Formyl-HIST1H1C (K74) Antibody

Shipped with Ice Packs
In Stock

Description

Functional Role of HIST1H1C in Cellular Processes

Histone H1.2 (encoded by HIST1H1C) is a linker histone essential for chromatin compaction and transcriptional regulation. Its roles include:

  • Chromatin Fiber Formation: Binds linker DNA between nucleosomes to stabilize higher-order chromatin structures .

  • Gene Expression Modulation: Regulates transcription via chromatin remodeling and DNA methylation .

  • Immune Signaling: Modulates dendritic cell (DC) maturation by activating p38 MAPK and NF-κB pathways .

  • Autophagy Regulation: Overexpression induces autophagy via upregulation of ATG proteins (e.g., ATG12–ATG5, LC3B-II) .

Chromatin Immunoprecipitation (ChIP)

The antibody has been validated in ChIP assays to study histone modifications. For example:

  • β-Globin Promoter Analysis: Immunoprecipitation with this antibody quantified DNA enrichment at the β-globin promoter in HeLa cells treated with sodium butyrate .

  • Procedure: Cells are treated with micrococcal nuclease, sonicated, and immunoprecipitated with 5 µg antibody. DNA is analyzed via real-time PCR .

Dendritic Cell Maturation Studies

  • Key Finding: Anti-histone H1 antibodies block DC maturation by inhibiting p38 and IκBα phosphorylation, reducing CD80/CD86 expression and IL-1β/IL-6 secretion .

  • Mechanism: Histone H1 translocation from nuclei to cytoplasm is critical for DC activation and T-cell proliferation .

Autophagy in Retinal Cells

  • Overexpression Effects: HIST1H1C overexpression in rMC-1 retinal cells increases autophagy markers (ATG12–ATG5, LC3B-II) and reduces SQSTM1/p62 levels, indicating enhanced autophagic flux .

Research Implications

  • Epigenetic Regulation: Enables mapping of formyl-lysine modifications in chromatin remodeling studies .

  • Therapeutic Potential: Targeting histone H1.2 could modulate immune responses (e.g., transplant tolerance) or autophagy-related diseases .

  • Limitations: Cross-reactivity with non-histone proteins requires validation via knockout controls .

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 your order within 1-3 business days of receipt. Delivery time may vary depending on the shipping method and location. For specific delivery timelines, please consult your local distributor.
Synonyms
H1 histone family member 2 antibody; H1.a antibody; H12_HUMAN antibody; H1F2 antibody; H1s-1 antibody; HIST1H1C antibody; Histone 1 H1c antibody; Histone cluster 1 H1c antibody; Histone H1.2 antibody; Histone H1c antibody; Histone H1d antibody; Histone H1s-1 antibody; MGC3992 antibody
Target Names
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 function as regulators of individual gene transcription through chromatin remodeling, nucleosome spacing, and DNA methylation.
Gene References Into Functions
  1. Research indicates that a network of E2F target genes is susceptible to the regulatory influence of H1.2. H1.2 enhances the global association of pRb with chromatin, amplifies transcriptional repression by pRb, and facilitates pRb-dependent cell-cycle arrest. PMID: 28614707
  2. BRG1 participates in gene repression by interacting with H1.2, facilitating its deposition and stabilizing nucleosome positioning around the transcription start site. PMID: 27390128
  3. Studies have shown that histones H1.2 and H1.4 are present in MDA-MB-231 metastatic breast cancer cells. Phosphorylation at S173 of histone H1.2 and S172, S187, T18, T146, and T154 of H1.4 significantly increases during the M phase, suggesting that these events are cell cycle-dependent. The study also reports the observation of the H1.2 SNP variant A18V in MCF-10A cells. PMID: 26209608
  4. Integration with apoptotic intermediates (via C-terminal tail interactions) may represent a more generalized function of linker histone isoforms in apoptotic cascades. PMID: 24525734
  5. Histone H1.2-T165 post-translational modifications are dispensable for chromatin binding and cell proliferation, while the H1.4-K26 modifications are essential for proper cell cycle progression. PMID: 24873882
  6. H1.2 interacts with Cul4A and PAF1 to activate developmental regulatory genes. PMID: 24360965
  7. H1.2 is less abundant than other histone H1 variants at the transcription start sites of inactive genes. Promoters enriched in H1.2 differ from those enriched in other histone H1 variants and tend to be repressed. PMID: 24476918
  8. Mutations in linker histone genes HIST1H1 B, C, D, and E; OCT2 (POU2F2); IRF8; and ARID1A are implicated in the pathogenesis of follicular lymphoma. PMID: 24435047
  9. Evidence suggests that the p53 acetylation-H1.2 phosphorylation cascade serves as a unique mechanism for triggering p53-dependent DNA damage response pathways. PMID: 22249259
  10. Research has confirmed N-terminal acetylation on all isoforms, plus a single internal acetylation site. Phosphorylation sites were located on peptides containing the cyclin-dependent kinase (CDK) consensus motif. PMID: 15595731
  11. The binding of histone H1 to a general amyloid-like motif indicates that histone H1 may play a significant role in diseases associated with amyloid-like fibrils. PMID: 16854430
  12. Histone H1.2 was translocated from the nucleus to the mitochondria after treatment with bleomycin and co-localized with Bak in mitochondria. PMID: 17879944
  13. Research suggests that the recruitment of YB1, PURalpha, and H1.2 to the p53 target gene Bax is required for repression of p53-induced transcription. PMID: 18258596

Show More

Hide All

Database Links

HGNC: 4716

OMIM: 142710

KEGG: hsa:3006

STRING: 9606.ENSP00000339566

UniGene: Hs.7644

Protein Families
Histone H1/H5 family
Subcellular Location
Nucleus. Chromosome. Note=Mainly localizes in euchromatin. Distribution goes in parallel with DNA concentration.

Q&A

What is HIST1H1C and what biological roles does it play?

HIST1H1C (also known as Histone H1.2) is a linker histone variant that belongs to the H1 histone family. It functions in higher-order chromatin structure formation by binding to nucleosomes and facilitating chromatin compaction. HIST1H1C is one of several somatic H1 variants (including H1.0, H1.1, H1.2, H1.3, H1.4, H1.5, and H1X) that are expressed in various cell types .

Specifically, HIST1H1C plays critical roles in:

  • Chromatin compaction and stabilization

  • Regulation of transcriptional accessibility

  • Modulation of DNA replication and repair

  • Cell differentiation processes, particularly in the myeloid lineage

Recent studies have demonstrated that HIST1H1C and H1.4 affect neutrophil lineage determination, with their depletion reducing neutrophil differentiation and promoting eosinophil cell fate . This suggests a previously unappreciated role for specific H1 variants in immune cell development.

What is the significance of lysine formylation at position K74?

Lysine formylation at position 74 (K74) represents a specific post-translational modification of HIST1H1C that may alter its functional properties. While acetylation at this position has been more extensively studied, formylation represents a distinct modification with potentially unique biological significance .

Key points about K74 formylation include:

  • It occurs in a region that may affect DNA binding properties of the histone

  • It potentially influences interactions with chromatin remodeling complexes

  • It may represent a marker for specific cellular processes or conditions

  • It could affect the protein's stability or subcellular localization

Unlike acetylation, which is typically associated with chromatin relaxation and increased gene expression, the specific functional consequences of formylation at K74 are still under investigation in the research community.

What are the basic specifications of Formyl-HIST1H1C (K74) antibody?

The commercially available Formyl-HIST1H1C (K74) antibody has the following specifications:

ParameterSpecification
Antibody TypePrimary Antibody
ClonalityPolyclonal
HostRabbit
ReactivityHuman (Homo sapiens)
IsotypeIgG
LabelUnconjugated
ApplicationsChIP, ELISA, IF
ImmunogenPeptide sequence around site of Formyl-Lys (74) derived from Human Histone H1.2
FormLiquid
BufferPreservative: 0.03% Proclin 300; Constituents: 50% Glycerol
Purification MethodAntigen affinity purified
StorageMaintain refrigerated at 2-8°C for up to 2 weeks; long-term storage at -20°C

The antibody recognizes the formylated lysine at position 74 of HIST1H1C (UniProt ID: P16403) .

How can Formyl-HIST1H1C (K74) antibody be effectively used in Chromatin Immunoprecipitation (ChIP) assays?

ChIP is one of the validated applications for the Formyl-HIST1H1C (K74) antibody. For optimal ChIP results, researchers should consider the following methodological approach:

  • Sample preparation:

    • Use approximately 4×10^6 cells per immunoprecipitation

    • Treat samples with Micrococcal Nuclease to fragment chromatin

    • Follow with sonication to achieve optimal chromatin fragment size (200-500bp)

  • Immunoprecipitation:

    • Use 5μg of Formyl-HIST1H1C (K74) antibody per IP reaction

    • Include a control IP with normal rabbit IgG

    • Incubate chromatin-antibody mixture overnight at 4°C with rotation

    • Capture complexes using protein A/G magnetic beads

  • Washing and elution:

    • Wash complexes stringently to remove non-specific binding

    • Elute chromatin and reverse crosslinks

    • Purify DNA for downstream applications

  • Analysis methods:

    • Real-time qPCR for targeted loci analysis

    • ChIP-seq for genome-wide distribution profiling

    • Consider comparing with other histone modifications (H3K4me3, H3K9me3) to establish correlation patterns

Researchers should note that H1 histones have been reported to have complex genomic distributions, with evidence suggesting both broad genome-wide presence and specific enrichment patterns .

How does HIST1H1C distribution compare with other H1 variants across the genome?

Research on H1 variant distribution has revealed interesting patterns that may inform experimental design and data interpretation when using Formyl-HIST1H1C (K74) antibody:

  • General distribution patterns:

    • All H1 variants occur across the genome, but with variant-specific features

    • H1.2 (HIST1H1C) shows distinctive distribution patterns at promoters and genome-wide

    • H1 variants show differential enrichment at specific genomic features

  • Correlation with chromatin features:

    • H1 variants show positive correlation with H3K9me3 (repressive mark)

    • Negative correlation with H3K4me3 (active mark)

    • Differential enrichment at GC- and gene-rich regions

    • Variable presence at active promoters

  • Cell-type specificity:

    • Distribution patterns may differ between cell types

    • H1.5 shows zones of enrichment in differentiated cells but not in embryonic stem cells

    • These patterns associate with gene repression and chromatin compaction

Understanding these distribution patterns is critical when designing ChIP experiments and interpreting data obtained with the Formyl-HIST1H1C (K74) antibody.

What are the functional differences between histone H1 variants and how might formylation affect their roles?

Different H1 variants exhibit distinct functional properties despite their structural similarities:

H1 VariantFunctional CharacteristicsSpecific Roles
H1.2 (HIST1H1C)Critical for neutrophil differentiationAffects lineage determination in granulopoiesis
H1.4Similar to H1.2 in neutrophil differentiationAffects lineage determination in granulopoiesis
H1.1, H1.3, H1.5Opposite effects to H1.2/H1.4Accelerate maturation when depleted
H1.0Specific role in dendritic cellsFewer dendritic cells but normal granulocytes in H1.0-deficient mice

H1.2 and H1.4 deficiency has been shown to:

  • Reduce expression of neutrophil differentiation markers (e.g., CD11b)

  • Promote unrestricted cell growth

  • Increase cell viability during differentiation

  • Upregulate eosinophil genes

  • Shift bone marrow stem cell differentiation from neutrophil to eosinophil fate

Formylation at K74 could potentially modulate these functions by altering:

  • Chromatin binding affinity

  • Interactions with transcription factors (e.g., GATA-2)

  • Residence time on chromatin

  • Recruitment of chromatin modifiers

How can researchers distinguish between different modifications at the K74 position?

Researchers often need to differentiate between various modifications at the same position (K74), which can include acetylation, methylation, and formylation. Methodological approaches include:

  • Antibody specificity validation:

    • Western blotting with modified and unmodified peptides

    • Competition assays with specific modified peptides

    • Dot blot analysis with a panel of modified histone peptides

    • Analysis with mass spectrometry to confirm modification identity

  • Parallel ChIP experiments:

    • Perform ChIP with antibodies against different modifications at K74

    • Compare genomic distribution patterns

    • Identify regions of unique or overlapping enrichment

  • Functional validation:

    • Use site-specific mutants (K74R, K74Q) to mimic or prevent modification

    • Employ mass spectrometry to quantify modification abundance

    • Analyze effects of enzymatic inhibitors that affect specific modifications

When comparing Formyl-HIST1H1C (K74) with Acetyl-HIST1H1C (K74), researchers should be aware that both modifications occur at the same residue but may have distinct functional consequences and genomic distribution patterns .

What controls should be included when using Formyl-HIST1H1C (K74) antibody in experimental workflows?

Robust experimental design requires appropriate controls. For experiments using Formyl-HIST1H1C (K74) antibody, consider:

  • Negative controls:

    • Isotype control (normal rabbit IgG) for immunoprecipitation

    • No-antibody control in immunoprecipitation

    • Peptide competition with unmodified K74 peptide

    • Samples from HIST1H1C knockout or knockdown cells

  • Positive controls:

    • Known targets or genomic regions enriched for this modification

    • Nuclear extracts with confirmed presence of formylated HIST1H1C

    • Synthetic peptides containing formyl-K74 for antibody validation

  • Experimental validation controls:

    • Parallel ChIP with antibodies against total HIST1H1C

    • ChIP with antibodies against associated histone marks

    • Comparison with histone H3 distribution to normalize for nucleosome occupancy

    • Replicate experiments under identical conditions to assess reproducibility

How can ChIP-seq data for Formyl-HIST1H1C (K74) be properly analyzed and interpreted?

Analysis of ChIP-seq data for histone modifications requires specialized approaches:

  • Data processing pipeline:

    • Quality control and read filtering

    • Alignment to reference genome

    • Peak calling using appropriate algorithms for histone modifications

    • Input subtraction to remove background signal

    • Normalization for sequencing depth

  • Analytical approaches:

    • Calculate normalized average read density in enriched locations

    • Generate genome browser tracks for visualization

    • Perform statistical tests (e.g., Kolmogorov-Smirnov) to assess significance

    • Compare with random genomic windows of equal width as controls

  • Correlation with genomic features:

    • Analyze enrichment at regulatory regions

    • Examine correlation with other histone modification peaks

    • Assess association with CpG islands and lamina-associated domains (LADs)

    • Integrate with gene expression data to identify functional correlations

  • Comparative analysis:

    • Compare distribution with other H1 variants

    • Analyze differential enrichment between cell types or conditions

    • Correlate with transcription factor binding sites

How can Formyl-HIST1H1C (K74) antibody be used to study cellular differentiation processes?

Given the role of HIST1H1C in cellular differentiation, particularly in the neutrophil lineage, this antibody can be valuable for studying differentiation processes:

  • Differentiation models:

    • Use PLB-985 cell line as a neutrophil differentiation model

    • Monitor HIST1H1C formylation during myeloid differentiation

    • Compare with other cell lines (e.g., THP-1 for monocytic differentiation)

  • Methodological approach:

    • Track formylation levels during differentiation time course

    • Perform ChIP-seq at different differentiation stages

    • Correlate formylation patterns with expression of differentiation markers (e.g., CD11b)

    • Compare wild-type cells with H1.2/H1.4-deficient cells

  • Functional readouts:

    • Cell proliferation and viability measurements

    • Surface marker expression (flow cytometry)

    • Functional assays (e.g., oxidative burst, NET formation)

    • Gene expression profiling by RNA-seq

Studies have shown that H1.2 and H1.4 deficiency affects differentiation trajectories, with cells showing:

  • Decreased CD11b expression

  • Unrestricted growth until day 7 of differentiation

  • Enhanced viability compared to controls

  • Altered gene expression profiles favoring eosinophil rather than neutrophil fate

What is the relevance of studying Formyl-HIST1H1C (K74) in cancer research?

Histone H1 variants have been differentially implicated in cancer processes, making Formyl-HIST1H1C (K74) potentially relevant for cancer research:

  • Cancer-related applications:

    • Map formylation patterns in breast cancer cell lines

    • Compare formylation levels between normal and cancer cells

    • Correlate formylation with gene expression changes in cancer

    • Study the impact of formylation on cancer cell differentiation state

  • Experimental approaches:

    • ChIP-seq in cancer cell lines (e.g., T47D, MCF7)

    • Correlation with transcriptomic data

    • Integration with other epigenetic marks

    • Functional studies using genetic manipulation (CRISPR/Cas9)

  • Potential significance:

    • Formylation may mark specific regulatory regions in cancer cells

    • Changes in formylation patterns might correlate with cancer progression

    • H1 variant distribution differs between differentiated cells and stem-like cells

    • Differential regulation of H1 variants occurs during cancer development

What are common challenges when working with histone H1 antibodies and how can they be addressed?

Working with H1 histone antibodies presents unique challenges:

  • ChIP efficiency issues:

    • H1 histones can be more difficult to immunoprecipitate than core histones

    • Solution: Optimize crosslinking conditions; try dual crosslinking with DSG and formaldehyde

    • Solution: Adjust sonication conditions to improve chromatin accessibility

  • Specificity concerns:

    • Cross-reactivity between H1 variants due to sequence similarity

    • Solution: Validate specificity using peptide competition assays

    • Solution: Include appropriate controls (H1 variant knockout/knockdown)

  • Signal-to-noise ratio:

    • High background in ChIP experiments

    • Solution: Increase wash stringency during immunoprecipitation

    • Solution: Optimize antibody concentration

    • Solution: Pre-clear chromatin more thoroughly

  • Reproducibility issues:

    • Variation between experimental replicates

    • Solution: Standardize cell culture conditions and harvest procedures

    • Solution: Maintain consistent chromatin fragmentation size

    • Solution: Use internal normalization controls

How do experimental conditions affect the detection of formylated HIST1H1C?

The detection of histone formylation can be affected by various experimental factors:

  • Cell culture conditions:

    • Serum levels can affect histone modification patterns

    • Cell density impacts histone modifications

    • Hypoxia can influence formylation levels

    • Recommendation: Standardize culture conditions across experiments

  • Sample preparation:

    • Formyl modifications may be less stable than other modifications

    • Oxidative conditions during preparation may alter modification patterns

    • Recommendation: Add protease and deacetylase inhibitors to buffers

    • Recommendation: Process samples quickly and maintain cold temperatures

  • Fixation parameters:

    • Overfixation can mask epitopes

    • Insufficient fixation leads to poor chromatin recovery

    • Recommendation: Optimize formaldehyde concentration (typically 1%) and fixation time (10-15 minutes)

    • Recommendation: Consider dual crosslinking approaches for improved H1 recovery

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.