HIST1H1D (Ab-146) Antibody

Shipped with Ice Packs
In Stock

Description

Target Biology

Histone H1.3 (encoded by HIST1H1D) is a linker histone that binds to DNA between nucleosomes, facilitating chromatin compaction into higher-order structures . Key functional roles include:

  • Chromatin remodeling: Regulates nucleosome spacing and DNA methylation .

  • Transcriptional control: Modulates gene expression by altering chromatin accessibility .

  • Disease associations: Dysregulation linked to cancers (e.g., ovarian, colorectal) and neurodegenerative disorders .

Western Blot

  • Detects endogenous histone H1.3 in PC-3 cell lysates, mouse spleen, and rat spleen tissues at 23 kDa .

  • Secondary antibody: Goat anti-rabbit IgG (1:50,000 dilution) .

Immunohistochemistry

  • Validated in paraffin-embedded human ovarian cancer tissues at 1:100 dilution .

Immunofluorescence

  • Localizes histone H1.3 to nuclei in HeLa cells using Alexa Fluor 488-conjugated secondary antibodies .

Epigenetic Studies

  • Identifies histone H1.3 dynamics during chromatin remodeling and transcriptional silencing .

Cancer Biology

  • Used to analyze histone H1.3 expression patterns in ovarian and colorectal cancer biopsies .

Developmental Biology

  • Maps chromatin structural changes during cellular differentiation .

Comparative Insights

While HIST1H1D (Ab-146) detects the unmodified form of histone H1.3, its phosphorylated counterpart (e.g., at Thr-146) requires specialized antibodies (e.g., PACO56661) to study cell cycle-dependent modifications .

Quality Controls

  • Specificity: No cross-reactivity with non-target histones confirmed via knockout cell line testing .

  • Batch Consistency: Rigorous validation across multiple lots ensures reproducibility .

Limitations

  • Species-restricted reactivity (non-primate models require validation) .

  • Not suited for flow cytometry due to fixation-dependent epitope accessibility .

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 the products within 1-3 business days after receiving your order. Delivery timelines may vary depending on the purchasing method or location. For specific delivery times, please consult your local distributors.
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. Research indicates that histone H1.3 was only identified 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 within the nucleolus, which is crucial for nucleolar structure and integrity. PMID: 25584861
  3. Overexpression of histone cluster 1 has been 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 biological functions does it serve?

HIST1H1D, also known as Histone H1.3, is a member of the H1 histone family that plays a critical role in chromatin organization and gene regulation. Histone H1 proteins bind to linker DNA between nucleosomes, forming the macromolecular structure known as the chromatin fiber. This binding is essential for the condensation of nucleosome chains into higher-order structured fibers . Beyond its structural role, HIST1H1D functions as a regulator of individual gene transcription through mechanisms including chromatin remodeling, nucleosome spacing, and DNA methylation . The protein is particularly important in epigenetic processes that control gene expression patterns. Dysregulation of histone H1.3 has been implicated in various diseases, including cancer and neurodegenerative disorders, highlighting its significance in maintaining normal cellular function .

What are the key characteristics of the HIST1H1D (Ab-146) Antibody?

The HIST1H1D (Ab-146) Antibody (PACO56663) is a rabbit polyclonal antibody specifically designed to recognize and bind to the region around the Threonine 146 site of human Histone H1.3 protein . This antibody has been affinity-purified against the target antigen to ensure high specificity and reduced background . It is provided in liquid form, stored in a buffer containing 0.03% Proclin 300, 50% Glycerol, and 0.01M PBS at pH 7.4 to maintain stability . The antibody demonstrates cross-reactivity with human, mouse, and rat samples, making it versatile for comparative studies across these species . It has been validated for multiple applications including Western blot (WB), immunohistochemistry (IHC), immunofluorescence (IF), and ELISA, with specific recommended dilutions for each application .

How does the HIST1H1D (Ab-146) Antibody differ from other HIST1H1D antibodies?

The HIST1H1D (Ab-146) Antibody is specifically designed to recognize the region surrounding Threonine 146 in human Histone H1.3 protein . This distinguishes it from other HIST1H1D antibodies that target different epitopes or post-translational modifications. For example, there are antibodies specifically designed to detect phosphorylated Threonine 146 (pThr146) , while others target different regions such as amino acids 137-149 or 135-164 . Some antibodies recognize specific modifications like methylated Lysine 16 (meLys16) . The specificity for the region around Threonine 146 without requiring phosphorylation makes this antibody ideal for detecting total HIST1H1D protein regardless of its phosphorylation status . In Western blot applications, this antibody detects a band at approximately 23 kDa, corresponding to the predicted molecular weight of HIST1H1D . When selecting a HIST1H1D antibody, researchers should consider which specific epitope or modification is most relevant to their research question.

What sample types can be effectively analyzed using this antibody?

The HIST1H1D (Ab-146) Antibody has demonstrated effective detection in various sample types from human, mouse, and rat origins . For Western blot applications, successful detection has been validated in whole cell lysates from multiple cell lines including PC-3 (human prostate cancer cells) . Additionally, the antibody effectively detects the target protein in tissue samples such as rat and mouse spleen tissues . For immunohistochemistry applications, the antibody can be used on both frozen and paraffin-embedded tissue sections when proper antigen retrieval methods are employed. In immunofluorescence studies, the antibody works well with fixed and permeabilized cell preparations. The versatility across multiple sample types makes this antibody a valuable tool for researchers conducting comparative studies across different experimental models or investigating HIST1H1D expression in various cellular contexts.

What are the optimal dilutions and conditions for different applications of the HIST1H1D (Ab-146) Antibody?

For optimal results with the HIST1H1D (Ab-146) Antibody, specific dilution ranges have been validated for different applications:

ApplicationRecommended DilutionSpecial Considerations
ELISA1:2000-1:10000Higher dilutions suitable for this sensitive method
Western Blot1:200-1:20001.5 μg/ml concentration has been validated with goat anti-rabbit IgG secondary at 1:50000
IHC1:20-1:200May require optimization based on tissue type and fixation method
IF1:50-1:200Lower dilutions typically needed compared to WB

For Western blot applications, the antibody has been successfully used at 1.5 μg/ml with secondary goat polyclonal to rabbit IgG at 1:50000 dilution . The predicted band size is 23 kDa, which matches the observed band size in validated samples . When designing experiments, researchers should consider running an initial dilution series to determine the optimal concentration for their specific sample type and experimental conditions. For all applications, proper blocking (typically 5% BSA or 5% non-fat milk in TBST) is recommended to minimize background staining.

How should I design a Western blot protocol for detecting HIST1H1D using this antibody?

When designing a Western blot protocol for HIST1H1D detection using the (Ab-146) antibody, follow these methodological steps for optimal results:

  • Sample preparation: Extract proteins using a buffer containing protease inhibitors. For histone extraction, consider specialized protocols that effectively extract nuclear proteins.

  • Protein quantification: Use Bradford or BCA assay to ensure equal loading (typically 20-30 μg total protein per lane).

  • Gel electrophoresis: Use 12-15% SDS-PAGE gels as HIST1H1D is a relatively small protein (23 kDa) .

  • Transfer: Transfer proteins to PVDF or nitrocellulose membrane using standard transfer buffer at 100V for 1 hour or 30V overnight.

  • Blocking: Block membrane with 5% non-fat milk or BSA in TBST for 1 hour at room temperature.

  • Primary antibody incubation: Dilute HIST1H1D (Ab-146) Antibody 1:200-1:2000 in blocking buffer and incubate overnight at 4°C . A concentration of 1.5 μg/ml has been validated for PC-3 whole cell lysate, rat spleen tissue, and mouse spleen tissue .

  • Washing: Wash membrane 3×10 minutes with TBST.

  • Secondary antibody: Incubate with goat anti-rabbit IgG-HRP at 1:50000 dilution for 1 hour at room temperature .

  • Washing: Wash membrane 3×10 minutes with TBST.

  • Detection: Develop using ECL substrate and image using appropriate detection system.

Include positive controls such as PC-3, rat spleen, or mouse spleen tissue lysates, which have been validated with this antibody . The expected band size is 23 kDa.

What controls and optimization strategies should I implement for immunofluorescence studies?

For robust immunofluorescence studies using the HIST1H1D (Ab-146) Antibody, implement the following controls and optimization strategies:

Essential controls:

  • Positive control: Include cells or tissues known to express HIST1H1D, such as PC-3 cells which have been validated with this antibody .

  • Negative control: Omit primary antibody while maintaining all other steps to assess background from secondary antibody.

  • Isotype control: Use non-specific rabbit IgG at the same concentration as the primary antibody to evaluate non-specific binding.

  • Peptide competition: Pre-incubate antibody with the immunizing peptide to confirm specificity.

Optimization strategies:

  • Fixation method: Compare paraformaldehyde (4%) with methanol fixation, as histone epitopes can be sensitive to fixation method.

  • Permeabilization: Test different permeabilization reagents (0.1-0.5% Triton X-100, 0.1-0.5% Saponin) and durations (5-15 minutes).

  • Antigen retrieval: For tissue sections, compare heat-induced epitope retrieval methods (citrate buffer pH 6.0 vs. EDTA buffer pH 8.0).

  • Antibody dilution: Test a range within 1:50-1:200 as recommended for this antibody .

  • Incubation conditions: Compare room temperature (1-2 hours) versus 4°C (overnight) incubation.

  • Blocking reagent: Test different blocking solutions (5% normal goat serum, 3% BSA, or commercial blocking reagents).

When analyzing results, look for nuclear localization of HIST1H1D, which would be consistent with its role in chromatin organization. Document all optimization parameters systematically to establish a reproducible protocol.

How can I validate the specificity of HIST1H1D (Ab-146) Antibody in my experimental system?

Validating antibody specificity is crucial for generating reliable data. For the HIST1H1D (Ab-146) Antibody, implement these methodological approaches:

  • Multi-application validation: Confirm target detection across different techniques (WB, IHC, IF, ELISA) as specificity issues may be revealed in certain applications but not others. This antibody has been validated for all these applications .

  • Knockdown/knockout validation:

    • Perform siRNA or shRNA knockdown of HIST1H1D

    • Use CRISPR-Cas9 to generate HIST1H1D knockout cells

    • Compare antibody reactivity between wildtype and knockdown/knockout samples; signal should be significantly reduced or absent in the latter

  • Peptide competition assay:

    • Pre-incubate the antibody with excess immunizing peptide (sequence around Thr146 of HIST1H1D)

    • Run parallel experiments with blocked and unblocked antibody

    • Specific signals should be abolished or significantly reduced with peptide-blocked antibody

  • Cross-reactivity assessment:

    • Test the antibody against recombinant proteins of related histone variants (HIST1H1A, HIST1H1B, HIST1H1C, HIST1H1E)

    • The antibody should show higher affinity for HIST1H1D compared to other variants

  • Molecular weight verification:

    • Confirm detected band size matches the predicted 23 kDa for HIST1H1D

    • Be aware of potential post-translational modifications that might alter migration

  • Comparison with alternative antibodies:

    • Test another validated HIST1H1D antibody targeting a different epitope

    • The staining pattern should be similar if both antibodies are specific

Document validation results thoroughly to support the reliability of subsequent experimental findings.

Why might I observe non-specific binding or high background when using this antibody?

Non-specific binding and high background are common challenges when working with antibodies. When using the HIST1H1D (Ab-146) Antibody, several factors may contribute to these issues:

  • Insufficient blocking: Histone proteins are abundant nuclear components that can lead to high background. Optimize blocking by:

    • Increasing blocking agent concentration (try 5-10% BSA or normal serum)

    • Extending blocking time from 1 hour to 2 hours

    • Using specialized blocking reagents designed for nuclear proteins

    • Adding 0.1-0.2% Tween-20 to the blocking solution

  • Antibody concentration: Using too high a concentration can increase non-specific binding. Perform a dilution series starting from the higher end of the recommended range (1:200 for WB and 1:50 for IF) and systematically test more dilute solutions.

  • Cross-reactivity with related proteins: Histone family proteins share sequence homology. While this antibody is designed for HIST1H1D specificity, it may recognize related histone proteins. Validate specificity using the methods described in question 2.4.

  • Sample preparation issues:

    • Incomplete permeabilization for IF can cause uneven staining

    • Excessive antigen retrieval can expose non-specific epitopes

    • Improper fixation can alter protein conformation

  • Secondary antibody problems:

    • Secondary antibody concentration may be too high

    • Consider using highly cross-adsorbed secondary antibodies

    • Try a different secondary antibody from another vendor

  • Buffer composition: The storage buffer contains 50% glycerol and 0.03% Proclin 300 , which at high antibody concentrations could affect staining quality. Ensure proper dilution in appropriate buffers for each application.

What factors might affect epitope accessibility and antibody binding to HIST1H1D?

Several factors can significantly impact epitope accessibility and binding efficiency of the HIST1H1D (Ab-146) Antibody:

  • Chromatin compaction state: Histone H1.3 is integral to chromatin structure, and its accessibility may vary depending on chromatin compaction. The antibody targets the region around Threonine 146 , which may be differentially exposed based on chromatin state. Consider using chromatin relaxation methods (like TSA treatment) before fixation if detecting chromatin-bound HIST1H1D.

  • Post-translational modifications (PTMs): The region around Threonine 146 can undergo various PTMs including phosphorylation, which could directly affect antibody binding. The antibody was raised against the unmodified sequence , so phosphorylation at Thr146 might reduce recognition. Consider parallel experiments with phospho-specific antibodies if this modification is relevant to your research.

  • Protein-protein interactions: HIST1H1D interacts with linker DNA and other nuclear proteins, potentially masking the epitope. Optimize fixation and extraction methods to preserve the native state while maintaining epitope accessibility.

  • Fixation methods:

    • Formaldehyde crosslinking may obscure nuclear epitopes

    • Methanol fixation might better preserve nuclear protein epitopes

    • Test different fixation protocols to determine optimal conditions

  • Antigen retrieval parameters:

    • pH conditions (citrate buffer pH 6.0 vs. EDTA buffer pH 8.0)

    • Retrieval duration (10-20 minutes)

    • Temperature (95-100°C vs. lower temperature for longer time)

  • Sample processing artifacts:

    • Freeze-thaw cycles can degrade epitopes

    • Prolonged storage of fixed samples may reduce antigenicity

    • Embedding procedures can mask nuclear antigens

  • Cell/tissue type variations: Different cell types may express varying levels of HIST1H1D and exhibit different chromatin organizations, affecting epitope accessibility and requiring application-specific optimization.

How can I troubleshoot weak or absent signals when using HIST1H1D (Ab-146) Antibody?

When encountering weak or absent signals with the HIST1H1D (Ab-146) Antibody, systematically troubleshoot using this methodological approach:

  • Confirm target expression:

    • Verify HIST1H1D expression in your sample through RT-PCR or RNA-seq

    • Include positive control samples like PC-3, rat spleen, or mouse spleen tissues

    • Consider cell cycle phase, as histone expression may vary

  • Antibody handling:

    • Check antibody storage conditions (should be stored at -20°C in aliquots)

    • Avoid repeated freeze-thaw cycles that can degrade antibody

    • Verify antibody hasn't expired or been contaminated

    • Consider ordering fresh antibody if problems persist

  • Protocol optimization for Western blot:

    • Increase protein loading (up to 50-75 μg)

    • Reduce antibody dilution (try 1:100-1:200 range)

    • Extend primary antibody incubation (overnight at 4°C)

    • Use more sensitive detection systems (ECL Plus or Super Signal)

    • Ensure transfer efficiency using Ponceau S staining

    • Consider specialized extraction buffers for nuclear proteins

  • Protocol optimization for IHC/IF:

    • Test different antigen retrieval methods

    • Extend primary antibody incubation time (overnight at 4°C)

    • Use signal amplification systems (tyramide signal amplification)

    • Ensure adequate permeabilization for nuclear antigen

    • Try lower antibody dilutions (1:20-1:50 range for IHC)

  • Epitope masking issues:

    • The region around Thr146 may be masked by protein interactions

    • Try alternative extraction/lysis methods

    • Consider mild denaturation steps to expose the epitope

  • Extraction efficiency:

    • Nuclear proteins require specialized extraction methods

    • Use histone extraction protocols with acid extraction

    • Ensure complete nuclear lysis and histone solubilization

  • Detection system issues:

    • Verify secondary antibody works with a different primary antibody

    • Check detection reagents with a control sample

Document all troubleshooting steps systematically to establish optimal conditions for your specific experimental system.

Why might I see different band sizes than expected in Western blot using this antibody?

When Western blotting with HIST1H1D (Ab-146) Antibody shows unexpected band sizes beyond the expected 23 kDa , consider these scientific explanations and methodological approaches:

  • Post-translational modifications:

    • Phosphorylation, particularly at Thr146 (the region recognized by this antibody), can cause mobility shifts

    • Ubiquitination adds approximately 8.5 kDa per ubiquitin moiety

    • SUMOylation can add 11-20 kDa

    • ADP-ribosylation can significantly alter migration patterns

  • Protein isoforms:

    • Alternative splicing variants of HIST1H1D may exist

    • Verify against database information for known isoforms

    • Consider RNA-seq data to identify potential novel variants in your system

  • Proteolytic cleavage:

    • Incomplete protease inhibition during sample preparation

    • Endogenous nuclear proteases may partially cleave HIST1H1D

    • Store samples at -80°C and add additional protease inhibitors

  • Protein aggregation or oligomerization:

    • Incomplete denaturation can lead to dimers/multimers (look for bands at ~46 kDa or higher)

    • Increase SDS concentration or β-mercaptoethanol in loading buffer

    • Heat samples at 95°C for 5-10 minutes before loading

  • Protein-protein interactions:

    • Strong interaction partners resistant to SDS denaturation

    • Try more stringent lysis conditions or stronger reducing agents

  • Gel artifacts:

    • Uneven polymerization causing irregular migration

    • Air bubbles or contamination affecting band patterns

    • Use pre-cast gels to ensure consistency

  • Cross-reactivity:

    • The antibody may recognize related histone variants

    • HIST1H1A, HIST1H1B, HIST1H1C, and HIST1H1E have similar molecular weights but may migrate differently

    • Perform peptide competition assays to determine specificity

  • Methodology to verify true bands:

    • Use mass spectrometry to identify proteins in unexpected bands

    • Perform siRNA knockdown of HIST1H1D and observe which bands diminish

    • Compare patterns with another validated HIST1H1D antibody targeting a different epitope

    • Use recombinant HIST1H1D as a positive control

When reporting unexpected bands, clearly document their sizes, consistency across experiments, and any verification steps taken to confirm or refute their identity as HIST1H1D-related proteins.

How can I use HIST1H1D (Ab-146) Antibody for chromatin immunoprecipitation (ChIP) studies?

While the HIST1H1D (Ab-146) Antibody hasn't been explicitly validated for ChIP in the provided data, related HIST1H1D antibodies have been used successfully for this application . Here's a methodological approach to adapt this antibody for ChIP studies:

  • ChIP protocol optimization:

    • Start with standard ChIP protocols but optimize for histone linker proteins

    • Use dual crosslinking (DSG followed by formaldehyde) to better preserve protein-DNA interactions

    • Sonication conditions should be carefully optimized (typically 15-25 cycles of 30s on/30s off) to generate 200-500 bp fragments

    • Verify sonication efficiency by agarose gel electrophoresis

  • Antibody amount determination:

    • Titrate antibody amount (2-10 μg per reaction)

    • Include IgG control at the highest antibody concentration

    • Include a positive control antibody targeting core histones (H3 or H4)

    • Consider pre-clearing chromatin with protein A/G beads

  • Experimental validation:

    • Perform ChIP-qPCR on regions known to be associated with H1 histones

    • Include both heterochromatic and euchromatic regions

    • Compare enrichment patterns with published H1 ChIP-seq datasets

    • Verify antibody specificity by performing ChIP after HIST1H1D knockdown

  • Advanced ChIP applications:

    • For genome-wide studies, consider ChIP-seq with 10-20 million reads minimum

    • For spatiotemporal dynamics, combine with proximity ligation assays

    • For protein complex identification, consider ChIP followed by mass spectrometry

    • For studying relationships with other chromatin features, perform sequential ChIP

  • Data analysis considerations:

    • HIST1H1D typically shows broad enrichment patterns rather than sharp peaks

    • Analysis should account for linker histone dynamics and potential partial occupancy

    • Compare with marks of heterochromatin (H3K9me3, H3K27me3) and euchromatin (H3K4me3, H3K27ac)

    • Consider nucleosome positioning data to interpret HIST1H1D binding patterns

  • Technical challenges to anticipate:

    • The region around Thr146 may be involved in DNA binding, potentially affecting epitope accessibility

    • Chromatin compaction state influences HIST1H1D occupancy and ChIP efficiency

    • Cell cycle variations affect linker histone occupancy and should be considered in experimental design

This methodological approach leverages the specificity of the HIST1H1D (Ab-146) Antibody for advanced chromatin studies while accounting for the unique challenges of linker histone ChIP experiments.

How can I study the impact of Thr146 phosphorylation on HIST1H1D function?

The HIST1H1D (Ab-146) Antibody recognizes the region around Threonine 146 , making it valuable for studying this key regulatory site. To comprehensively investigate the impact of Thr146 phosphorylation on HIST1H1D function, implement this multi-faceted research strategy:

  • Comparative antibody approach:

    • Use both HIST1H1D (Ab-146) Antibody (total protein) and phospho-specific antibodies targeting pThr146

    • Compare signal ratios to determine phosphorylation state across conditions

    • Use Lambda phosphatase treatment to confirm phosphorylation-dependent signals

  • Phosphorylation induction and inhibition studies:

    • Treatment with CDK inhibitors (CDK2 and CDK1 are known to phosphorylate H1)

    • Cell cycle synchronization to study phase-specific phosphorylation

    • Stress conditions (UV, oxidative stress) that may trigger histone phosphorylation

    • Analyze samples using Western blotting with both antibodies

  • Mutagenesis approaches:

    • Generate Thr146 to Ala mutants (phospho-deficient)

    • Generate Thr146 to Glu mutants (phospho-mimetic)

    • Express tagged versions in cells for localization and functional studies

    • Perform rescue experiments in HIST1H1D-depleted backgrounds

  • Chromatin structural analysis:

    • Micrococcal nuclease sensitivity assays with wild-type vs. mutant HIST1H1D

    • FRAP (Fluorescence Recovery After Photobleaching) to measure chromatin binding dynamics

    • Electron microscopy to visualize chromatin compaction states

    • ATAC-seq to assess chromatin accessibility changes

  • Gene expression analysis:

    • RNA-seq comparing cells with wild-type, phospho-deficient, and phospho-mimetic HIST1H1D

    • ChIP-seq to correlate HIST1H1D phosphorylation with genomic binding sites

    • Nascent RNA sequencing to detect immediate transcriptional effects

    • Combined with proteomics to identify altered protein interactions

  • Mass spectrometry analysis:

    • Identify other PTMs that co-occur with Thr146 phosphorylation

    • Quantify phosphorylation stoichiometry across conditions

    • Identify protein interaction partners specific to phosphorylated vs. non-phosphorylated states

    • Develop targeted MS methods for Thr146 phosphopeptide detection

  • Functional consequences:

    • DNA damage response assays (γH2AX foci, comet assay)

    • Cell cycle progression analysis

    • Apoptosis sensitivity

    • Transcriptional reporter assays at HIST1H1D-regulated genes

This comprehensive approach leverages the specificity of both the HIST1H1D (Ab-146) Antibody and phospho-specific antibodies to elucidate the functional significance of this critical modification in chromatin biology.

What techniques can I combine with immunoprecipitation to study HIST1H1D protein interactions?

To comprehensively characterize HIST1H1D protein interactions using the HIST1H1D (Ab-146) Antibody for immunoprecipitation (IP), consider these advanced methodological approaches:

  • IP-Mass Spectrometry (IP-MS):

    • Use HIST1H1D (Ab-146) Antibody for IP from nuclear extracts

    • Analyze by LC-MS/MS to identify all interacting proteins

    • Implement SILAC or TMT labeling for quantitative comparison across conditions

    • Use stringent controls (IgG IP, HIST1H1D-depleted cells)

    • Cross-validate top hits with reciprocal IP using antibodies against identified partners

  • Proximity-dependent labeling:

    • Generate BioID or TurboID fusion with HIST1H1D

    • Express in cells and activate biotin labeling

    • Use streptavidin pulldown followed by MS

    • Compare interactome with conventional IP results

    • Identify transient or weak interactions missed by standard IP

  • Co-IP with sequential elution:

    • Use HIST1H1D (Ab-146) Antibody for initial IP

    • Perform sequential elution with increasing salt or detergent stringency

    • Analyze fractions to distinguish high-affinity from weak interactions

    • Validate interaction strength with biophysical methods (SPR, ITC)

  • ChIP-MS approaches:

    • Perform ChIP with HIST1H1D (Ab-146) Antibody

    • Instead of DNA purification, analyze protein content

    • Identify chromatin-associated interaction partners

    • Compare with soluble nuclear fraction IP to distinguish chromatin-dependent interactions

  • IP followed by enzymatic activity assays:

    • After IP with HIST1H1D (Ab-146) Antibody, test immunoprecipitates for:

    • Histone deacetylase (HDAC) activity

    • Histone methyltransferase activity

    • ATP-dependent chromatin remodeling activity

    • DNA methyltransferase activity

  • FRET/BRET interaction studies:

    • Generate fluorescent protein fusions with HIST1H1D and candidate partners

    • Perform live-cell interaction studies

    • Validate IP results in the native cellular environment

    • Study dynamics of interactions during cell cycle or stress

  • IP combined with chromatin analysis:

    • IP HIST1H1D-containing complexes

    • Extract and sequence associated DNA

    • Perform ChIP-seq on specific partners identified by IP-MS

    • Create interaction maps that incorporate genomic localization

  • Cross-linking IP (X-IP):

    • Use protein cross-linkers before IP to capture transient interactions

    • Identify cross-linked peptides by MS

    • Map interaction interfaces between HIST1H1D and partners

    • Provide structural insights into complex formation

Each approach provides complementary information about HIST1H1D interactions, from identifying novel partners to characterizing the nature and context of these interactions, ultimately building a comprehensive understanding of HIST1H1D's role in nuclear processes.

How can I use HIST1H1D (Ab-146) Antibody to investigate epigenetic changes in disease models?

The HIST1H1D (Ab-146) Antibody can be instrumental in investigating epigenetic alterations in disease models through these methodological approaches:

  • Comparative expression profiling:

    • Analyze HIST1H1D levels in healthy versus diseased tissues using Western blot and IHC

    • Quantify differences in expression levels, subcellular localization, and post-translational modifications

    • Create tissue microarrays for high-throughput screening across multiple disease samples

    • Correlate findings with clinical parameters and patient outcomes

  • Cell-type specific analysis in complex tissues:

    • Combine IF using HIST1H1D (Ab-146) Antibody with cell type-specific markers

    • Implement multiplex immunofluorescence to assess HIST1H1D in different cell populations within heterogeneous tissues

    • Use confocal microscopy with spectral unmixing for high-resolution localization

    • Apply quantitative image analysis to measure nuclear HIST1H1D intensity across cell types

  • Chromatin accessibility correlation:

    • Perform HIST1H1D ChIP-seq or CUT&RUN in disease models

    • Integrate with ATAC-seq or DNase-seq data to correlate HIST1H1D binding with chromatin accessibility changes

    • Compare accessibility profiles between normal and disease states

    • Identify disease-specific regulatory regions with altered HIST1H1D occupancy

  • Epigenetic mark co-localization:

    • Perform sequential ChIP with HIST1H1D (Ab-146) Antibody followed by antibodies against:

      • DNA methylation (5mC, 5hmC)

      • Histone modifications (H3K9me3, H3K27me3, H3K4me3)

      • Chromatin remodelers (BRG1, CHD4)

    • Map disease-specific changes in epigenetic landscapes

  • Therapeutic intervention studies:

    • Monitor HIST1H1D levels and PTMs during treatment with epigenetic drugs (HDAC inhibitors, DNA methyltransferase inhibitors)

    • Assess chromatin structural changes using MNase sensitivity assays

    • Correlate changes in HIST1H1D binding patterns with therapeutic response

    • Identify potential biomarkers for treatment efficacy

  • Genetic perturbation models:

    • Generate HIST1H1D knockdown/knockout in disease model systems

    • Rescue experiments with wild-type vs. mutant HIST1H1D (particularly Thr146 mutants)

    • Monitor disease phenotype progression

    • Identify genes and pathways affected by HIST1H1D alteration using RNA-seq

  • Single-cell approaches:

    • Adapt HIST1H1D (Ab-146) Antibody for single-cell protein analysis (CyTOF or imaging mass cytometry)

    • Combine with single-cell RNA-seq to correlate HIST1H1D levels with transcriptional states

    • Identify rare cell populations with altered HIST1H1D patterns in heterogeneous disease tissues

    • Track epigenetic heterogeneity in cancer progression

  • Liquid biopsy applications:

    • Develop protocols for detecting HIST1H1D in circulating nucleosomes from cancer patients

    • Correlate with disease progression or treatment response

    • Use as potential non-invasive biomarker for monitoring epigenetic changes

These methodologies leverage the specificity of the HIST1H1D (Ab-146) Antibody to provide insights into how alterations in linker histone biology contribute to disease pathogenesis and response to therapy.

How should I interpret changes in HIST1H1D localization patterns in immunofluorescence studies?

Changes in HIST1H1D localization patterns revealed by immunofluorescence using the HIST1H1D (Ab-146) Antibody can provide significant insights into cellular processes and disease states:

  • Nuclear distribution patterns:

    • Homogeneous nuclear staining: Indicates normal chromatin distribution with HIST1H1D evenly associated with chromatin

    • Peripheral nuclear localization: Often associated with heterochromatin formation at the nuclear lamina

    • Nucleolar exclusion/enrichment: Changes in nucleolar association may indicate ribosomal DNA transcription alterations

    • Punctate nuclear foci: May represent specialized chromatin domains or DNA damage sites

  • Cell cycle-related changes:

    • Prophase: Look for initial chromosome condensation with strong HIST1H1D association

    • Metaphase: HIST1H1D should show chromosome-associated patterns

    • Anaphase/telophase: Observe redistribution as chromosomes segregate

    • G1 vs S phase: Compare intensity and distribution patterns between these phases

    • Mitotic chromosome association: Reduced binding during mitosis may indicate phosphorylation-mediated dissociation

  • Stress response localization changes:

    • DNA damage: Formation of HIST1H1D foci at damage sites

    • Oxidative stress: Potential redistribution to protect specific chromatin regions

    • Heat shock: Reorganization associated with stress response gene activation

    • Hypoxia: Changes in nuclear architecture reflected in HIST1H1D patterns

  • Disease-associated patterns:

    • Cancer cells: Often show altered distribution reflecting abnormal chromatin organization

    • Senescent cells: May exhibit SAHF (senescence-associated heterochromatic foci) with HIST1H1D enrichment

    • Neurodegenerative disease models: Potential abnormal aggregation or depletion patterns

    • Viral infection: Virus-induced changes in chromatin structure reflected in HIST1H1D reorganization

  • Technical considerations for accurate interpretation:

    • Compare with DNA counterstain (DAPI/Hoechst) to normalize for chromatin distribution

    • Use Z-stack imaging to capture the full nuclear volume

    • Implement quantitative measurements (intensity, colocalization coefficients)

    • Include appropriate controls for antibody specificity

  • Quantitative analysis approaches:

    • Nuclear/cytoplasmic ratio quantification

    • Radial distribution analysis (center to periphery intensity)

    • Colocalization analysis with other chromatin marks

    • Texture analysis to quantify pattern changes (granularity, homogeneity)

When interpreting changes, consider that alterations may reflect either redistribution of existing HIST1H1D or changes in the accessibility of the epitope recognized by the antibody due to conformational changes or post-translational modifications around the Thr146 region.

What do different band patterns in Western blot reveal about HIST1H1D post-translational modifications?

Western blot analysis using the HIST1H1D (Ab-146) Antibody can reveal valuable information about post-translational modifications (PTMs) of HIST1H1D through band pattern interpretation:

  • Single band at 23 kDa:

    • Represents the unmodified or predominant form of HIST1H1D

    • Serves as the baseline for comparison with other conditions

    • Expected in most normal cellular contexts

  • Higher molecular weight bands:

    • 25-30 kDa bands: Likely phosphorylated forms, particularly relevant as the antibody targets the region around Thr146

    • ~31-32 kDa: Potentially mono-ubiquitinated HIST1H1D

    • ~35-40 kDa: Could indicate multiple phosphorylation sites or other bulky modifications

    • 40 kDa: May represent poly-ubiquitinated forms or SUMOylation

  • Lower molecular weight bands:

    • 15-20 kDa: Potential proteolytic fragments, which could be biologically relevant or sample preparation artifacts

    • 10-15 kDa: Severe degradation or specific cleavage products

  • Interpreting treatment-induced changes:

    • Cell cycle synchronization: Compare G1, S, G2/M for changes in phosphorylation status

    • Phosphatase treatment: Should eliminate bands caused by phosphorylation

    • Proteasome inhibitors: May increase ubiquitinated forms

    • HDAC inhibitors: Can alter acetylation patterns, potentially affecting mobility

  • Condition-specific patterns:

    • Stress conditions: Oxidative stress, DNA damage, or heat shock may induce specific modification patterns

    • Differentiation: Changes during cellular differentiation may reflect chromatinremodeling events

    • Disease models: Cancer cells often show altered modification patterns

  • Verification approaches:

    • Use phospho-specific antibodies in parallel (such as pThr146-specific antibodies)

    • Perform mass spectrometry to identify the exact modifications

    • Treat samples with specific enzymes (phosphatases, deubiquitinases) to confirm modification types

    • Compare with site-specific mutants (T146A, T146E) to confirm phosphorylation-dependent bands

  • Quantitative analysis:

    • Calculate the ratio of modified to unmodified forms across conditions

    • Track changes in modification patterns over time courses

    • Correlate modifications with functional outcomes

The HIST1H1D (Ab-146) Antibody is particularly valuable for studying modifications around Thr146, a key regulatory site . When interpreting Western blot results, consider that the antibody may have differential affinity for modified versus unmodified forms, potentially affecting band intensity independently of protein abundance.

How can I distinguish between different histone H1 variants in my experimental data?

Distinguishing between different histone H1 variants when using the HIST1H1D (Ab-146) Antibody requires strategic approaches due to the high sequence similarity among H1 family members:

  • Antibody-based discrimination:

    • The HIST1H1D (Ab-146) Antibody targets the region around Thr146 , which may have sequence differences from other H1 variants

    • Compare blots or staining patterns with variant-specific antibodies targeting HIST1H1A, HIST1H1B, HIST1H1C, and HIST1H1E

    • Perform peptide competition assays with peptides derived from each H1 variant to determine cross-reactivity

  • Molecular weight-based differentiation:

    • Although all H1 variants have similar molecular weights, subtle differences exist:

      • HIST1H1A: ~21.8 kDa

      • HIST1H1B: ~22.6 kDa

      • HIST1H1C: ~21.4 kDa

      • HIST1H1D: ~22.9 kDa (detected by this antibody)

      • HIST1H1E: ~22.0 kDa

    • Use high-resolution SDS-PAGE (12-15%) or gradient gels for better separation

    • Include recombinant proteins as standards for each variant

  • Genetic manipulation approaches:

    • Perform selective knockdown/knockout of HIST1H1D

    • Compare signal intensity before and after genetic manipulation

    • Express tagged variants for unambiguous identification

  • Mass spectrometry differentiation:

    • Analyze immunoprecipitated proteins by mass spectrometry

    • Identify variant-specific peptides for unambiguous assignment

    • Quantify relative abundance of each variant

  • Cell type and context considerations:

    • Different cell types express distinct patterns of H1 variants

    • HIST1H1D expression may be cell-cycle regulated

    • Literature review of expected H1 variant distribution in your model system

  • Chromatin fractionation approaches:

    • Different H1 variants may associate with distinct chromatin fractions

    • Extract histone fractions with increasing salt concentrations

    • Analyze variant distribution across fractions

  • Bioinformatic analysis of expression data:

    • Use RNA-seq or qPCR to determine which variants are expressed in your system

    • Correlate protein levels with transcript abundance

    • Consider cell type-specific expression patterns from reference databases

  • Functional discrimination:

    • Different H1 variants may respond differently to treatments

    • Compare mobility after CDK inhibitor treatment

    • Analyze distribution after stress conditions

When reporting results, clearly state the potential for cross-reactivity and the limitations of the methods used for variant discrimination, especially when making variant-specific claims about biological functions or regulation.

What can co-localization studies with HIST1H1D reveal about chromatin organization and gene regulation?

Co-localization studies using HIST1H1D (Ab-146) Antibody combined with markers of chromatin states can provide profound insights into nuclear organization and gene regulation:

  • Heterochromatin co-localization analysis:

    • Combine HIST1H1D (Ab-146) antibody with markers such as H3K9me3, H3K27me3, or HP1

    • Strong co-localization suggests HIST1H1D involvement in transcriptional repression

    • Quantify Pearson's or Mander's correlation coefficients between signals

    • Analyze changes in co-localization patterns during differentiation or disease progression

    • Compare with DNA methylation patterns using 5mC antibodies

  • Euchromatin association patterns:

    • Co-stain with H3K4me3, H3K27ac, or RNA Pol II

    • Limited co-localization is expected as HIST1H1D typically associates with compact chromatin

    • Identify potential active regions where HIST1H1D is depleted

    • Examine boundaries between active and repressed chromatin domains

    • Correlate with nascent RNA synthesis using EU incorporation

  • Nuclear compartment analysis:

    • Co-stain with nucleolar markers (fibrillarin, nucleolin)

    • Examine relationship with nuclear speckles (SC35) or PML bodies

    • Analyze lamina association (lamin B1)

    • Investigate relationship with nuclear pore complexes

    • Quantify radial distribution from the nuclear periphery to center

  • Cell cycle-dependent co-localization:

    • Analyze mitotic chromosomes (co-stain with H3S10ph)

    • Compare patterns in G1, S, and G2 phases

    • Examine changes during DNA replication (co-stain with PCNA)

    • Quantify changes in co-localization coefficients across cell cycle

  • DNA damage response:

    • Co-stain with γH2AX to identify damage sites

    • Analyze recruitment or exclusion of HIST1H1D from damage foci

    • Monitor dynamics using live-cell imaging with fluorescently tagged proteins

    • Correlate with DNA repair protein recruitment (53BP1, BRCA1)

  • Advanced imaging approaches:

    • Super-resolution microscopy (STORM, PALM) for nanoscale organization

    • FRET analysis to detect direct molecular proximity

    • Live-cell imaging to capture dynamic interactions

    • 3D image reconstruction to analyze volumetric co-localization

  • Quantitative co-localization methods:

    • Object-based co-localization for discrete structures

    • Intensity correlation analysis for continuous distributions

    • Distance-based measurements from defined nuclear landmarks

    • Machine learning approaches for pattern recognition

  • Integration with genomic data:

    • Correlate microscopy findings with ChIP-seq data for HIST1H1D

    • Compare with gene expression profiles from the same cell types

    • Relate to chromatin accessibility maps (ATAC-seq, DNase-seq)

    • Create integrated models of HIST1H1D function in genome organization

By systematically analyzing HIST1H1D co-localization with different chromatin components, researchers can build a comprehensive understanding of how this linker histone contributes to nuclear architecture and gene regulation in normal development and disease states.

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.