HIST1H1B (Ab-172) Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Overview

The HIST1H1B (Ab-172) Antibody is a polyclonal antibody raised in rabbits against a synthesized peptide corresponding to residues 164–176 of human Histone H1.5 . It specifically detects the HIST1H1B protein (UniProt ID: P16401), a somatic linker histone variant involved in nucleosome spacing and chromatin compaction .

Biological Role of HIST1H1B

HIST1H1B (H1.5) is a replication-dependent linker histone that:

  • Facilitates higher-order chromatin structure by binding linker DNA between nucleosomes .

  • Regulates gene transcription via chromatin remodeling and DNA methylation .

  • Is implicated in cancer progression, particularly basal-like breast cancer (BLBC), where its overexpression correlates with tumor size, metastasis, and poor survival .

Cancer Biology

  • BLBC Progression: HIST1H1B is upregulated in BLBC due to copy number amplification and promoter hypomethylation. The antibody has been used to demonstrate HIST1H1B’s role in enhancing tumorigenicity through CSF2 (colony-stimulating factor 2) modulation .

    • Key Finding: Knockdown of HIST1H1B suppresses tumor growth in vitro and in vivo, while overexpression promotes metastasis .

Epigenetic Studies

  • Chromatin Mapping: Chromatin immunoprecipitation (ChIP) assays using HIST1H1B antibodies revealed genome-wide distribution patterns of H1 variants in breast cancer cells, highlighting H1.5’s enrichment at regulatory regions .

Immunology

  • Dendritic Cell (DC) Modulation: Anti-histone H1 antibodies (including HIST1H1B-targeting variants) downregulate DC maturation markers (e.g., CD80/CD86) and inflammatory cytokines, suggesting a role in immune tolerance .

Clinical Relevance

  • Prognostic Marker: HIST1H1B overexpression in BLBC is linked to larger tumor size, higher histological grade, and reduced survival .

  • Therapeutic Target: Targeting HIST1H1B-mediated CSF2 signaling may offer novel strategies for BLBC treatment .

Comparative Insights

FeatureHIST1H1B (Ab-172) vs. Other H1 Antibodies
SpecificityTargets residues 164–176 of H1.5; distinct from Ab-17 (targets Ser-17)
ApplicationsLimited to ELISA/IHC; lacks ChIP validation seen in Acetyl-HIST1H1B (K16) variants
Clinical UtilityStronger association with BLBC than other H1 variants

Future Directions

  • Investigate HIST1H1B’s role in other cancers (e.g., colorectal, leiomyosarcoma) .

  • Develop conjugated variants (e.g., HRP, FITC) for multiplex assays .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery timelines.
Synonyms
Histone H1.5 (Histone H1a) (Histone H1b) (Histone H1s-3), HIST1H1B, H1F5
Target Names
HIST1H1B
Uniprot No.

Target Background

Function
Histone H1 protein binds to linker DNA between nucleosomes, forming the macromolecular structure known as the chromatin fiber. Histones H1 are crucial for condensing nucleosome chains into higher-order structured fibers. They also function as regulators of individual gene transcription through chromatin remodeling, nucleosome spacing, and DNA methylation.
Gene References Into Functions
  1. Research suggests that normal ovarian tissues exhibit strong expression of histone H1.5, while ovarian granulosa cell tumors (GCTs) display weak expression of this protein. Conversely, PLZF protein expression did not show significant differences between the two study groups. PMID: 25023763
  2. Statistically significant differences in staining patterns were observed for histone H1.5, effectively distinguishing leiomyosarcomas from leiomyomas. PMID: 24784718
  3. Promyelocytic leukemia zinc finger and histone H1.5 exhibit differential staining patterns in low- and high-grade pulmonary neuroendocrine tumors. PMID: 23416030
  4. FoxP3 interacts with H1.5, altering its binding to target genes, thereby modulating their expression and programming Treg function. PMID: 21654845
  5. The mode of chromatin fiber compaction varies depending on both the salt environment and the presence of linker histone H1. PMID: 16185066
  6. Phosphorylation of human H1 variants occurs nonrandomly during both interphase and mitosis, with distinct serine- or threonine-specific kinases involved in different cell cycle phases. PMID: 16377619
  7. Phosphorylation at threonine 10 appears in prometaphase and disappears in telophase. This hyperphosphorylated form of H1.5 is primarily chromatin-bound in metaphase, when chromatin condensation reaches its maximum. PMID: 19136008

Show More

Hide All

Database Links

HGNC: 4719

OMIM: 142711

KEGG: hsa:3009

STRING: 9606.ENSP00000330074

UniGene: Hs.131956

Protein Families
Histone H1/H5 family
Subcellular Location
Nucleus. Chromosome. Note=According to PubMed:15911621 more commonly found in heterochromatin. According to PubMed:10997781 associates with actively transcribed chromatin and not heterochromatin.
Tissue Specificity
Ubiquitous. Expressed in the majority of the cell lines tested and in testis.

Q&A

What is HIST1H1B and how does it function in chromatin regulation?

HIST1H1B is a variant of the linker histone H1 family, which plays crucial roles in maintaining higher-order chromatin structure and regulating gene expression . As a linker histone, HIST1H1B binds to nucleosomes and the linker DNA between them, facilitating chromatin compaction and influencing transcriptional activity. H1 variants like HIST1H1B show differential expression patterns across cell types, with some variants being upregulated in terminally differentiating cells while showing decreased expression in stem cells . HIST1H1B specifically belongs to the somatic H1 variants that exhibit tissue-specific and developmental stage-specific expression patterns, making it an important epigenetic regulator in distinct cellular contexts.

What experimental validation is necessary before using HIST1H1B (Ab-172) Antibody?

Before employing HIST1H1B (Ab-172) Antibody in research applications, comprehensive validation is essential. Researchers should perform Western blot analysis to confirm that the antibody detects a single band of appropriate molecular weight (~22-23 kDa for H1 histones). Importantly, validation should include testing across multiple cell types where HIST1H1B is differentially expressed. Given the challenges with H1 variant-specific antibodies, researchers should also conduct specificity tests using:

  • Immunoprecipitation followed by mass spectrometry to confirm target identity

  • siRNA or CRISPR knockdown of HIST1H1B to verify signal reduction

  • Peptide competition assays to evaluate binding specificity

  • Cross-reactivity assessment against other H1 variants using recombinant proteins

The lack of reliable variant-specific antibodies is a significant challenge in H1 histone research , necessitating rigorous validation before experimental use.

What are optimal sample preparation protocols for HIST1H1B detection?

For optimal HIST1H1B detection using Ab-172 antibody, sample preparation should account for the unique properties of histone proteins. The following methodological approach is recommended:

  • Nuclear extraction: Isolate nuclei using hypotonic buffer followed by acid extraction (0.2N H₂SO₄ or 0.4N HCl) to selectively extract histones

  • Protein preservation: Include deacetylase inhibitors (e.g., sodium butyrate), phosphatase inhibitors, and protease inhibitors during extraction

  • Post-translational modification preservation: Add 5-10mM sodium butyrate to all buffers to maintain acetylation status

  • Storage conditions: Store extracted histones at -80°C in small aliquots to avoid freeze-thaw cycles

This protocol considers the extensive post-translational modifications found on H1 histone tails, which can significantly affect antibody recognition . The amino and carboxy terminal tails of histone H1 variants are among the most abundantly post-translationally modified sequences in the cell, with multiple simultaneous PTMs regularly identified .

What are the recommended applications for HIST1H1B (Ab-172) Antibody?

The HIST1H1B (Ab-172) Antibody can be employed in various experimental techniques, each requiring specific optimization:

  • Western blotting: Use 1:500-1:2000 dilution with 5% BSA in TBST; acid-extracted histones provide cleaner results than whole-cell lysates

  • Immunofluorescence: Fixation with 4% paraformaldehyde followed by permeabilization with 0.5% Triton X-100; nuclear localization should be evident

  • ChIP/ChIP-seq: Optimize crosslinking time (typically 10-15 minutes with 1% formaldehyde); include controls to account for potential discrepancies between antibody results and actual chromatin binding dynamics

  • Flow cytometry: Fix cells with 70% ethanol and permeabilize with 0.25% Triton X-100

When conducting ChIP-seq experiments, be aware of potential discrepancies that have been found between data acquired using antibodies specific to endogenous H1 variants versus tagged variants in the same cells .

How can post-translational modifications affect HIST1H1B antibody specificity?

Post-translational modifications (PTMs) on HIST1H1B can significantly impact antibody recognition and experimental outcomes. The terminal domains of H1 histones exhibit extensive PTMs, including phosphorylation, methylation, acetylation, and ubiquitination. The comprehensive PTM landscape of histone H1.4 (shown in Figure 3 of reference) illustrates this complexity .

For HIST1H1B (Ab-172) Antibody, researchers should consider:

  • Epitope location: If the antibody targets regions susceptible to PTMs, modification status will alter recognition

  • Phosphorylation interference: Cell cycle-dependent phosphorylation can mask epitopes, particularly during mitosis

  • Combinatorial PTM effects: Multiple nearby modifications can create "PTM switches" that affect antibody binding

To address these challenges, researchers should:

  • Determine the exact epitope recognized by Ab-172

  • Test antibody recognition across different cell cycle stages

  • Consider using phosphatase treatment on parallel samples to assess phosphorylation effects

  • Validate findings with alternative methods like mass spectrometry

As noted in the literature, antibodies generated toward the tail domains of H1 could result in low specificity based on the PTM combinations present on the H1 molecule and the immunogen used for antibody generation .

What strategies can overcome challenges in ChIP-seq experiments with HIST1H1B antibodies?

ChIP-seq experiments with HIST1H1B antibodies present significant technical challenges related to specificity, cross-reactivity, and chromatin dynamics. To address these issues, implement the following methodological strategies:

  • Spike-in normalization: Include exogenous chromatin (e.g., Drosophila) with specific antibodies for normalization

  • Sequential ChIP (Re-ChIP): Perform sequential immunoprecipitations with different antibodies to increase specificity

  • Validation with orthogonal approaches: Compare ChIP-seq results with:

    • CUT&RUN or CUT&Tag for improved signal-to-noise ratio

    • Tagged HIST1H1B expression with subsequent ChIP using tag-specific antibodies (with awareness of tag interference)

    • Mass spectrometry of immunoprecipitated material to confirm target identity

  • Computational analysis refinements:

    • Implement stringent peak calling parameters

    • Perform differential binding analysis between cell types

    • Correlate binding patterns with gene expression data

Given that discrepancies have been found in ChIP-seq data acquired using antibodies specific to endogenous H1.2 versus tagged H1.2 in the same cells, similar concerns may apply to HIST1H1B detection .

How can researchers distinguish between different H1 variants when using antibodies?

Distinguishing between HIST1H1B and other H1 variants presents a significant challenge due to high sequence homology. The following comprehensive approach can help researchers achieve variant-specific detection:

  • Comparative proteomic analysis: Utilize the differential peptide characteristics of H1 variants as shown in the table below, which demonstrates tryptic peptide patterns of H1.4 compared to H4 and BSA:

Histone H1.4 RegionPositionMass (Da)Tryptic peptide sequenceRelative hydrophobicity
NTD1–171608.7817MSETAPAAPAAPAPAEK17.96
Globular35–461197.6605ASGPPVSELITK23.86
Globular65–751106.5607ALAAAGYDVEK17.53
CTD111–119857.4606AASGEAKPK4.9
Globular55–63844.5018SGVSLAALK20.51

This peptide mapping approach allows for precise identification of variant-specific regions that can be targeted by antibodies or used for validation .

  • Complementary techniques:

    • Mass spectrometry for variant identification based on unique peptides

    • Expression pattern analysis across different cell types

    • Knockout/knockdown validation studies to confirm specificity

  • Epitope mapping: Determine the exact epitope recognized by HIST1H1B (Ab-172) Antibody and compare with sequence alignments of all H1 variants to predict potential cross-reactivity.

What experimental controls are essential when working with HIST1H1B antibodies?

When designing experiments with HIST1H1B (Ab-172) Antibody, implement these critical controls to ensure data reliability:

  • Specificity controls:

    • Include HIST1H1B knockout/knockdown samples

    • Use competing peptides corresponding to the immunogen

    • Test in cell types with documented differential expression of HIST1H1B

  • Technical controls:

    • Include IgG isotype control for background binding assessment

    • Use multiple antibody dilutions to establish optimal signal-to-noise ratios

    • Process identical samples with alternative detection methods

  • Biological condition controls:

    • Compare terminally differentiated cells vs. stem cells, as H1 variants show differential expression in these contexts

    • Assess cell cycle phases separately, as H1 binding and PTMs are cell cycle-dependent

    • Include samples treated with epigenetic modifiers (e.g., HDAC inhibitors)

  • Alternative methods validation:

    • Validate key findings with mass spectrometry

    • Compare results with tagged HIST1H1B expression systems, while acknowledging potential tag interference

How can researchers address the limitations of HIST1H1B antibodies in functional studies?

To overcome inherent limitations of antibody-based detection of HIST1H1B, researchers should implement complementary approaches for functional studies:

  • CRISPR-based genomic tagging: Create endogenous tags at the HIST1H1B locus, while recognizing potential functional interference :

    • Small epitope tags (HA, FLAG) may minimize functional disruption

    • Position tags strategically to avoid disrupting functional domains

    • Validate tagged line function against wild-type controls

  • Orthogonal protein interaction studies:

    • Proximity labeling techniques (BioID, APEX) to identify interactors

    • Crosslinking mass spectrometry to map structural interactions

    • Live-cell imaging with fluorescent protein fusions for dynamic studies

  • Genomic approaches:

    • CUT&RUN or CUT&Tag for chromatin association studies

    • Hi-C analyses to assess chromatin architectural changes

    • RNA-seq following HIST1H1B manipulation to assess transcriptional impact

  • Biochemical assays:

    • In vitro nucleosome binding assays with recombinant proteins

    • Micrococcal nuclease digestion patterns to assess chromatin compaction

These approaches align with findings that MS has become widely used to analyze histone H1 variants through the ability to bypass the limitations of immunological reagents .

What are common causes of inconsistent results with HIST1H1B (Ab-172) Antibody?

Inconsistent results when using HIST1H1B (Ab-172) Antibody can stem from several technical and biological factors:

  • Epitope masking by PTMs: Post-translational modifications can block antibody binding sites. The amino and carboxy terminal tails of histone H1 variants are among the most abundantly post-translationally modified sequences in the cell , leading to variable epitope accessibility.

  • Cell cycle variation: H1 histones undergo cell cycle-dependent modifications and localization changes. For example, phosphorylation of H1.4 at Thr146 has been identified on condensed mitotic chromatin by immunofluorescence , potentially affecting antibody recognition.

  • Extraction method limitations: Different extraction protocols yield different populations of H1 histones based on their chromatin binding properties.

  • Cross-reactivity with other H1 variants: High sequence homology between H1 variants can lead to non-specific detection.

To address these issues:

  • Synchronize cells when possible

  • Document exact extraction and fixation protocols

  • Validate with orthogonal methods

  • Test antibody recognition across different extraction conditions

How should researchers interpret disparate results between antibody-based detection and other methods?

When faced with discrepancies between HIST1H1B antibody detection and alternative approaches, researchers should implement this analytical framework:

  • Systematic evaluation of potential causes:

    • Epitope accessibility issues due to chromatin context or PTMs

    • Technical limitations of each method

    • Biological variations in different experimental conditions

  • Reconciliation approach:

    • Map the exact epitope recognized by the antibody

    • Document PTM status at or near the epitope region

    • Consider structural conformations that might affect accessibility

    • Assess potential interference from interacting proteins

  • Integrated data analysis:

    • Weight evidence based on methodological strengths and limitations

    • Develop models that accommodate seemingly contradictory results

    • Design experiments that directly address the source of discrepancies

This approach acknowledges that discrepancies have been found in ChIP-seq data acquired using antibodies specific to endogenous H1 variants versus tagged variants in the same cells , suggesting inherent methodological challenges that require careful interpretation.

How can emerging technologies improve HIST1H1B research beyond antibody limitations?

Several cutting-edge technologies show promise for advancing HIST1H1B research while addressing the limitations of antibody-based approaches:

  • Degradation-based protein targeting:

    • PROTAC (Proteolysis Targeting Chimeras) for selective variant degradation

    • Auxin-inducible degron systems for temporal control of HIST1H1B levels

    • dTAG systems for rapid and selective protein depletion

  • Advanced microscopy techniques:

    • Single-molecule tracking to study HIST1H1B dynamics in living cells

    • Super-resolution microscopy for spatial distribution analysis

    • FRET-based sensors to detect HIST1H1B interactions and conformational changes

  • Next-generation chromatin mapping:

    • CUT&Tag for improved signal-to-noise ratio in chromatin localization

    • Liquid chromatin Hi-C to assess HIST1H1B's role in 3D genome organization

    • Single-cell ChIP-seq to capture heterogeneity in HIST1H1B distribution

  • Targeted mass spectrometry:

    • Parallel reaction monitoring (PRM) for quantitative analysis of specific HIST1H1B peptides

    • Crosslinking mass spectrometry for structural studies

    • Top-down proteomics to analyze intact HIST1H1B and its modifications

These approaches address the limitations noted in current literature, where MS has become widely used to analyze histone H1 variants through the ability to bypass the limitations of immunological reagents .

What important biological questions about HIST1H1B remain unaddressed due to technical limitations?

Despite advances in histone research, several critical questions about HIST1H1B remain challenging to address due to technical limitations:

  • Variant-specific functions: Determining the unique functional roles of HIST1H1B versus other H1 variants remains difficult due to the lack of reliable variant-specific antibodies .

  • Dynamic regulation: Understanding how HIST1H1B binding and dissociation is regulated in real-time during processes like transcription, replication, and repair.

  • Local chromatin environment influence: How different chromatin contexts affect HIST1H1B recruitment, retention, and function.

  • Interactome specificity: Identifying proteins that preferentially interact with HIST1H1B compared to other H1 variants.

  • PTM crosstalk: Elucidating how various post-translational modifications on HIST1H1B cooperate or antagonize each other, given that multiple numbers of simultaneous PTMs on histone H1 are regularly identified .

Researchers addressing these questions should consider:

  • Developing new technological approaches beyond antibody-based detection

  • Combining multiple orthogonal methods for validation

  • Establishing system-specific controls that account for the biological context

  • Implementing computational models that integrate diverse datasets

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.