HIST1H1D encodes histone H1.3, which:
Mouse knockout studies reveal that triple deletion of H1d/c/e causes embryonic lethality due to chromatin decompaction, emphasizing its structural role .
The antibody enables:
HIST1H1D dysregulation correlates with:
Immunofluorescence:
Specificity Controls:
Feature | Ab-16 (PACO56589) | Ab-146 (PACO56663) | Ab-106 (PACO60594) |
---|---|---|---|
Epitope | Lys16 | Thr146 | Lys106 |
Applications | ELISA, IF | WB, IHC, IF | WB, IHC |
Species Reactivity | Human | Human, Mouse, Rat | Human |
Key Findings | Mitotic chromatin | Cancer biomarker | Chromatin remodeling |
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.
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:
Property | Description |
---|---|
Host | Rabbit |
Clonality | Polyclonal |
Target | HIST1H1D (Histone H1.3) |
Immunogen | Peptide sequence around site of Lys (16) derived from human Histone H1.3 |
Reactivity | Human |
Applications | ELISA, IF (Immunofluorescence) |
Form | Liquid |
Diluent Buffer | Preservative: 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 .
The HIST1H1D (Ab-16) Antibody has been validated for specific applications with recommended dilutions:
Application | Validated | Recommended Dilution |
---|---|---|
ELISA | Yes | 1:2000-1:10000 |
Immunofluorescence (IF) | Yes | 1:50-1:200 |
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 .
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 Variant | Nuclear Distribution Pattern |
---|---|
H1.2, H1.3, H1.5 | Enriched at the nuclear periphery, coinciding with more condensed-DNA nuclear areas |
H1.0 | Distributed throughout the nucleus with certain enrichment territories that tend to be peripheral |
H1.4 | Homogeneously distributed throughout the nucleus, only partially mimicking DNA staining pattern |
H1X | Homogeneously 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
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
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.
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:
In human T47D breast cancer cells with inducible knockdown of H1.2 and H1.4:
In cell lines naturally lacking H1.3 and H1.5:
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 .
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:
Controls and validation:
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
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
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:
Multi-technique validation:
Mass spectrometry validation:
Perform immunoprecipitation followed by mass spectrometry
Confirm presence of HIST1H1D and assess presence of other proteins
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
Research suggests notable differences in HIST1H1D function and distribution between normal and cancer cells:
Expression patterns:
Nuclear distribution:
Functional consequences:
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
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
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