The Phospho-HIST1H1B (S17) Antibody (PAC065057) is a specialized immunological reagent produced in rabbits that specifically recognizes and binds to the phosphorylated form of Hist1H1B protein at serine 17 in human samples . HIST1H1B, also known as Histone H1.5, belongs to the histone H1 family and plays critical roles in chromatin organization, DNA packaging, and transcriptional regulation within the cell nucleus. The phosphorylation at serine 17 represents a specific post-translational modification that affects the protein's functional properties and interactions within the nuclear environment . This antibody's high specificity makes it an invaluable tool for researchers investigating histone modifications and their implications in cellular processes.
The antibody is typically supplied as a liquid preparation and requires specific storage conditions for maintaining its reactivity and specificity. The product is formulated in a buffer containing preservatives like Proclin 300 and glycerol, which help maintain stability during storage and prevent bacterial contamination . Proper handling and storage are essential to preserve the antibody's functionality, with recommendations typically involving storage at -20°C to -80°C to prevent degradation and maintain consistent performance across experiments.
HIST1H1B (Histone H1.5) is a linker histone that plays crucial roles in chromatin structure and function. The protein is involved in higher-order chromatin organization and acts as a regulator of DNA accessibility, influencing processes such as gene expression, DNA replication, and DNA repair . The molecular weight of HIST1H1B is approximately 22,580 daltons, and it primarily localizes to the nucleus where it associates with nucleosomes and linker DNA.
Phosphorylation of HIST1H1B at serine 17 represents a specific post-translational modification that affects the protein's interactions and functions within the chromatin environment. This modification has been linked to various cellular processes, including:
Chromatin remodeling and structural changes
Regulation of gene expression through altered DNA accessibility
Cell cycle progression and mitotic events
Response to DNA damage and cellular stress
Involvement in epigenetic reprogramming mechanisms
The specific phosphorylation at serine 17 may influence the binding affinity of the histone to DNA, thereby affecting chromatin compaction and accessibility to transcription factors and other regulatory proteins . Understanding these phosphorylation events provides insights into the dynamic regulation of chromatin structure and function in normal cellular processes and disease states.
The Phospho-HIST1H1B (S17) Antibody serves as a valuable tool in various research applications focused on understanding histone modifications and their roles in cellular processes. This antibody has been validated for specific experimental techniques, enabling researchers to investigate the dynamics of HIST1H1B phosphorylation in different cellular contexts.
The primary applications of Phospho-HIST1H1B (S17) Antibody include:
Western Blot Analysis: Enables detection of phosphorylated HIST1H1B in cell or tissue lysates, allowing for quantitative assessment of phosphorylation levels under different experimental conditions .
Enzyme-Linked Immunosorbent Assay (ELISA): Provides a platform for quantitative measurement of phosphorylated HIST1H1B in complex biological samples .
Immunocytochemistry (ICC): Allows visualization of the subcellular localization of phosphorylated HIST1H1B in fixed cells, providing spatial information about phosphorylation patterns .
Each application requires specific optimization of antibody dilutions and experimental conditions to achieve optimal results. For ICC applications, a recommended dilution range of 1:10 to 1:100 has been established, though researchers may need to adjust these parameters based on specific sample types and detection methods .
The Phospho-HIST1H1B (S17) Antibody finds application in various research areas, including:
Epigenetics research: Investigating the role of histone modifications in gene regulation and chromatin structure
Cancer biology: Examining alterations in histone phosphorylation patterns in cancer cells and their potential as biomarkers or therapeutic targets
Cell cycle studies: Analyzing the dynamics of histone phosphorylation during different phases of the cell cycle
Developmental biology: Understanding the role of histone modifications in cellular differentiation and development
Neuroscience: Investigating epigenetic mechanisms in neuronal function and neurological disorders
Researchers in these fields utilize the antibody to gain insights into the specific roles of HIST1H1B phosphorylation in normal cellular functions and disease mechanisms .
To contextualize the utility of Phospho-HIST1H1B (S17) Antibody, it is valuable to compare it with other phospho-specific antibodies targeting related residues or proteins. This comparison highlights the specificity and unique applications of each antibody.
The Anti-Phospho-Histone H1 (T17) HIST1H1B Antibody (A06717T17) targets the threonine 17 phosphorylation site of HIST1H1B, in contrast to the serine 17 site targeted by the Phospho-HIST1H1B (S17) Antibody . This represents an important distinction, as phosphorylation at different amino acid residues can trigger distinct signaling pathways and functional outcomes. The T17 antibody demonstrates reactivity with human, monkey, and mouse samples, offering broader species coverage than the S17-specific antibody . Additionally, the T17 antibody has been validated for a wider range of applications, including immunohistochemistry and immunofluorescence, providing researchers with flexibility in experimental design .
Other phospho-specific antibodies, such as the Phospho-Src-S17 Rabbit Polyclonal Antibody (CABP0522), target different proteins but similar serine phosphorylation sites . The Src antibody specifically recognizes phosphorylation at serine 17 of the Src protein, a non-receptor tyrosine kinase involved in cell signaling pathways distinct from those associated with histone proteins . These antibodies serve different research purposes but share common methodological applications such as Western blotting and ELISA.
Similarly, the Anti-Histone H1 (Phospho-Thr17) Antibody recognizes threonine 17 phosphorylation on histone H1 but may recognize this modification across multiple H1 variants rather than specifically targeting HIST1H1B . This broader specificity may be advantageous for certain research questions but lacks the precision offered by the HIST1H1B-specific antibody.
Effective utilization of Phospho-HIST1H1B (S17) Antibody requires attention to several technical considerations to ensure reliable and reproducible results in experimental applications.
To achieve optimal results with Phospho-HIST1H1B (S17) Antibody:
Titration experiments should be performed to determine the optimal antibody concentration for each application and sample type
Appropriate blocking buffers should be used to minimize non-specific binding
Inclusion of phosphatase inhibitors in sample preparation is essential to preserve phosphorylation status
Positive and negative controls should be included to validate specificity
For Western blot applications, optimization of transfer conditions is important for efficient detection of histone proteins
Careful attention to these technical details helps ensure reliable and reproducible results when working with phospho-specific antibodies.
The study of histone phosphorylation, including HIST1H1B at serine 17, continues to expand our understanding of epigenetic regulation and its implications in health and disease. Future research utilizing Phospho-HIST1H1B (S17) Antibody may explore several promising directions:
Investigating the dynamic changes in HIST1H1B phosphorylation during different phases of the cell cycle and in response to various cellular stresses
Elucidating the enzymes (kinases and phosphatases) responsible for regulating HIST1H1B phosphorylation at serine 17
Developing targeted therapeutic approaches based on modulating HIST1H1B phosphorylation in diseases characterized by epigenetic dysregulation
Exploring the interplay between HIST1H1B phosphorylation and other histone modifications in the context of the histone code hypothesis
Utilizing advanced proteomic approaches to identify protein complexes associated with phosphorylated HIST1H1B
These research directions highlight the continuing importance of phospho-specific antibodies in advancing our understanding of chromatin biology and epigenetic regulation.
HIST1H1B is a member of the histone H1 family of proteins that plays a crucial role in chromatin structure and gene regulation. Phosphorylation of HIST1H1B at various serine and threonine residues impacts gene expression and numerous cellular processes including cell cycle progression, DNA replication, and chromatin condensation. The phosphorylation status of histone H1 proteins is dynamically regulated during different phases of the cell cycle and is frequently altered in pathological conditions such as cancer . Understanding these phosphorylation patterns provides insights into epigenetic regulatory mechanisms in both normal and disease states.
Several phosphorylation sites have been identified on HIST1H1B and related H1 variants. Mass spectrometry and phospho-specific antibody studies have confirmed multiple phosphorylation sites, including:
| Histone Variant | Phosphorylation Sites | Kinases Involved | Cell Cycle Association |
|---|---|---|---|
| H1.2 (HIST1H1C) | T154, S173 | CDKs | G2/M phase |
| H1.4 (HIST1H1E) | S27, T154, S172, S187 | CDKs | G2/M phase |
| HIST1H1B | T17, S172 | CDKs | G2/M phase |
These phosphorylation events typically occur at consensus cyclin-dependent kinase (CDK) sites with the phosphorylation occurring at (S/T)PXK sequences . The different phosphorylation patterns may contribute to distinct functional outcomes in chromatin organization.
Unlike core histone modifications (H2A, H2B, H3, H4) that often function as binding platforms for reader proteins, HIST1H1B phosphorylation primarily affects the electrostatic interactions between the histone and DNA. Phosphorylation adds negative charges to the otherwise positively charged histone, weakening its interaction with DNA and potentially loosening chromatin structure. This mechanistic difference results in broader effects on chromatin accessibility rather than the targeted recruitment of specific factors typically seen with modifications like H3K4me3 or H3K27ac. Additionally, while core histone modifications often persist through multiple cell generations as epigenetic marks, HIST1H1B phosphorylation tends to be more dynamic and cell cycle-dependent .
Designing experiments to distinguish between different phosphorylation sites requires a multi-faceted approach:
Phospho-specific antibodies: Use site-specific antibodies that recognize distinct phosphorylation sites (e.g., anti-phospho-T17 vs. anti-phospho-S172 antibodies) . Validate antibody specificity using appropriate controls.
Mass spectrometry analysis: Employ phosphopeptide enrichment strategies followed by MS/MS analysis to identify and quantify specific phosphorylation sites. This approach can simultaneously detect multiple phosphorylation events and their relative abundances .
Phosphorylation site mutants: Generate constructs with specific serine/threonine-to-alanine mutations to prevent phosphorylation at particular sites. Express these in cells and analyze their functional consequences.
Kinase inhibitors: Use specific CDK inhibitors to block phosphorylation and assess which sites are affected under different conditions. This can provide insights into the regulation of specific phosphorylation events.
Cell cycle synchronization: Since histone H1 phosphorylation is cell cycle-dependent, synchronize cells at different stages to track phosphorylation dynamics across the cell cycle using flow cytometry in combination with phospho-specific antibodies .
Essential controls for studying HIST1H1B phosphorylation include:
Phosphatase treatment controls: Treat samples with lambda phosphatase to remove phosphorylation and confirm antibody specificity.
Peptide competition assays: Pre-incubate phospho-specific antibodies with phosphorylated and non-phosphorylated peptides to validate specificity.
Phospho-site mutants: Include HIST1H1B constructs with alanine substitutions at the phosphorylation site of interest.
Cell cycle markers: Include markers for different cell cycle phases (e.g., phospho-histone H3 for mitosis) to correlate HIST1H1B phosphorylation with cell cycle progression.
Total HIST1H1B controls: Always analyze total HIST1H1B levels alongside phosphorylation status to distinguish between changes in phosphorylation and changes in protein abundance.
Cell viability controls: Include viability assays when using kinase inhibitors or other treatments that might affect cell survival, to avoid misinterpreting changes due to cell death .
To integrate HIST1H1B phosphorylation into broader epigenetic studies:
Combine with chromatin accessibility assays: Correlate HIST1H1B phosphorylation with ATAC-seq or DNase-seq data to determine how phosphorylation affects chromatin accessibility.
Link with ChIP-seq of other histone marks: Perform sequential ChIP or parallel ChIP-seq experiments to correlate HIST1H1B phosphorylation with other histone modifications.
Connect to transcriptomic data: Integrate phosphorylation analysis with RNA-seq to identify genes whose expression correlates with changes in HIST1H1B phosphorylation.
3D chromatin organization: Use Hi-C or related methods to determine if HIST1H1B phosphorylation affects higher-order chromatin organization.
Multiple time points: Analyze phosphorylation dynamics across developmental processes, cell differentiation, or disease progression to identify temporal patterns.
This multi-omics approach provides a comprehensive understanding of how HIST1H1B phosphorylation contributes to epigenetic regulation in different biological contexts.
The choice of method should be guided by the specific research question, with multiple complementary approaches often providing the most comprehensive insights.
Optimizing histone extraction for phosphorylation analysis requires special considerations:
Rapid extraction: Perform extraction quickly to prevent phosphatase activity from dephosphorylating histones.
Phosphatase inhibitors: Include phosphatase inhibitors (e.g., sodium fluoride, sodium orthovanadate, β-glycerophosphate) in all buffers.
Acid extraction: Use acid extraction methods (0.2M H₂SO₄ or 0.4N HCl) to efficiently extract histones while minimizing protein degradation.
Sample handling: Keep samples cold throughout the procedure and avoid repeated freeze-thaw cycles.
Protein extraction buffers: Use appropriate lysis buffers supplemented with protease inhibitors and phosphatase inhibitors as described in research protocols .
Quantification: Accurately quantify total protein to ensure equal loading for subsequent analyses.
Storage: Store extracted histones at -80°C with phosphatase inhibitors to maintain phosphorylation status until analysis.
These optimizations help preserve the native phosphorylation state of HIST1H1B for accurate downstream analysis.
When using phospho-specific HIST1H1B antibodies, consider these technical aspects:
Antibody validation: Verify antibody specificity using peptide competition assays, phosphatase treatment controls, and phospho-site mutants.
Cross-reactivity: Test for cross-reactivity with other H1 variants due to sequence similarity. Many commercially available antibodies may recognize phosphorylation sites conserved across multiple H1 variants .
Optimal conditions: Determine optimal antibody dilution, incubation time, and temperature for each application (WB, IF, IHC, flow cytometry).
Fixation effects: Different fixation methods may affect epitope accessibility; compare paraformaldehyde, methanol, and other fixatives.
Storage and handling: Follow manufacturer recommendations for antibody storage (typically -20°C) and avoid repeated freeze-thaw cycles .
Batch variation: Be aware of potential batch-to-batch variations; include consistent positive controls across experiments.
Species reactivity: Confirm species reactivity of the antibody. The anti-phospho-HIST1H1B antibodies have variable cross-species reactivity (human, monkey, mouse) .
Application suitability: Verify that the antibody has been validated for your specific application (ELISA, IF, IHC-P, WB) .
HIST1H1B phosphorylation appears to play significant roles in cancer progression through several mechanisms:
Correlation with tumor grade: Research has shown that phosphorylation of histone H1 at threonine 146 correlates with tumor grade in breast cancer, with higher phosphorylation levels observed in more aggressive tumors .
Cell cycle dysregulation: Cancer cells often exhibit altered cell cycle regulation, and the cell cycle-dependent phosphorylation of HIST1H1B (particularly during G2/M phase) may contribute to these abnormalities .
Response to estrogen stimulation: In breast cancer models, estradiol stimulation increases histone H1 phosphorylation, suggesting a link between hormone signaling and epigenetic regulation through histone phosphorylation .
Altered chromatin accessibility: Increased phosphorylation of histone H1 may lead to chromatin decompaction, potentially enabling aberrant gene expression patterns that promote cancer progression.
Therapeutic response marker: Changes in histone H1 phosphorylation in response to PI3K inhibitor LY294002 indicate its potential utility as a biomarker for monitoring therapeutic response in cancer treatment .
Invasiveness indicator: Studies in bladder carcinogenesis have demonstrated that increased global phosphorylation of histone H1 is associated with cancer invasiveness, suggesting its role in promoting metastatic potential .
These findings highlight the potential of HIST1H1B phosphorylation as both a biomarker and a mechanistic contributor to cancer development and progression.
When faced with contradictory phosphorylation data, consider these analytical approaches:
Cell type-specific mechanisms: Different cell types may have distinct regulatory pathways controlling HIST1H1B phosphorylation. Compare the expression levels of relevant kinases (CDKs) and phosphatases across cell types.
Methodology differences: Evaluate whether methodological differences might explain contradictory results. Different antibodies, extraction methods, or detection techniques can yield varying results.
Context-dependent regulation: Consider that phosphorylation may be regulated differently depending on cellular context (proliferation status, differentiation state, stress conditions).
Multiple phosphorylation sites: Assess whether the contradictions involve different phosphorylation sites. Phosphorylation at one site may influence modification at other sites through cross-talk mechanisms.
Temporal dynamics: Examine the temporal aspects of phosphorylation. Contradictory data might reflect different time points in a dynamic process.
Signaling pathway integration: Map the phosphorylation data to known signaling pathways to identify potential points of convergence or divergence that might explain the contradictions.
Biological variability: Consider the inherent biological variability in phosphorylation levels, particularly in primary samples compared to cell lines.
Systematically analyzing these factors can help reconcile apparently contradictory data and may even reveal novel regulatory mechanisms.
Emerging approaches for studying HIST1H1B phosphorylation dynamics include:
Phospho-specific antibody engineering: Advanced antibody design techniques are enabling the development of highly specific antibodies that can distinguish between closely related phosphorylation sites .
Live-cell imaging of phosphorylation: Genetically encoded biosensors that report on the phosphorylation status of specific sites in real-time are being developed for histone modifications.
Single-cell phosphoproteomics: Emerging technologies that allow phosphorylation analysis at the single-cell level can reveal heterogeneity in HIST1H1B phosphorylation within populations.
Targeted mass spectrometry: Multiple reaction monitoring (MRM) assays specifically developed for HIST1H1B phosphorylation sites enable more sensitive and quantitative detection than traditional approaches .
CRISPR-based approaches: Precise genome editing to introduce mutations at endogenous loci or to tag HIST1H1B allows for physiological studies of phosphorylation dynamics.
Proximity labeling techniques: Methods like BioID or APEX combined with phospho-enrichment can identify proteins that interact specifically with phosphorylated HIST1H1B.
Computational modeling: Integration of phosphorylation data with structural information to predict how specific phosphorylation events affect histone-DNA interactions and chromatin structure.
These innovative approaches are expanding our ability to study the complex dynamics of HIST1H1B phosphorylation in various biological contexts.
Common artifacts and their solutions include:
Dephosphorylation during sample preparation:
Artifact: Loss of phosphorylation signal due to phosphatase activity
Solution: Use phosphatase inhibitors, maintain cold temperatures, and minimize preparation time
Cell cycle heterogeneity:
Non-specific antibody binding:
Artifact: False positive signals from cross-reactivity
Solution: Validate antibodies with phosphatase treatment and peptide competition assays
Cell death-induced changes:
Fixation artifacts:
Artifact: Loss of epitope recognition due to over-fixation
Solution: Optimize fixation protocols and compare multiple fixation methods
Extraction bias:
Artifact: Preferential extraction of certain histone variants or modification states
Solution: Compare multiple extraction methods and validate with spike-in controls
Batch effects in antibody performance:
Artifact: Variability between experiments due to antibody lot differences
Solution: Use consistent antibody lots and include standard controls across experiments
Recognizing and addressing these potential artifacts is crucial for generating reliable and reproducible phosphorylation data.
To correlate HIST1H1B phosphorylation with functional outcomes:
Phosphomimetic and phospho-deficient mutants: Generate S/T to D/E (phosphomimetic) or S/T to A (phospho-deficient) mutants and assess their impact on cellular processes.
ChIP-seq analysis: Perform ChIP-seq with phospho-specific antibodies to identify genomic regions associated with phosphorylated HIST1H1B and correlate with gene expression.
Kinase inhibitor studies: Use specific CDK inhibitors to modulate phosphorylation and observe resulting phenotypic changes, as demonstrated with LY294002 treatment .
Cell cycle analysis: Correlate phosphorylation with cell cycle progression using synchronized cells and flow cytometry to establish functional relationships .
Chromatin accessibility assays: Combine phosphorylation analysis with ATAC-seq or MNase sensitivity assays to determine how phosphorylation affects chromatin compaction.
Protein interaction studies: Identify proteins that differentially interact with phosphorylated versus non-phosphorylated HIST1H1B using immunoprecipitation followed by mass spectrometry.
Functional genomics screens: Use CRISPR/Cas9 to modulate HIST1H1B phosphorylation and screen for phenotypic outcomes.
These approaches can establish causal relationships between specific phosphorylation events and their functional consequences in cellular processes.
Appropriate statistical approaches include:
For Western blot densitometry:
Normalize phospho-signal to total HIST1H1B
Use ANOVA with post-hoc tests for multiple conditions
Apply non-parametric tests (Kruskal-Wallis) if normality assumptions are violated
For mass spectrometry quantification:
Consider specialized software (MaxQuant, Skyline) for phosphopeptide quantification
Use linear mixed-effects models to account for technical and biological variability
Apply multiple testing corrections (Benjamini-Hochberg) when analyzing multiple phosphorylation sites
For flow cytometry data:
For correlation with clinical parameters:
Use Spearman's rank correlation for ordinal variables like tumor grade
Apply Kaplan-Meier survival analysis to correlate phosphorylation with patient outcomes
Consider multivariate Cox regression to adjust for confounding factors
For time-course experiments:
Use repeated measures ANOVA or mixed models
Consider time-series analysis methods for identifying temporal patterns