HIST1H1C Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze / thaw cycles.
Lead Time
Typically, we can ship your order within 1-3 business days of receiving it. Delivery time may vary depending on the purchasing method or location. Please consult your local distributor for specific delivery timelines.
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 is known to bind to the linker DNA between nucleosomes, playing a crucial role in forming the macromolecular structure of the chromatin fiber. These histones are essential for the condensation of nucleosome chains into higher-order structured fibers. Additionally, they act as regulators of individual gene transcription through chromatin remodeling, nucleosome spacing, and DNA methylation.
Gene References Into Functions
  1. Research has identified a network of E2F target genes that are susceptible to the regulatory influence of H1.2. This variant enhances the global association of pRb with chromatin, leading to increased transcriptional repression by pRb and facilitated 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 revealed the presence of histones H1.2 and H1.4 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 of the cell cycle, suggesting a cell cycle-dependent nature of these events. Moreover, the study 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 broader function of linker histone isoforms in apoptotic cascades. PMID: 24525734
  5. Post-translational modifications of histone H1.2-T165 are dispensable for chromatin binding and cell proliferation, while 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 have been 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, along with a single internal acetylation site. Phosphorylation sites have been identified 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 has been observed to translocate from the nucleus to the mitochondria after treatment with bleomycin and co-localize with Bak in mitochondria. PMID: 17879944
  13. Research has demonstrated that the recruitment of YB1, PURalpha, and H1.2 to the p53 target gene Bax is necessary for repressing 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 the functional significance of HIST1H1C in chromatin structure and immune regulation?

HIST1H1C is a somatic linker histone variant that stabilizes nucleosomal structure and contributes to higher-order chromatin folding. Beyond structural roles, HIST1H1C has emerged as an important regulator of innate immunity. HIST1H1C significantly upregulates interferon-β (IFN-β) production, with its post-translational modifications playing critical regulatory roles . Specifically:

  • HIST1H1C interacts with the influenza virus NS2 protein via its C-terminal domain in the nucleus

  • The histone's phosphorylation mutant (T146A) decreases IFN-β production

  • Methylation mutants (K34A, K187A) increase IFN-β by releasing nucleosome constraints and promoting IRF3 binding to the IFN-β promoter

  • Influenza virus NS2 protein interacts with HIST1H1C, reducing H1C-IRF3 interaction and inhibiting IFN-β production

This demonstrates HIST1H1C's dual role in both structural genomic organization and epigenetic regulation of immune responses.

How should researchers select the appropriate HIST1H1C antibody for their specific experimental applications?

Selection of HIST1H1C antibodies requires careful consideration of several technical factors:

ApplicationRecommended Antibody CharacteristicsValidation Methods
Western BlottingPolyclonal antibodies targeting middle regionCell lysate positive controls, knockout cell validation
ChIPHigh-specificity antibodies, validated for chromatin bindingChIP-qPCR with known targets
ImmunofluorescenceNon-phospho-specific antibodies for general localizationCSK buffer treatment, co-localization studies
PTM StudiesModification-specific antibodies (e.g., acLys96, meLys45)Peptide competition assays

When selecting antibodies, researchers should consider:

  • Target epitope region (N-terminal, C-terminal, or middle region)

  • Post-translational modification status

  • Cross-reactivity with other H1 variants (sequence homology 74-87%)

  • Validated applications and species reactivity

For highest specificity, choose antibodies targeting the amino or carboxy termini, as these regions show greatest sequence divergence between H1 variants .

What experimental controls are essential when using HIST1H1C antibodies in immunological techniques?

Proper experimental design for HIST1H1C antibody usage requires rigorous controls:

Essential Controls for HIST1H1C Antibody Experiments:

  • HIST1H1C knockout cell lines (e.g., A549-H1C-KO cells) serve as negative controls to validate antibody specificity

  • Peptide competition assays using the immunizing peptide

  • Isotype control antibodies to assess non-specific binding

  • Multiple antibodies targeting different HIST1H1C epitopes to confirm findings

  • siRNA knockdown controls to verify signal reduction

  • Positive controls using cells known to express HIST1H1C

For ChIP experiments specifically, additional controls include:

  • IgG negative controls

  • RNA polymerase II antibody as positive control for active promoters

  • Input DNA normalization

  • Known target regions for validation (e.g., IFN-β promoter)

Without these controls, results may be compromised by cross-reactivity with other H1 variants due to high sequence homology.

How can researchers validate the specificity of commercial HIST1H1C antibodies?

Commercial HIST1H1C antibodies require thorough validation before use in critical experiments. Recommended validation protocols include:

  • Western blot analysis:

    • Compare wild-type and HIST1H1C knockout cell lysates

    • Verify single band at expected molecular weight (~30 kDa)

    • Test cross-reactivity with recombinant H1 variants

  • Immunoprecipitation validation:

    • Perform IP followed by mass spectrometry identification

    • Confirm enrichment of HIST1H1C peptides

    • Verify absence of peptides unique to other H1 variants

  • Chromatin immunoprecipitation verification:

    • ChIP-qPCR at known HIST1H1C binding sites

    • Verify enrichment patterns match published data

    • Compare with ChIP using alternative HIST1H1C antibodies

  • Knockout/knockdown validation:

    • CRISPR/Cas9 knockout cells (example: A549-H1C-KO cells)

    • siRNA knockdown verification

    • Signal reduction following target depletion

One validated commercial option is antibody ab4086, which shows specificity for HIST1H1C in wild-type A549 cells, with signal loss in HIST1H1C knockout cell lines .

What are the common technical challenges when working with HIST1H1C antibodies and how can they be addressed?

Researchers face several technical challenges when working with HIST1H1C antibodies:

ChallengeCauseSolution
Cross-reactivityHigh sequence homology (74-87%) among H1 variants Use antibodies targeting divergent N/C-terminal regions
Epitope maskingAbundant PTMs on terminal tailsSelect antibodies recognizing unmodified regions or specific PTM states
Inconsistent resultsBatch-to-batch variabilityValidate each lot, maintain reference samples
Background signalNon-specific bindingOptimize blocking, increase washing stringency
Poor chromatin accessibilityCompact chromatin structureInclude appropriate chromatin fragmentation and extraction steps

Additional technical considerations:

  • For immunofluorescence studies, CSK buffer treatment helps distinguish chromatin-bound from soluble HIST1H1C

  • For ChIP applications, using high pressure antigen retrieval with 10 mM citrate buffer (pH 6.0) can improve epitope accessibility

  • Mass spectrometry offers an antibody-independent alternative for analyzing HIST1H1C variants and their PTMs

How can researchers effectively study post-translational modifications of HIST1H1C using specific antibodies?

HIST1H1C undergoes extensive post-translational modifications that regulate its function. Studying these PTMs requires specialized approaches:

Methodological Workflow for HIST1H1C PTM Analysis:

  • PTM-specific antibody selection:

    • Validated modification-specific antibodies for HIST1H1C include:

      • Anti-phospho-Thr146 (regulates chromatin condensation during mitosis)

      • Anti-methylLys34 and anti-methylLys187 (enhance IFN-β production)

      • Anti-acetylLys96 (alters chromatin binding properties)

  • Sample preparation considerations:

    • Utilize phosphatase inhibitors for phosphorylation studies

    • Include deacetylase inhibitors (e.g., sodium butyrate) for acetylation studies

    • Extract histones using acid extraction to maintain PTM integrity

  • Validation approaches:

    • Compare wild-type HIST1H1C with PTM site mutants (e.g., K34A, K187A, T146A)

    • Combine with mass spectrometry for comprehensive PTM profiling

    • Use phosphatase/deacetylase treatments as negative controls

  • Functional analysis:

    • ChIP-seq with PTM-specific antibodies to map genomic distribution

    • Correlate with transcriptional outcomes through RNA-seq

    • Assess impact on protein interactions through Co-IP experiments

Research shows that HIST1H1C phosphorylation and methylation states significantly impact interferon responses, with K34A and K187A methylation mutants enhancing IFN-β production while T146A phosphorylation mutants decrease it .

What are the most effective protocols for studying HIST1H1C interactions with viral proteins such as influenza NS2?

Studying HIST1H1C interactions with viral proteins requires multifaceted approaches:

Recommended Protocol for HIST1H1C-Viral Protein Interaction Studies:

  • Co-Immunoprecipitation (Co-IP):

    • Infect A549 cells with influenza virus (MOI=10)

    • Lyse cells at 10h post-infection

    • Perform Co-IP using NS2 polyclonal antibody

    • Analyze by SDS-PAGE and western blotting

  • Recombinant protein expression for validation:

    • Co-transfect HEK293T cells with Flag-H1C and HA-NS2

    • Perform reciprocal Co-IP with anti-HA and anti-Flag antibodies

    • Validate interaction domain using HIST1H1C truncation mutants

  • Subcellular localization studies:

    • Extract subcellular fractions using differential centrifugation

    • Analyze nuclear vs. cytoplasmic distribution

    • Perform immunofluorescence to visualize co-localization

  • Functional impact assessment:

    • Compare wild-type and HIST1H1C knockout cells for viral replication

    • Measure IFN-β production and IRF3 activity

    • Analyze how NS2 affects HIST1H1C-IRF3 interaction

These approaches have revealed that NS2 interacts with HIST1H1C's C-terminal domain, reducing H1C-IRF3 interaction and inhibiting IFN-β production, suggesting a novel mechanism for influenza virus to antagonize innate immune responses .

How should researchers design and interpret ChIP-seq experiments for genome-wide mapping of HIST1H1C binding sites?

ChIP-seq for HIST1H1C requires specialized protocols due to linker histone dynamics and chromatin structure:

HIST1H1C ChIP-seq Optimization Protocol:

  • Cross-linking optimization:

    • Dual cross-linking with 1.5 mM EGS followed by 1% formaldehyde

    • Cross-linking time must be optimized (typically 10-15 minutes)

    • Quench with 125 mM glycine

  • Chromatin preparation:

    • Sonicate to 200-500 bp fragments

    • Verify fragmentation by agarose gel electrophoresis

    • Pre-clear with protein A/G beads to reduce background

  • Immunoprecipitation:

    • Use validated ChIP-grade HIST1H1C antibodies

    • Include IgG control and input samples

    • Wash stringently to reduce background

  • Library preparation and sequencing:

    • Prepare libraries with adapters for paired-end sequencing

    • Sequence at >30 million reads per sample

    • Include spike-in controls for normalization

  • Bioinformatic analysis:

    • Peak calling algorithms: MACS2 with broad peak settings

    • Analyze distribution relative to genomic features

    • Compare with histone modification marks (H3K9me3, H3K4me3)

Genome-wide mapping studies have revealed that HIST1H1C is depleted from GC- and gene-rich regions and active promoters, with positive correlation with H3K9me3 and negative correlation with H3K4me3, as well as overrepresentation in major satellites . This contrasts with H1X, which is enriched at gene-rich chromosomes and RNA polymerase II-enriched regions .

What experimental approaches effectively demonstrate the functional impact of HIST1H1C on viral replication and interferon response?

To establish HIST1H1C's role in viral replication and interferon response, researchers should implement a comprehensive experimental strategy:

Experimental Design for Functional Studies:

  • HIST1H1C manipulation models:

    • CRISPR/Cas9 knockout cell lines (A549-H1C-KO)

    • siRNA knockdown (transient depletion)

    • Overexpression of wild-type and mutant HIST1H1C proteins (K34A, K187A, T146A)

  • Viral infection assessment:

    • Measure virus replication by plaque assay

    • Quantify viral NP mRNA by real-time PCR

    • Detect viral protein expression by western blotting

    • Generate viral growth curves at multiple timepoints (12, 24, 36, 48h)

  • Interferon response characterization:

    • Quantify IFN-β mRNA by RT-PCR

    • Measure additional cytokines (TNF-α, CXCL10)

    • Analyze downstream ISG expression (MX1, OASL)

    • Stimulate with synthetic IFN inducers (poly(I:C)) to assess pathway integrity

  • Mechanistic investigation:

    • Perform ChIP-qPCR for IRF3 binding to IFN-β promoter

    • Analyze H1C-IRF3 interaction by Co-IP

    • Examine impact of viral NS2 on these interactions

Research using these approaches has demonstrated that:

  • Influenza virus replicates more efficiently in H1C-KO cells compared to wild-type

  • HIST1H1C overexpression inhibits viral replication

  • K34A and K187A mutations enhance HIST1H1C's antiviral activity

  • T146A mutation reduces this inhibitory effect

  • HIST1H1C significantly upregulates IFN-β and TNF-α production

How can researchers effectively generate and validate HIST1H1C knockout cell lines for functional studies?

Generation of HIST1H1C knockout cell lines requires careful design and validation:

Protocol for HIST1H1C Knockout Generation:

  • CRISPR/Cas9 design:

    • Target guide sequence (example): 5'-AACCAATGTCACCGGCGCCGGCC-3'

    • Design multiple guide RNAs targeting early exons

    • Clone into appropriate CRISPR/Cas9 vectors (e.g., px335-H1C plasmid)

  • Cell transfection and selection:

    • Transfect target cells (e.g., A549)

    • Culture for 2 days, repeat transfection

    • Perform single-cell dilution and expansion

    • Screen clones for knockout

  • Validation methods:

    • Genomic PCR and sequencing to confirm mutations

    • Western blotting to verify protein absence

    • RNA-seq to confirm transcript disruption

    • Functional rescue experiments with wild-type HIST1H1C

  • Characterization:

    • Compare cell proliferation and morphology

    • Analyze chromatin structure

    • Assess response to stimuli (viral infection, IFN inducers)

    • Document any compensatory changes in other H1 variants

Commercial knockout cell lines like the A549-H1C-KO (validated by Next Generation Sequencing and Western blot) can serve as alternatives to in-house generation . Studies using HIST1H1C knockout cells have revealed enhanced viral replication, demonstrating the histone's role in antiviral defense mechanisms .

How does HIST1H1C contribute to epigenetic regulation during cellular senescence and aging?

HIST1H1C plays a complex role in cellular senescence and aging processes:

Research Approaches for Studying HIST1H1C in Senescence:

  • Senescence models:

    • Replicative senescence in fibroblasts

    • Oncogene-induced senescence

    • DNA damage-induced senescence

    • Premature aging syndromes associated with chromatin defects

  • HIST1H1C dynamics analysis:

    • Monitor changes in HIST1H1C expression during senescence

    • Analyze post-translational modifications associated with aging

    • Map genomic redistribution using ChIP-seq

    • Compare with other H1 variants (particularly H1.0)

  • Functional studies:

    • Manipulate HIST1H1C levels in senescent cells

    • Assess impact on senescence markers (SA-β-gal, p16, p21)

    • Analyze chromatin compaction and accessibility

    • Evaluate effects on senescence-associated secretory phenotype (SASP)

Research on the related H1 variant HIST1H1E has shown that mutations in its C-terminal tail cause accelerated senescence and premature aging . Cells expressing these mutant proteins demonstrate:

  • Dramatically reduced proliferation

  • Difficulty entering S phase

  • Accelerated senescence

  • Association with premature aging phenotypes

These findings suggest that proper linker histone function is critical for preventing premature cellular aging, with implications for understanding age-related diseases.

What are the current technical innovations and emerging trends in studying HIST1H1C variant-specific functions in chromatin regulation?

Recent technological advances are transforming HIST1H1C research:

Emerging Approaches in HIST1H1C Research:

  • Advanced imaging techniques:

    • Super-resolution microscopy to visualize HIST1H1C distribution

    • Live-cell imaging with fluorescently tagged HIST1H1C variants

    • Single-molecule tracking to analyze dynamics and residence time

    • FRAP (Fluorescence Recovery After Photobleaching) to measure mobility

  • Genome editing and functional genomics:

    • CRISPR/Cas9 for precise modification of endogenous HIST1H1C

    • Homology-directed repair to introduce specific mutations (K34A, K187A, T146A)

    • CUT&RUN and CUT&Tag as alternatives to traditional ChIP

    • CRISPR screens to identify HIST1H1C functional pathways

  • Combinatorial epigenomic approaches:

    • Multi-omics integration (ChIP-seq, ATAC-seq, RNA-seq)

    • Single-cell analyses to capture heterogeneity

    • Proteomics to identify HIST1H1C interactome

    • Correlation between HIST1H1C binding and 3D chromatin structure

Recent work with endogenously tagged or antibody-detected H1 variants has revealed distinct genomic distributions for different variants, with H1X enriched at active regions while HIST1H1C is predominantly associated with repressed chromatin . Emerging research suggests HIST1H1C depletion, particularly when combined with H1.4 knockdown, triggers interferon responses via activation of heterochromatic repeats, indicating its role in maintaining cell homeostasis .

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.