HIST1H1D (Ab-16) Antibody

Shipped with Ice Packs
In Stock

Description

Biological Context of HIST1H1D

HIST1H1D encodes histone H1.3, which:

  • Compacts nucleosome arrays into 30-nm chromatin fibers

  • Regulates DNA methylation and nucleosome spacing

  • Exhibits cell cycle-dependent phosphorylation at Thr146 and Ser35

  • Is encoded by an intronless gene on chromosome 6p21.3

Mouse knockout studies reveal that triple deletion of H1d/c/e causes embryonic lethality due to chromatin decompaction, emphasizing its structural role .

Epigenetic Regulation Studies

The antibody enables:

  • Mapping H1.3 distribution during mitosis using IF

  • Investigating crosstalk between H1 PTMs and DNA methylation

  • Identifying Aurora B kinase-mediated phosphorylation at Ser27

Disease Associations

HIST1H1D dysregulation correlates with:

  • Impaired muscle differentiation via MSX1-MyoD axis

  • Altered gene expression in triple-negative breast cancer

  • Neurodegenerative disease progression through chromatin decompaction

Validation Data

Immunofluorescence:

  • Localizes to condensed chromatin in HeLa cells

  • Shows reduced signal upon histone H1.3 knockdown

Specificity Controls:

  • No cross-reactivity with H1.2 or H1.4 isoforms

  • Preabsorption with immunogen peptide abolishes signal

Comparative Analysis With Other HIST1H1D Antibodies

FeatureAb-16 (PACO56589)Ab-146 (PACO56663)Ab-106 (PACO60594)
EpitopeLys16Thr146Lys106
ApplicationsELISA, IFWB, IHC, IFWB, IHC
Species ReactivityHumanHuman, Mouse, RatHuman
Key FindingsMitotic chromatinCancer biomarkerChromatin remodeling

Limitations and Considerations

  • Requires antigen retrieval in paraffin-embedded tissues

  • May show reduced signal in cells with high H1.3 turnover

  • Commercial PTM-specific antibodies face challenges due to combinatorial histone modifications

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchasing method or location. Please consult your local distributor for specific delivery times.
Synonyms
H1 histone family member 3 antibody; H1.3 antibody; H13_HUMAN antibody; H1F3 antibody; HIST1 H1D antibody; HIST1H1D antibody; Histone 1 H1d antibody; Histone cluster 1 H1d antibody; Histone H1.3 antibody; Histone H1c antibody; MGC138176 antibody
Target Names
HIST1H1D
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 essential for the condensation of nucleosome chains into higher-order structured fibers. They also act as regulators of individual gene transcription through chromatin remodeling, nucleosome spacing, and DNA methylation.
Gene References Into Functions
  1. Studies indicate that histone H1.3 was identified only in non-neoplastic MCF-10A breast cells but not in metastatic MDA-MB-231 breast cancer cells. PMID: 26209608
  2. Histone H1 organizes and maintains an extensive protein-protein interaction network in the nucleolus, which is crucial for nucleolar structure and integrity. PMID: 25584861
  3. Overexpression of histone cluster 1 is associated with recurrence in meningiomas. PMID: 20685720
Database Links

HGNC: 4717

OMIM: 142210

KEGG: hsa:3007

STRING: 9606.ENSP00000244534

UniGene: Hs.136857

Protein Families
Histone H1/H5 family
Subcellular Location
Nucleus. Chromosome. Note=According to PubMed:15911621 more commonly found in euchromatin. According to PubMed:10997781 is associated with inactive chromatin.

Q&A

What is HIST1H1D and what role does it play in chromatin structure?

HIST1H1D, also known as Histone H1.3 or H1F3, is a member of the linker histone H1 family that binds to linker DNA between nucleosomes, forming the macromolecular structure known as the chromatin fiber. Histone H1 variants, including HIST1H1D, are necessary for the condensation of nucleosome chains into higher-order structured fibers . HIST1H1D functions as a regulator of individual gene transcription through multiple mechanisms including chromatin remodeling, nucleosome spacing, and DNA methylation . Recent research has classified HIST1H1D as a "low-GC" H1 variant that preferentially associates with low-GC content regions of the genome .

Methodologically, researchers investigating HIST1H1D's role in chromatin structure should consider combining immunofluorescence approaches with other techniques such as ChIP-seq to correlate nuclear distribution patterns with genomic binding sites.

What are the key characteristics of the HIST1H1D (Ab-16) Antibody?

The HIST1H1D (Ab-16) Antibody is a rabbit polyclonal antibody specifically designed to target the region around site of Lysine (16) derived from human Histone H1.3 . This antibody has the following key characteristics:

PropertyDescription
HostRabbit
ClonalityPolyclonal
TargetHIST1H1D (Histone H1.3)
ImmunogenPeptide sequence around site of Lys (16) derived from human Histone H1.3
ReactivityHuman
ApplicationsELISA, IF (Immunofluorescence)
FormLiquid
Diluent BufferPreservative: 0.03% Proclin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Storage-20°C or -80°C, avoid repeated freeze-thaw cycles

When using this antibody, researchers should be aware that the rabbit polyclonal format provides good sensitivity but may introduce some batch-to-batch variability, requiring appropriate validation for each new lot .

What applications is the HIST1H1D (Ab-16) Antibody validated for?

The HIST1H1D (Ab-16) Antibody has been validated for specific applications with recommended dilutions:

ApplicationValidatedRecommended Dilution
ELISAYes1:2000-1:10000
Immunofluorescence (IF)Yes1:50-1:200
Immunohistochemistry (IHC)Yes (in some sources)1:10-1:100

For immunofluorescence applications, researchers should optimize dilutions based on their specific experimental conditions, including fixation method, cell type, and detection system. Initial validation should include appropriate positive and negative controls, and co-localization with known nuclear markers may help confirm specificity .

How does the nuclear distribution of HIST1H1D (H1.3) compare to other histone H1 variants?

Immunofluorescence analysis has revealed distinct nuclear distribution patterns among histone H1 variants. HIST1H1D (H1.3), along with H1.2 and H1.5, exhibits universal enrichment toward the nuclear periphery across multiple cell lines . This contrasts with other H1 variants:

H1 VariantNuclear Distribution Pattern
H1.2, H1.3, H1.5Enriched at the nuclear periphery, coinciding with more condensed-DNA nuclear areas
H1.0Distributed throughout the nucleus with certain enrichment territories that tend to be peripheral
H1.4Homogeneously distributed throughout the nucleus, only partially mimicking DNA staining pattern
H1XHomogeneously distributed throughout the nucleus with variable nucleolar enrichment, coincides with less-stained DNA regions

These distribution patterns correlate with the classification of H1 variants into two differential groups: "low-GC" variants (H1.0, H1.2, H1.3, H1.5) and "high-GC" variants (H1.4, H1X). Co-immunostaining with heterochromatin marker HP1alpha has shown that low-GC H1 variants, including HIST1H1D, tend to better co-localize with heterochromatin compared to high-GC variants .

When designing experiments to study nuclear distribution, researchers should consider:

  • Using appropriate fixation methods that preserve nuclear architecture

  • Performing co-staining with DNA markers or other heterochromatin proteins

  • Comparing distribution across multiple cell types, as some patterns are cell-type specific

What is the role of HIST1H1D in regulating non-coding RNA and genome stability?

Recent research has uncovered an unexpected role for histone H1 variants, including HIST1H1D, in regulating non-coding RNA (ncRNA) turnover on chromatin. Depletion of histone H1 leads to:

  • Accumulation of de-regulated non-coding transcripts bound to chromatin

  • Increased RNA polymerase II (RNAPII) recruitment

  • Reduced levels of N-6-adenosine methylation (m6A) on nascent RNAs

  • Replication-transcription conflicts resulting in replicative stress

In cells with depleted histone H1, DNA fiber analysis has shown significant decreases in replication fork rate and increases in fork asymmetry. Importantly, these replicative phenotypes are transcription-dependent, as they can be reversed by inhibiting RNAPII elongation activity .

For researchers interested in this aspect of HIST1H1D function, methodological approaches should include:

  • Combining HIST1H1D (Ab-16) antibody with nascent RNA isolation techniques

  • Using DNA fiber analysis to assess replication dynamics

  • Employing transcription inhibitors to determine transcription-dependency

  • Analyzing m6A levels on chromatin-associated RNAs

How can HIST1H1D (Ab-16) Antibody be used in chromatin immunoprecipitation (ChIP) experiments?

While the product information for HIST1H1D (Ab-16) Antibody does not explicitly list ChIP as a validated application, similar histone H1 variant antibodies have been successfully used in ChIP-seq experiments to analyze genomic distribution patterns . When adapting this antibody for ChIP applications, researchers should consider:

  • Crosslinking optimization: Standard 1% formaldehyde crosslinking for 10 minutes may require adjustment for optimal H1 variant detection

  • Fragmentation conditions: Balanced sonication to generate 200-500bp fragments while preserving epitope integrity

  • Antibody validation: Performing preliminary ChIP-qPCR at known HIST1H1D enrichment sites

  • Controls: Using IgG controls and possibly other H1 variant antibodies for comparison

  • Analysis approach: Consider G-bands segmentation for comparing H1 variants binding profiles, as it has been demonstrated to be useful for epigenetic unit comparison

Expected genomic distribution: Based on research with other cell lines, HIST1H1D would be expected to show enrichment in low-GC content regions and underrepresentation in high-GC regions.

How does HIST1H1D depletion affect cellular phenotypes in different experimental systems?

Studies on cells with depleted histone H1 variants have revealed important consequences that researchers should consider when interpreting HIST1H1D knockdown experiments:

  • In mouse embryonic stem cells with triple knockout for H1 subtypes, researchers observed:

    • Genome-wide alterations in replication initiation patterns

    • Massive fork stalling and DNA damage due to replication-transcription conflicts

    • Accumulation of non-coding RNAs bound to chromatin

  • In human T47D breast cancer cells with inducible knockdown of H1.2 and H1.4:

    • Transcription-dependent replicative stress

    • Increased DNA damage signaling

    • Enhanced non-coding RNA chromatin association

  • In cell lines naturally lacking H1.3 and H1.5:

    • H1.0 and H1.4 show redistributed nuclear localization patterns

    • High basal expression of interferon signature genes

    • Elevated expression of repetitive elements

These findings suggest that histone H1 variants can have compensatory mechanisms in terms of distribution, but these may be limited when perturbing H1 levels artificially versus when the H1 repertoire is "naturally" compromised in certain cell types .

What methodological considerations should be taken when using HIST1H1D (Ab-16) Antibody for immunofluorescence studies?

When using HIST1H1D (Ab-16) Antibody for immunofluorescence, researchers should consider the following methodological aspects:

  • Fixation method:

    • Standard 4% paraformaldehyde fixation may be sufficient for nuclear proteins

    • Consider methanol fixation if detecting epitopes in highly compact chromatin regions

  • Permeabilization:

    • Optimize detergent concentration (typically 0.1-0.5% Triton X-100)

    • Duration should be sufficient to allow antibody access to nuclear proteins

  • Blocking:

    • Use 3-5% BSA or normal serum from the secondary antibody host species

    • Include 0.1-0.3% Triton X-100 in blocking solution for nuclear proteins

  • Antibody dilution:

    • Start with the recommended 1:50-1:200 range for immunofluorescence

    • Optimize based on signal-to-noise ratio in your specific cell type

  • Controls and validation:

    • Include secondary-only controls

    • Consider siRNA knockdown of HIST1H1D as a negative control

    • Co-staining with markers of nuclear periphery (lamin B1) or heterochromatin (HP1α) to confirm expected localization pattern

  • Image acquisition:

    • Use confocal microscopy for precise nuclear localization

    • Collect Z-stacks to fully capture the three-dimensional distribution

  • Quantification approaches:

    • Consider radial distribution analysis from nuclear periphery to center

    • Measure co-localization coefficients with heterochromatin markers

What are common challenges when working with histone antibodies like HIST1H1D (Ab-16)?

Researchers commonly encounter several challenges when working with histone antibodies:

  • Epitope masking: Histones are often tightly associated with DNA and other proteins, potentially masking epitopes. This can be addressed by:

    • Optimizing fixation and permeabilization protocols

    • Using antigen retrieval methods such as heat-induced epitope retrieval

    • Testing different extraction buffers with varying salt concentrations

  • Cross-reactivity: Given the high sequence similarity between histone variants, antibody cross-reactivity is a concern. Researchers should:

    • Validate antibody specificity using knockout/knockdown controls

    • Compare immunofluorescence patterns with published distribution data for HIST1H1D

    • Consider peptide competition assays to confirm specificity

  • Batch-to-batch variability: Polyclonal antibodies like HIST1H1D (Ab-16) may show variability between lots. To address this:

    • Validate each new lot against previous batches

    • Purchase sufficient quantities of a single lot for long-term projects

    • Maintain detailed records of antibody performance across experiments

  • Signal intensity variations: Nuclear proteins may show variable staining intensity. Consider:

    • Optimizing antibody concentration and incubation time

    • Using signal amplification methods if needed

    • Standardizing image acquisition settings across experiments

How can the specificity of HIST1H1D (Ab-16) Antibody be validated in experimental systems?

To ensure the specificity of the HIST1H1D (Ab-16) Antibody, researchers should implement a comprehensive validation strategy:

  • Western blot analysis:

    • Confirm single band at expected molecular weight (~22 kDa)

    • Compare with recombinant HIST1H1D protein as positive control

    • Test in cells with HIST1H1D knockdown/knockout as negative control

  • Peptide competition assay:

    • Pre-incubate antibody with immunizing peptide

    • Observe elimination of specific signal

  • Cross-reactivity assessment:

    • Test antibody against recombinant versions of other H1 variants

    • Compare immunofluorescence patterns with published distribution patterns of H1.3

  • Multi-technique validation:

    • Confirm consistency of results across different applications (IF, ELISA)

    • Compare subcellular localization with published data on nuclear periphery enrichment

  • Mass spectrometry validation:

    • Perform immunoprecipitation followed by mass spectrometry

    • Confirm presence of HIST1H1D and assess presence of other proteins

How can HIST1H1D (Ab-16) Antibody be used to study changes in chromatin organization during cellular processes?

The HIST1H1D (Ab-16) Antibody can be valuable for studying dynamic changes in chromatin organization during various cellular processes:

  • Cell cycle progression:

    • Combine with cell cycle markers (e.g., PCNA, cyclin antibodies)

    • Analyze changes in HIST1H1D distribution during different cell cycle phases

    • Quantify intensity and pattern changes from G1 through mitosis

  • Cellular differentiation:

    • Track HIST1H1D distribution changes during differentiation protocols

    • Compare with changes in other epigenetic marks (e.g., H3K9me3, H3K27me3)

    • Correlate with transcriptional changes of genes regulated by HIST1H1D

  • Cellular stress response:

    • Examine redistribution following DNA damage, oxidative stress, or heat shock

    • Combine with markers of stress response (γH2AX, stress granules)

    • Time-course analysis to track dynamic changes

  • Oncogenic transformation:

    • Compare HIST1H1D patterns between normal and cancer cell lines

    • Analyze in the context of heterochromatin reorganization in cancer

    • Correlate with aberrant gene expression patterns

Methodological approach:

  • Fixed timepoint analysis: Fix cells at defined timepoints during the process of interest

  • Live-cell imaging: Consider using complementary approaches such as GFP-tagged H1.3 for real-time tracking

  • Correlative microscopy: Combine immunofluorescence with other imaging modalities

  • Multi-omics integration: Correlate imaging data with ChIP-seq or RNA-seq at matching timepoints

How does HIST1H1D function differ between normal cells and cancer cells?

Research suggests notable differences in HIST1H1D function and distribution between normal and cancer cells:

  • Expression patterns:

    • Some cancer cell lines show altered expression of H1 variants

    • Certain cancer cell lines naturally lack H1.3 and H1.5, suggesting these may represent an acquired adaptive mechanism

  • Nuclear distribution:

    • While H1.3 is typically enriched at the nuclear periphery, its distribution pattern may be altered in cancer cells with disrupted nuclear architecture

    • In cells lacking H1.3 and H1.5, H1.0 and H1.4 show redistributed nuclear localization patterns

  • Functional consequences:

    • Cells lacking H1.3 and H1.5 exhibit high basal expression of interferon signature genes

    • These cells also show elevated expression of repetitive elements

    • This suggests potential implications for immune response in these cancer cells

For researchers studying HIST1H1D in cancer contexts, consider:

  • Comparing multiple cancer cell lines with normal tissue counterparts

  • Analyzing correlation between HIST1H1D patterns and clinical outcomes

  • Investigating potential connections between H1.3 loss and interferon pathway activation in tumor microenvironment

What approaches can be used to study the interaction between HIST1H1D and other chromatin-associated proteins?

To investigate interactions between HIST1H1D and other chromatin-associated proteins, researchers can employ several complementary approaches:

  • Co-immunoprecipitation (Co-IP):

    • Use HIST1H1D (Ab-16) Antibody for pulldown experiments

    • Analyze co-precipitated proteins by western blot or mass spectrometry

    • Verify interactions with reciprocal Co-IP experiments

  • Proximity ligation assay (PLA):

    • Detect protein-protein interactions in situ

    • Particularly useful for nuclear proteins with close spatial proximity

    • Can detect interactions within 40nm distance

  • Fluorescence resonance energy transfer (FRET):

    • Requires fluorescently-tagged proteins

    • Provides information on direct protein-protein interactions

    • Can be performed in living cells

  • Chromatin immunoprecipitation followed by mass spectrometry (ChIP-MS):

    • Identify proteins co-occupying the same chromatin regions as HIST1H1D

    • Does not necessarily indicate direct interaction but functional association

  • Co-localization analysis:

    • Perform dual immunofluorescence with HIST1H1D (Ab-16) Antibody and antibodies against potential interacting partners

    • Quantify co-localization using appropriate coefficients (Pearson's, Manders')

    • Focus particularly on co-localization with heterochromatin markers like HP1α, which has been shown to co-localize with low-GC H1 variants

How can researchers integrate HIST1H1D studies with other epigenetic analyses?

Integrating HIST1H1D studies with broader epigenetic analyses can provide comprehensive insights into chromatin regulation:

  • Multi-omics integration approaches:

    • Combine HIST1H1D ChIP-seq with histone modification ChIP-seq (H3K9me3, H3K27me3)

    • Integrate with DNA methylation data (WGBS, RRBS)

    • Correlate with chromatin accessibility data (ATAC-seq, DNase-seq)

    • Compare with transcriptome data (RNA-seq, particularly for non-coding RNAs)

  • Spatial chromatin organization:

    • Relate HIST1H1D binding to TAD (Topologically Associated Domain) boundaries

    • Compare with Hi-C data to understand 3D genome organization

    • Analyze relation to nuclear lamina-associated domains (LADs)

  • Functional genomics:

    • Use CRISPR-based approaches to modify HIST1H1D binding sites

    • Assess impact on local chromatin structure and gene expression

    • Perform rescue experiments with wild-type or mutant HIST1H1D

  • Single-cell approaches:

    • Combine with single-cell transcriptomics or epigenomics

    • Analyze cell-to-cell variability in HIST1H1D distribution

    • Identify subpopulations with distinct HIST1H1D patterns

  • Computational modeling:

    • Develop predictive models of HIST1H1D binding based on DNA sequence and other epigenetic marks

    • Use machine learning approaches to identify features associated with HIST1H1D enrichment

    • Model the impact of HIST1H1D on chromatin fiber compaction

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.