The HIST1H1B (Ab-154) Antibody is a polyclonal rabbit-derived immunoglobulin (IgG) designed to specifically target the Thr154 residue of human histone H1.5 (HIST1H1B), a linker histone critical for chromatin compaction and gene regulation. This antibody is optimized for applications such as ELISA, immunohistochemistry (IHC), and potentially Western blotting (WB) or immunofluorescence (IF), depending on the supplier. Its specificity for Thr154 enables precise detection of HIST1H1B in human tissues, making it a valuable tool in epigenetics, cancer biology, and chromatin dynamics research .
HIST1H1B (UniProt: P16401) belongs to the histone H1 family and binds linker DNA between nucleosomes, stabilizing chromatin architecture. Key features include:
Biological Role: Regulates chromatin compaction, nucleosome spacing, and transcriptional activity through interactions with chromatin-modifying enzymes .
HIST1H1B is implicated in diseases such as basal-like breast cancer (BLBC), where its overexpression correlates with tumorigenicity .
HIST1H1B (Ab-154) is used to investigate chromatin remodeling and nucleosome dynamics. For example:
Chromatin Compaction: Detects HIST1H1B’s role in organizing higher-order chromatin structures .
Gene Regulation: Reveals interactions with histone deacetylases and DNA methyltransferases .
A study on basal-like breast cancer (BLBC) demonstrated that HIST1H1B overexpression enhances colony formation in vitro and tumor growth in vivo, suggesting its potential as a prognostic marker . While this study did not explicitly use the Ab-154 antibody, such tools are critical for validating HIST1H1B’s role in oncogenesis.
The antibody is validated for IHC on paraffin-embedded tissues (e.g., human testis), enabling spatial analysis of HIST1H1B localization .
Histone H1 protein binds to linker DNA between nucleosomes, forming the chromatin fiber. These histones are essential for the condensation of nucleosome chains into higher-order structures. Additionally, Histone H1 plays a regulatory role in gene transcription through chromatin remodeling, nucleosome spacing, and DNA methylation.
HIST1H1B, also known as Histone H1.5, is a linker histone protein that binds to DNA between nucleosomes, contributing to chromatin fiber formation and structural organization. Phosphorylation at Threonine 154 (pThr154) represents one of several post-translational modifications that can alter HIST1H1B's interactions with DNA and its regulatory functions . This specific modification impacts chromatin compaction and accessibility, potentially affecting gene expression patterns in various cellular contexts. Phosphorylation of linker histones generally correlates with looser chromatin structure and increased transcriptional activity in the affected regions.
The HIST1H1B antibody targeting phosphorylated Threonine 154 has been validated for multiple research applications, including:
ELISA (Enzyme-Linked Immunosorbent Assay)
Western Blotting (WB)
Immunofluorescence (IF)
These techniques allow researchers to detect, quantify, and localize phosphorylated HIST1H1B in various experimental contexts, from protein expression analysis to chromatin-association studies. The antibody is particularly valuable for investigating phosphorylation-dependent functions of HIST1H1B in human cells and tissues.
Based on empirical validation, the following dilution ranges are recommended for optimal results when using the HIST1H1B (pThr154) antibody:
| Application | Recommended Dilution Range |
|---|---|
| Western Blotting | 1:200-1:2000 |
| Immunofluorescence | 1:50-1:200 |
| ELISA | Typically 1:2000-1:10000 (based on similar antibodies) |
| ChIP | Application-specific optimization required |
These dilutions should be further optimized based on your specific experimental conditions, sample type, and detection method.
For rigorous experimental design with the HIST1H1B (pThr154) antibody, incorporate these essential controls:
Positive control: Use cell lines or tissues known to express phosphorylated HIST1H1B at Thr154, such as proliferating breast cancer cell lines (e.g., MDA-468, BT20) .
Negative control:
Antibody controls:
Isotype control (rabbit IgG)
Secondary antibody-only control
Pre-immune serum (for polyclonal antibodies)
Phosphorylation induction/inhibition: Compare samples treated with kinase activators versus phosphatase inhibitors to demonstrate specificity for the phosphorylated form.
These controls help validate antibody specificity and prevent misinterpretation of experimental results.
Optimizing ChIP for HIST1H1B (pThr154) requires several key considerations:
Crosslinking conditions: Since HIST1H1B is a linker histone with dynamic chromatin association, optimize formaldehyde concentration (0.5-1%) and incubation time (5-15 minutes) for efficient but reversible crosslinking.
Chromatin fragmentation: Aim for fragments of 200-500bp through careful sonication calibration. Histone binding may protect certain DNA regions, requiring optimization of sonication parameters.
Antibody incubation: The polyclonal nature of the antibody suggests overnight incubation at 4°C with 2-5μg antibody per ChIP reaction .
Washing stringency: Use increasingly stringent wash buffers to minimize non-specific binding while preserving specific interactions.
Elution and reversal: Carefully optimize elution conditions and reversal of crosslinks to maximize DNA recovery without introducing artifacts.
Controls: Include input chromatin (pre-immunoprecipitation sample) and IgG control in every experiment.
Specifically for HIST1H1B studies, consider analyzing promoter regions of genes known to be regulated by HIST1H1B, such as CSF2, which has been demonstrated to be directly bound by HIST1H1B .
To effectively study HIST1H1B phosphorylation at Thr154 and its impact on gene expression in cancer:
Establish cellular models:
Functional assays:
Molecular profiling:
RNA-seq to identify differentially expressed genes between models
ChIP-seq using the pThr154 antibody to map genomic binding sites
Integration with existing datasets on histone modifications and chromatin structure
Mechanistic investigation:
This approach can reveal how HIST1H1B phosphorylation affects oncogenic processes, particularly in breast cancer where HIST1H1B overexpression has been linked to basal-like subtypes and poor prognosis .
Investigating the crosstalk between HIST1H1B phosphorylation at Thr154 and other epigenetic modifications requires multi-layered approaches:
Sequential ChIP (Re-ChIP): Perform initial ChIP with anti-pThr154 antibody followed by a second immunoprecipitation with antibodies against other histone modifications to identify co-occurrence patterns .
Mass spectrometry-based approaches:
Analyze post-translational modification (PTM) patterns on histones in different cellular contexts
Identify proteins that differentially associate with phosphorylated versus non-phosphorylated HIST1H1B
Proximity ligation assays: Visualize and quantify spatial relationships between pThr154 HIST1H1B and other modified histones in situ.
Integrative genomics:
Compare ChIP-seq data for pThr154 HIST1H1B with datasets for other histone marks
Analyze correlation or anti-correlation patterns at specific genomic regions
Functional manipulation:
Use histone modification enzyme inhibitors to assess their impact on HIST1H1B phosphorylation
Employ CRISPR/Cas9-based approaches to modify specific histone-modifying enzymes
These methods can illuminate how HIST1H1B phosphorylation coordinates with broader epigenetic landscapes to regulate gene expression and chromatin structure.
Detecting phosphorylated HIST1H1B by Western blot presents several technical challenges:
Low abundance of phosphorylated form:
Enrich for nuclear proteins in your extraction protocol
Consider phosphoprotein enrichment methods
Load adequate protein amounts (50-100μg per lane)
Use high-sensitivity detection systems (ECL Plus or fluorescent secondary antibodies)
Cross-reactivity issues:
Phosphatase activity during sample preparation:
Include phosphatase inhibitors (e.g., sodium fluoride, sodium orthovanadate, β-glycerophosphate)
Maintain samples at 4°C during processing
Use acidic extraction buffers when appropriate
Detection of appropriate molecular weight:
Expect HIST1H1B at approximately 22-23 kDa
Phosphorylation may cause slight mobility shifts
Consider using gradient gels for better resolution
Stripping and reprobing challenges:
Mild stripping conditions may be insufficient to remove phospho-specific antibodies
Consider running parallel blots rather than stripping when comparing phosphorylated and total HIST1H1B
Methodical optimization of these parameters will improve detection specificity and sensitivity.
Discriminating between phosphorylated HIST1H1B and other H1 variants requires careful experimental design:
Sequence analysis and epitope mapping:
Analyze the amino acid sequence surrounding Thr154 in HIST1H1B
Compare with corresponding regions in other H1 variants
Determine if the epitope recognized by the antibody is unique to HIST1H1B
Validation experiments:
Perform immunoblotting against recombinant H1 variants
Use variant-specific siRNA knockdowns followed by immunoblotting
Employ variant-specific blocking peptides in competition assays
Mass spectrometry confirmation:
Immunoprecipitate with the phospho-specific antibody
Analyze precipitated proteins by mass spectrometry
Identify unique peptides that confirm variant identity
Combined immunoprecipitation approaches:
First precipitate with variant-specific antibodies
Then probe with phospho-specific antibodies
Or perform the reverse sequence
Genomic targeting analysis:
Compare ChIP-seq profiles using variant-specific versus phospho-specific antibodies
Identify overlapping and distinct binding patterns
These approaches help ensure that observed signals truly represent phosphorylated HIST1H1B rather than other phosphorylated H1 variants.
The HIST1H1B (pThr154) antibody offers valuable applications in breast cancer research:
Subtype classification:
Prognostic stratification:
Therapeutic target identification:
Mechanistic studies:
Investigate how phosphorylation at Thr154 affects HIST1H1B function in tumor progression
Explore kinase inhibitors that might reduce HIST1H1B phosphorylation
Biomarker development:
Validate pThr154 HIST1H1B as a biomarker using tissue microarrays
Correlate with clinical outcomes in prospective studies
These applications could advance both understanding of breast cancer biology and development of new diagnostic or therapeutic approaches.
Analyzing HIST1H1B phosphorylation in clinical samples requires specialized protocols:
Tissue preservation and processing:
Rapid fixation is critical to preserve phosphorylation status
Consider using phosphatase inhibitor-containing fixatives
For frozen samples, maintain strict temperature control throughout processing
Immunohistochemistry optimization:
Multiplexed immunofluorescence:
Combine pThr154 HIST1H1B detection with other markers (e.g., CSF2, proliferation markers)
Use spectral unmixing to resolve signal overlap
Quantify using digital pathology approaches
Protein extraction from FFPE samples:
Specialized protocols using Qproteome FFPE Tissue Kit or similar
Include phosphatase inhibitors throughout
Validate extraction efficiency with known phospho-proteins
Alternative approaches:
Proximity ligation assay for increased specificity
Laser capture microdissection to isolate specific cell populations
Reverse phase protein arrays for high-throughput screening
Normalization strategies:
Always compare to total HIST1H1B levels
Use housekeeping proteins stable in cancer tissues
Consider ratio-based reporting of phosphorylation status
These methods help overcome the challenges of working with clinical samples while maintaining detection specificity and sensitivity.
Integrating single-cell technologies with HIST1H1B phosphorylation analysis offers exciting research opportunities:
Single-cell CUT&Tag/CUT&RUN:
Adapt these techniques using the pThr154 antibody
Map phosphorylated HIST1H1B binding at single-cell resolution
Correlate with cell states or differentiation stages
Single-cell multi-omics:
Combine pThr154 HIST1H1B ChIP with scRNA-seq (CITE-seq approaches)
Integrate with single-cell ATAC-seq for chromatin accessibility correlation
Develop computational approaches to infer phosphorylation impact on gene regulation
In situ approaches:
Adapt proximity ligation assays for single-cell analysis
Develop multiplexed immunofluorescence protocols
Consider emerging in situ sequencing technologies
Micro-fluidic applications:
Develop microfluidic immunocapture methods for circulating tumor cells
Analyze pThr154 HIST1H1B in minimal sample volumes
Enable longitudinal monitoring in clinical contexts
Live-cell imaging:
Generate phospho-specific intrabodies or nanobodies
Monitor dynamics of phosphorylation in living cells
Correlate with chromatin dynamics and transcriptional output
These approaches would provide unprecedented insights into the heterogeneity and dynamics of HIST1H1B phosphorylation in complex biological systems.
While HIST1H1B has been well-studied in breast cancer , several compelling research directions exist for investigating its phosphorylation in other contexts:
Other cancer types:
Is HIST1H1B phosphorylation at Thr154 altered in additional aggressive cancers?
Does it correlate with similar prognostic patterns across malignancies?
Are there cancer-specific phosphorylation patterns on HIST1H1B?
Neurodegenerative diseases:
How does HIST1H1B phosphorylation change during neural aging?
Is there altered HIST1H1B phosphorylation in Alzheimer's or Parkinson's disease?
Could it represent an epigenetic component of neurodegeneration?
Inflammatory conditions:
Development and differentiation:
How does Thr154 phosphorylation change during cellular differentiation?
Does it play a role in maintaining stem cell identity or directing lineage commitment?
Therapeutic resistance mechanisms:
Does HIST1H1B phosphorylation status predict response to epigenetic therapies?
Could targeting the kinases responsible for Thr154 phosphorylation enhance treatment efficacy?
These questions represent promising avenues for expanding our understanding of HIST1H1B phosphorylation beyond its established role in breast cancer biology.