing both heavy and light chains, are first cloned. These cloned genes are then inserted into expression vectors, which are subsequently introduced into host cells via transfection. The host cells then produce and secrete the antibody, which undergoes affinity chromatography for purification. Subsequently, the antibody undergoes rigorous functionality testing in various applications, including ELISA, Western blotting, immunocytochemistry, and immunofluorescence, ensuring its accuracy in detecting the human HIST1H3A protein phosphorylated at S1.
Phosphorylation at H3.1 S1 is a key regulator of chromatin condensation, which in turn affects gene expression. During mitosis, histone H3.1 S1 phosphorylation plays a crucial role in chromosome condensation and segregation, ensuring accurate cell division by regulating chromosomal architecture. Phosphorylated H3.1 S1 is also actively involved in DNA damage response and repair processes. Furthermore, H3.1 S1 phosphorylation is intricately linked to cell cycle regulation, epigenetic signaling, and coordinated gene regulation.
Histone H3.1 serves as a core component of the nucleosome, a fundamental structure responsible for wrapping and compacting DNA into chromatin. This compaction restricts DNA accessibility to cellular machineries that require DNA as a template for various processes. Consequently, histones play a pivotal role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. The accessibility of DNA is dynamically regulated by a complex interplay of post-translational modifications of histones, collectively known as the histone code, along with nucleosome remodeling.
Histone H3.1 is one of the core components of the nucleosome, the basic unit of chromatin. It belongs to the H3 family of histone proteins and differs from the H3.3 variant by specific amino acid residues. H3.1 is primarily expressed during S-phase and incorporated into chromatin during DNA replication, making it a replication-coupled (RC) histone .
Phosphorylation at serine 1 (S1) of histone H3.1 is associated with:
Promotion of chromatin condensation leading to gene repression
Regulation of chromosome condensation during mitosis
Involvement in DNA damage response and repair mechanisms
Contribution to cell cycle regulation and epigenetic signaling
Unlike other phosphorylation sites such as S10 or S28, the S1 phosphorylation appears to be more directly involved in transcriptional silencing rather than activation.
Recombinant monoclonal antibodies offer several advantages over traditional hybridoma-derived antibodies:
Feature | Traditional Monoclonal Antibodies | Recombinant Monoclonal Antibodies |
---|---|---|
Production method | Hybridoma cell culture | In vitro expression systems with cloned antibody sequences |
Consistency | May vary between lots | Higher lot-to-lot consistency |
Specificity | Good | Better specificity and sensitivity |
Formulation | Animal-derived components may be present | Animal origin-free formulations possible |
Customization | Limited | Can be engineered for specific applications |
Reproducibility | Subject to hybridoma drift | Sequence-defined, highly reproducible |
For histone research specifically, recombinant antibodies can be designed to recognize precise post-translational modifications with minimal cross-reactivity to other histone marks, which is critical when studying the nuanced epigenetic code .
Based on manufacturer recommendations across multiple providers:
Application | Recommended Dilution Range | Notes |
---|---|---|
Western Blot (WB) | 1:500-1:5000 | Lower dilutions (1:500-1:2000) typically yield optimal results |
Immunocytochemistry (ICC) | 1:50-1:500 | Fixation method can impact results |
Immunofluorescence (IF) | 1:30-1:200 | May require optimization based on cell type |
ELISA | Varies by kit/protocol | Follow manufacturer's specific recommendations |
Optimization is essential as detection sensitivity may vary depending on:
Cell type or tissue used
Fixation and permeabilization methods
Extent of S1 phosphorylation in your experimental system
Rigorous validation of phospho-specific antibodies is crucial to ensure experimental accuracy:
Peptide competition assay: Pre-incubate the antibody with phosphorylated and non-phosphorylated peptides corresponding to the target epitope. A specific signal should be blocked only by the phosphorylated peptide.
Phosphatase treatment controls: Treat one sample with lambda phosphatase to remove phosphorylation and compare to untreated samples. The signal should disappear in phosphatase-treated samples.
Knock-in/knock-out validation: Use cells where the target site has been mutated (S1A) to prevent phosphorylation or use histone H3.1 knockout models as negative controls.
Kinase inhibition: Use specific kinase inhibitors that block the phosphorylation of the S1 residue to confirm specificity.
Orthogonal techniques: Confirm results using alternative methods such as mass spectrometry to detect the phosphorylation state of H3.1 S1.
Positive controls: Include samples known to have high levels of H3.1 S1 phosphorylation, such as cells synchronized at specific cell cycle stages .
Optimal detection of H3.1 S1 phosphorylation requires careful sample preparation:
Cell Culture Considerations:
Synchronization: Use techniques like thymidine block combined with nocodazole to enrich for mitotic cells when H3.1 phosphorylation is often elevated
Stress induction: Various cellular stresses can alter histone phosphorylation patterns
Inhibitor treatments: Calyculin A (phosphatase inhibitor) can be used to preserve phosphorylation states
Sample Preparation for Western Blot:
Add phosphatase inhibitors (sodium fluoride, sodium orthovanadate, β-glycerophosphate) to all buffers
Use acid extraction methods for histone isolation to preserve phosphorylation marks
Keep samples cold during processing to minimize phosphatase activity
Consider direct lysis in SDS sample buffer for rapid fixation of phosphorylation states
For Immunofluorescence:
Quick fixation with 4% paraformaldehyde is often preferred
Brief permeabilization with 0.1-0.2% Triton X-100
Include phosphatase inhibitors in buffers
Different histone H3 phosphorylation sites present unique technical challenges:
Phosphorylation Site | Detection Characteristics | Technical Considerations |
---|---|---|
H3.1 S1 | Less studied; can be masked by histone tail binding proteins | May require specific extraction methods; typically lower abundance than S10 |
H3 S10 | Widely studied; strong mitotic marker | High signal in M-phase; serves as a positive control for many experiments |
H3 S28 | Similar dynamics to S10 but less abundant | May require more sensitive detection methods |
H3.3 S31 | Variant-specific mark; not present on H3.1 | Requires variant-specific antibodies; useful for distinguishing H3 variants |
H3 T3 | Often co-occurs with S10 phosphorylation | Can be detected with specific antibodies for combinatorial analysis |
For multiplex detection of different modifications:
Consider sequential detection with appropriate stripping between antibodies
Use fluorescent multiplex Western blotting with differently labeled secondary antibodies
For imaging applications, select antibodies from different host species to allow simultaneous detection
These phosphorylation marks have distinct functions and dynamics in cellular processes:
Feature | H3.1 S1 Phosphorylation | H3 S10 Phosphorylation |
---|---|---|
Cell cycle pattern | Present in interphase and mitosis | Dramatically increases during mitosis |
Primary function | Gene repression; chromatin condensation | Mitotic chromosome condensation; gene activation |
Associated kinases | Multiple candidates including MSK1/2 | Aurora B kinase (mitotic); MSK1/2, PKA (interphase) |
Interplay with other marks | May prevent methylation/acetylation of nearby residues | Disrupts HP1 binding to adjacent H3K9me3 |
Gene expression effects | Generally repressive | Context-dependent: activating in interphase, associated with condensed chromatin in mitosis |
Conservation | Highly conserved across species | Highly conserved across species |
The spatial and temporal dynamics of these marks reflect their distinct roles in chromatin biology. While S10 phosphorylation is a robust marker of mitosis widely used in cell cycle studies, S1 phosphorylation appears to play more subtle roles in transcriptional regulation and chromatin organization .
Several complementary approaches can identify and characterize kinases that phosphorylate H3.1 at S1:
In vitro kinase assays:
Incubate purified recombinant H3.1 with candidate kinases
Detect phosphorylation using Phospho-H3.1 (S1) antibodies
Quantify using Western blot or ELISA-based methods
Can be performed with γ-32P-ATP for radiometric analysis
Kinase inhibitor screens:
Treat cells with panels of specific kinase inhibitors
Monitor changes in H3.1 S1 phosphorylation levels
Create dose-response curves to identify potential kinases
Genetic approaches:
CRISPR/Cas9 knockout or knockdown of candidate kinases
Overexpression of constitutively active or dominant negative kinase mutants
Monitor effects on global H3.1 S1 phosphorylation levels
Immunoprecipitation-based methods:
Co-immunoprecipitation to identify kinases physically associated with H3.1
Phosphorylation of immunoprecipitated H3.1 using cell extracts
Mass spectrometry to identify kinases in complex with H3.1
HTRF (Homogeneous Time-Resolved Fluorescence) assays:
Distinguishing between H3 variants requires specialized techniques due to their high sequence similarity:
Variant-specific antibodies:
Use antibodies that recognize the unique regions of H3.1 or H3.3
H3.3 S31 phosphorylation is a variant-specific mark (not present in H3.1)
For H3.1, antibodies targeting regions containing A31 (vs S31 in H3.3) can be used
Sequential chromatin immunoprecipitation (ChIP):
First IP with variant-specific antibody
Second IP with phosphorylation-specific antibody
Identifies genomic regions containing specific variant with specific modifications
Mass spectrometry approaches:
Can distinguish variant-specific peptides and their modifications
Enables quantitative analysis of modification stoichiometry
Can detect combinatorial modifications not possible with antibodies
Stable isotope labeling by amino acids in cell culture (SILAC):
Label histones during protein synthesis
Monitor turnover rates of different variants and their modifications
Particularly useful for studying replication-dependent vs. independent incorporation
Exogenous tagged histones:
H3.1 S1 phosphorylation functions within the complex network of histone modifications that constitute the histone code:
Cross-talk with adjacent modifications:
S1 phosphorylation may prevent methylation or acetylation of neighboring residues (K4, R2)
Can influence reader protein binding to nearby modified residues
May alter accessibility of the histone tail to other modifying enzymes
Combinatorial effects with other modifications:
The combination of S1ph with specific lysine methylation states (K4me3, K9me3, K27me3) may signal for distinct functional outcomes
Sequential establishment of modifications during cell cycle progression creates specific chromatin states
Experimental approaches to study cross-talk:
Use recombinant histones with defined modifications for in vitro studies
Mass spectrometry to identify co-occurring modifications
Sequential ChIP to identify genome regions with multiple modifications
Antibodies specifically recognizing combinatorial modifications
Functional consequences:
S1 phosphorylation may influence the binding of chromatin remodeling complexes
Can affect higher-order chromatin structure and accessibility
May target specific genomic regions for silencing or activation depending on context
Cell cycle dynamics:
Researchers frequently encounter these challenges when detecting H3.1 S1 phosphorylation:
Issue | Possible Causes | Solutions |
---|---|---|
Weak or no signal | Low phosphorylation levels; Phosphatase activity during sample preparation; Antibody concentration too low | Add phosphatase inhibitors to all buffers; Increase antibody concentration; Enrich for cells with high phosphorylation; Consider signal amplification methods |
High background | Non-specific binding; Concentration too high; Inadequate blocking | Optimize antibody dilution; Use alternative blockers (BSA vs milk); Include competitive peptides; Increase washing stringency |
Inconsistent results | Phosphorylation instability; Cell cycle variation; Lot-to-lot antibody variation | Standardize sample preparation; Synchronize cells; Use recombinant antibodies with better lot consistency |
Cross-reactivity | Antibody recognizing other phosphorylation sites | Validate with peptide competition; Use S1A mutant controls; Consider alternative antibody clones |
Epitope masking | Protein-protein interactions blocking antibody access | Use alternative extraction methods; Consider different fixation protocols for IF/ICC |
For Western blotting specifically:
Transfer efficiency: Use transfer methods optimized for low molecular weight proteins
Loading controls: Consider using total H3 antibodies or non-histone loading controls
Quantification: Normalize phospho-signal to total H3 signal for accurate comparisons
Optimizing phospho-histone detection in tissue samples requires special considerations:
Fixation protocols:
Overfixation can mask epitopes; consider shorter fixation times (4-24 hours)
Freshly prepared 4% paraformaldehyde is often optimal
Avoid acidic fixatives that may affect phospho-epitopes
Consider perfusion fixation for animal tissues when possible
Antigen retrieval methods:
Heat-induced epitope retrieval (HIER) in citrate buffer (pH 6.0) or Tris-EDTA (pH 9.0)
Optimize time and temperature (typically 95-100°C for 15-30 minutes)
Allow slow cooling to room temperature
Include phosphatase inhibitors in retrieval buffers
Signal amplification:
Consider tyramide signal amplification systems
Polymer-based detection systems may provide better sensitivity
Avoid excessive amplification that can increase background
Controls and validation:
Include phosphatase-treated sections as negative controls
Use tissues known to contain cells with high H3.1 S1 phosphorylation (e.g., proliferating tissues)
Consider mitotic cells (identifiable by DAPI) as internal positive controls
Quantification approaches:
Several advanced technologies are enhancing histone phosphorylation research:
Single-cell analysis approaches:
Mass cytometry (CyTOF) for multiparameter analysis of histone modifications
Single-cell Western blotting for phospho-histone analysis
Imaging mass spectrometry for spatial distribution of modifications
High-sensitivity detection methods:
HTRF (Homogeneous Time-Resolved Fluorescence) assays provide plate-based detection without Western blotting
Luminex bead-based multiplex assays for simultaneous detection of multiple phosphorylation sites
Digital ELISA platforms offering femtomolar sensitivity
Genomic approaches:
CUT&RUN and CUT&Tag methods for improved chromatin profiling
ChIP-seq combining phospho-specific antibodies with next-generation sequencing
CHIC/HiChIP methods combining chromatin conformation with histone modification analysis
Live-cell imaging techniques:
FRET-based biosensors for real-time monitoring of histone phosphorylation
Specific fluorescent probes for visualizing phosphorylated histones
Optogenetic tools to manipulate kinase activity with spatial and temporal precision
Synthetic biology approaches:
While H3.1 S1 phosphorylation is less studied than other histone marks in disease contexts, emerging research suggests several important connections:
Cancer associations:
Altered H3.1 S1 phosphorylation patterns observed in several cancer types
May contribute to aberrant gene silencing of tumor suppressors
Dysregulation of kinases responsible for S1 phosphorylation often occurs in tumors
Potential biomarker for specific cancer subtypes or progression stages
Neurodegenerative disorders:
Altered histone phosphorylation patterns observed in models of neurodegeneration
May contribute to transcriptional dysregulation in affected neurons
Could represent potential therapeutic targets
Inflammatory conditions:
Inflammation-associated signaling can affect histone phosphorylation patterns
May contribute to epigenetic reprogramming during chronic inflammation
Developmental disorders:
Proper histone modification dynamics are essential for development
Mutations affecting histone H3.1 or its modifying enzymes linked to developmental abnormalities
Experimental approaches:
Working with clinical specimens presents unique challenges for histone phosphorylation analysis:
Sample preservation:
Phosphorylation marks are labile and sensitive to post-collection handling
Flash freezing or immediate fixation is optimal
Document ischemia time and preservation method
Consider phosphatase inhibitors during sample collection
FFPE tissue considerations:
Formalin fixation can affect epitope accessibility
Standardize fixation protocols (time, buffer, temperature)
Optimize antigen retrieval methods specifically for phospho-epitopes
Consider using phosphatase inhibitors in retrieval buffers
Controls and normalization:
Use matched normal tissues when possible
Consider adjacent normal tissue within samples as internal control
Normalize phospho-signal to total H3 levels
Include phosphatase-treated sections as technical controls
Quantification methods:
Digital pathology approaches for objective quantification
Consider H-score methods (intensity × percentage positive cells)
Blinded scoring by multiple observers
Use image analysis software for consistent threshold application
Integration with other biomarkers: