HIST1H1C (Ab-96) Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Overview

Target: Histone H1.2 (encoded by HIST1H1C gene)
Epitope: Peptide sequence surrounding lysine residue 96 (K96) in human histone H1.2 .
Host Species: Rabbit .
Clonality: Polyclonal .
Isotype: IgG .

Table 1: Key Properties

PropertyDetails
ApplicationsELISA, Western Blot (WB), Immunohistochemistry (IHC), Chromatin Immunoprecipitation (ChIP), Immunofluorescence (IF) .
Species ReactivityHuman (Homo sapiens) .
ImmunogenSynthetic peptide corresponding to residues near K96 of human H1.2 .
Accession NumberUniProt: P16403 .
ValidationVerified for specificity in WB, IHC, and IF .

Recommended Dilutions

ApplicationDilution Range
WB1:500 – 1:2000 .
IHC1:20 – 1:200 .
IFManufacturer-specified protocols .

Research Use Cases

  • Chromatin Studies: Used to investigate histone H1.2’s role in autophagy, inflammation, and diabetic retinopathy .

  • Viral Infection Research: Validated in studies exploring H1.2’s interaction with viral replication mechanisms (e.g., EMCV inhibition via MDA5 activation) .

  • Dendritic Cell Maturation: Applied to study histone H1.2’s impact on MAPK/NF-κB signaling in immune cells .

Validation and Quality Control

  • Specificity: Recognizes endogenous histone H1.2 without cross-reactivity to other H1 variants (e.g., H1.0, H1.3, H1.4) .

  • Batch Consistency: Produced under ISO 9001-certified conditions .

  • Functional Assays:

    • Confirmed reduced H1.2 expression in siRNA-treated retinal cells .

    • Demonstrated cytoplasmic/nuclear localization in dendritic cells under stress .

Role in Diabetic Retinopathy

  • HIST1H1C (Ab-96) detected upregulated H1.2 in diabetic rodent retinas, correlating with autophagy activation and neuronal damage .

  • Knockdown of H1.2 via siRNA reduced retinal inflammation and glial activation .

Antiviral Activity

  • Overexpression of H1.2 enhanced MDA5-mediated interferon production, inhibiting EMCV replication .

Immune Regulation

  • Blocking H1.2 with this antibody suppressed dendritic cell maturation and T-cell proliferation .

Limitations

  • Restrictions: For research use only; not approved for diagnostics .

  • Species Limitation: Reactivity confirmed only in humans .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch products within 1-3 business days after receiving your order. Delivery times may vary depending on the method of purchase and location. Please contact your local distributor for specific delivery timeframes.
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 condensing 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 a network of E2F target genes is susceptible to regulation by 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, promoting 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 these events are cell cycle-dependent. Additionally, a study reports the observation of the H1.2 SNP variant A18V in MCF-10A cells. PMID: 26209608
  4. The integration of linker histone isoforms with apoptotic intermediates (via C-terminal tail interactions) may represent a generalized function in apoptotic cascades. PMID: 24525734
  5. Histone H1.2-T165 post-translational modifications are dispensable for chromatin binding and cell proliferation, whereas 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 linked to the pathogenesis of follicular lymphoma. PMID: 24435047
  9. Evidence suggests that the p53 acetylation-H1.2 phosphorylation cascade serves as a distinct 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 suggests that histone H1 may play a common 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. Studies have shown 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 are its primary cellular functions?

HIST1H1C (H1.2) is one of seven linker histone H1 variants present in human somatic cells with distinct prevalence across cell types. It serves as a key structural component of chromatin, binding to the nucleosome and facilitating higher-order chromatin structure . Beyond its structural role, HIST1H1C has recently been discovered to have regulatory functions in immune responses, particularly in the regulation of interferon-β (IFN-β) production, and it plays a significant role in autophagy regulation in certain disease conditions like diabetic retinopathy . HIST1H1C is primarily localized in the nucleus and is involved in nucleosome assembly and DNA binding as indicated by its GO terms .

How does HIST1H1C differ from other H1 histone variants?

While all H1 variants share the common function of binding to nucleosomes, HIST1H1C displays specific genomic distribution patterns. Studies using chromatin immunoprecipitation (ChIP) combined with quantitative PCR, tiling promoter arrays, and high-resolution sequencing have revealed that HIST1H1C (H1.2) has specific features both at promoters and genome-wide that distinguish it from other H1 variants such as H1.0, H1.3, H1.4, H1.5, and H1X . These distribution patterns suggest specific regulatory roles for HIST1H1C in gene expression that may differ from other H1 variants. The specificity of HIST1H1C function is also demonstrated by its unique interactions with viral proteins and cellular factors during immune responses that aren't observed with other H1 variants .

What are the most widely used applications for HIST1H1C antibodies in research?

HIST1H1C antibodies are primarily used for:

  • Western blotting - detecting HIST1H1C protein expression levels (typically recognizing a band of approximately 30 kDa)

  • Chromatin immunoprecipitation (ChIP) - studying genome-wide distribution and binding patterns

  • Immunofluorescence - examining cellular localization

  • Protein-protein interaction studies - investigating binding partners such as IRF3 and viral proteins like influenza NS2

  • Epigenetic research - exploring how HIST1H1C affects gene expression through chromatin modification

These applications are essential for understanding both the structural role of HIST1H1C in chromatin organization and its emerging functional roles in cellular processes.

What are the optimal conditions for using HIST1H1C antibodies in western blotting?

For optimal western blotting with HIST1H1C antibodies, researchers should follow these methodological guidelines:

  • Sample preparation: Use standard cell lysis buffers containing protease inhibitors; nuclear extraction protocols often yield better results for histone proteins

  • Protein loading: Load 20-30 μg of total protein per lane

  • Gel selection: Use 12-15% SDS-PAGE gels for optimal resolution of histone proteins

  • Transfer conditions: Transfer to PVDF membranes at lower voltage (30V) overnight at 4°C for better results with histone proteins

  • Antibody dilution: Use anti-HIST1H1C antibody at a dilution of 1:1000 as recommended for PrecisionAb polyclonal antibodies

  • Secondary antibody: Use goat anti-rabbit IgG (H/L):HRP conjugate (such as STAR208P) for detection

  • Expected results: Look for a specific band at approximately 30 kDa in cellular lysates such as Jurkat cells

For validation, positive controls like Jurkat cell lysates where HIST1H1C is known to be expressed should be included, while knockout or knockdown samples serve as negative controls to confirm antibody specificity.

How can researchers effectively perform HIST1H1C chromatin immunoprecipitation (ChIP) experiments?

For successful HIST1H1C ChIP experiments, researchers should:

  • Crosslinking: Use 1% formaldehyde for 10 minutes at room temperature to preserve protein-DNA interactions

  • Sonication: Optimize sonication conditions to generate DNA fragments of 200-500 bp

  • Antibody validation: Prior to full experiments, validate antibody specificity using western blotting and immunoprecipitation

  • Immunoprecipitation: Use 2-5 μg of HIST1H1C-specific antibody per ChIP reaction (alternatively, for tagged variants, use HA-tag antibodies with recombinant HA-tagged HIST1H1C)

  • Controls: Include IgG control, input control, and positive control regions (known HIST1H1C binding sites)

  • Analysis methods: Combine with qPCR for targeted analysis, or with sequencing for genome-wide distribution assessment

Studies have successfully used this approach to map HIST1H1C distribution across the genome and at specific promoters, enabling comparison with other histone variants and nucleosome positioning through parallel H3 ChIP experiments .

What are the best approaches for studying HIST1H1C post-translational modifications?

HIST1H1C undergoes critical post-translational modifications (PTMs) that affect its function, particularly in immune regulation. To study these PTMs:

  • Mass spectrometry approaches:

    • Use enrichment procedures specific for phosphorylated or methylated peptides

    • Apply high-resolution mass spectrometry to identify specific modification sites

  • Site-directed mutagenesis:

    • Generate point mutations at key modification sites (e.g., T146A phosphorylation mutant or K34A, K187A methylation mutants)

    • Express these mutants in relevant cell systems to assess functional impacts

  • Modification-specific antibodies:

    • Use antibodies targeting specific modifications like phosphorylated T146 or methylated K34/K187

    • Apply in western blotting or ChIP to assess modification status in different conditions

  • Functional analysis:

    • Compare wild-type HIST1H1C with modification-site mutants in functional assays

    • Research has shown that the phosphorylation mutant (T146A) decreases IFN-β, while methylation mutants (K34A, K187A) increase IFN-β by altering nucleosome dynamics and IRF3 binding

This methodological approach has revealed how different modifications on HIST1H1C regulate critical functions like interferon responses during viral infection.

How does HIST1H1C contribute to antiviral immune responses?

HIST1H1C plays a significant role in antiviral immunity through several mechanisms:

  • Regulation of IFN-β: HIST1H1C enhances IFN-β production, a critical antiviral cytokine. Research shows it accomplishes this by facilitating IRF3 binding to the IFN-β promoter .

  • Influenza virus antagonism: Studies demonstrate that influenza virus NS2 protein specifically targets HIST1H1C, interacting with its C-terminal region in the nucleus. This interaction reduces HIST1H1C-IRF3 binding, thereby inhibiting the IFN-β enhancement mediated by HIST1H1C .

  • Modification-dependent regulation:

    • Phosphorylation at T146 reduces HIST1H1C's ability to enhance IFN-β

    • Methylation at K34 and K187 increases HIST1H1C's enhancement of IFN-β by releasing nucleosome constraints

  • Impact on viral replication: Experimental evidence shows that:

    • H1N1 influenza virus replicates more efficiently in HIST1H1C knockout A549 cells compared to wild-type cells

    • Overexpression of HIST1H1C significantly reduces viral nucleoprotein expression and viral titers

    • Modification mutants (K34A, K187A) enhance this antiviral effect

These findings reveal HIST1H1C as a previously unrecognized component of the innate immune system that viruses have evolved mechanisms to counteract.

What is the role of HIST1H1C in autophagy regulation and disease pathogenesis?

HIST1H1C has been identified as a critical regulator of autophagy in pathological conditions, particularly in diabetic retinopathy:

  • Autophagy induction: Overexpression of HIST1H1C upregulates SIRT1 and HDAC1, maintaining the deacetylation status of H4K16, which leads to upregulation of ATG proteins and promotion of autophagy in retinal cells .

  • Inflammatory pathway activation: HIST1H1C overexpression promotes inflammation through:

    • Increased production of TNF-α

    • Enhanced expression of chemokines like CXCL10

    • These inflammatory mediators contribute to tissue damage

  • Disease progression mechanisms:

    • In diabetic retinopathy models, both HIST1H1C and autophagy proteins are upregulated in the retinas of type 1 diabetic rodents

    • AAV-mediated HIST1H1C overexpression in retinas leads to increased autophagy, inflammation, glial activation, and neuron loss – pathological changes similar to early diabetic retinopathy

    • Knockdown of HIST1H1C in diabetic mice significantly attenuates these pathological changes

  • Therapeutic potential: The research suggests that targeting HIST1H1C could offer a novel therapeutic approach for preventing diabetic retinopathy by modulating excessive autophagy and inflammation .

This represents an emerging area where histone variants like HIST1H1C play direct roles in disease pathogenesis beyond their classical chromatin functions.

How can HIST1H1C knockout or knockdown models be effectively generated and validated?

Creating reliable HIST1H1C knockout or knockdown models requires careful methodological considerations:

  • CRISPR/Cas9 knockout approach:

    • Design guide RNAs targeting exons of the HIST1H1C gene

    • Validate knockout through western blotting to confirm complete protein loss

    • Sequence the targeted region to confirm genetic modification

    • Researchers have successfully generated HIST1H1C knockout A549 cells (A549-H1C-KO) using this approach

  • siRNA/shRNA knockdown:

    • Design multiple siRNA sequences targeting different regions of HIST1H1C mRNA

    • Optimize transfection conditions for target cell types

    • Validate knockdown efficiency by real-time PCR and western blotting

    • Studies have demonstrated specific HIST1H1C silencing during infection using siRNA approaches

  • In vivo knockdown:

    • For retinal experiments, intravitreal injection of siRNA against Hist1h1c has been effective

    • AAV vectors can be used for both knockdown (shRNA delivery) and overexpression

    • Local delivery methods minimize off-target effects in other tissues

  • Functional validation:

    • Compare viral replication, cytokine production, or autophagy markers between knockout/knockdown and control cells

    • Rescue experiments by re-expressing HIST1H1C or its mutants confirm phenotype specificity

    • Studies have shown significantly increased virus proliferation in A549-H1C-KO cells compared to wild-type cells, which can be reversed by HIST1H1C re-expression

These approaches have enabled researchers to establish the causal relationship between HIST1H1C and various cellular processes.

How can researchers distinguish between specific and non-specific binding when using HIST1H1C antibodies?

Distinguishing specific from non-specific HIST1H1C antibody binding requires rigorous validation:

  • Multiple antibody approach:

    • Use antibodies from different suppliers or those targeting different epitopes

    • Consistent results across antibodies suggest specific detection

    • For recombinant studies, compare results between epitope tag antibodies (e.g., HA-tag) and direct HIST1H1C antibodies

  • Knockout/knockdown validation:

    • Test antibodies in HIST1H1C knockout or knockdown samples

    • Specific bands or signals should be absent or significantly reduced

    • Non-specific signals will remain unchanged in knockout samples

  • Peptide competition assays:

    • Pre-incubate antibody with excess immunogenic peptide

    • Specific signals should be blocked while non-specific signals persist

    • Particularly useful for immunohistochemistry applications

  • Recombinant protein standards:

    • Include purified HIST1H1C protein as a positive control

    • Confirm antibody detects the correct molecular weight (approximately 30 kDa)

    • Useful for confirming specificity in western blotting

  • Comparison with other H1 variants:

    • Test cross-reactivity with other H1 variants (H1.0, H1.3, H1.4, etc.)

    • Evaluate signals in cell types with different H1 variant expression profiles

    • This is particularly important as H1 variants share sequence similarities

These rigorous validation steps are essential for ensuring reliable and reproducible results in HIST1H1C research.

What are common pitfalls in data interpretation when studying HIST1H1C-mediated effects?

Researchers should be aware of several potential pitfalls when interpreting data related to HIST1H1C:

  • Distinguishing direct vs. indirect effects:

    • HIST1H1C can affect chromatin structure broadly

    • Changes in gene expression may reflect direct HIST1H1C binding or indirect effects via chromatin reorganization

    • ChIP-seq combined with transcriptomics helps distinguish these mechanisms

  • Cell type-specific functions:

    • HIST1H1C functions may vary significantly between cell types

    • Results from one cell line (e.g., A549 or T47D) may not translate to other systems

    • Always validate findings across multiple relevant cell types

  • Compensation by other H1 variants:

    • Knockdown or knockout of HIST1H1C may lead to compensatory upregulation of other H1 variants

    • This may mask phenotypes in knockout models

    • Measure levels of other H1 variants when manipulating HIST1H1C

  • Post-translational modification complexity:

    • Different modifications can have opposing effects (e.g., phosphorylation vs. methylation)

    • PTMs may be regulated in complex, context-dependent patterns

    • Single-modification studies may oversimplify biological responses

  • Overexpression artifacts:

    • Extreme overexpression may cause non-physiological effects

    • Use moderate expression levels and compare with endogenous protein functions

    • Complementary loss-of-function approaches should support overexpression findings

Understanding these complexities helps researchers design more rigorous studies and interpret their data with appropriate caution.

How can contradictory results in HIST1H1C research be reconciled?

When faced with contradictory findings in HIST1H1C research, consider the following analytical approaches:

  • Methodological differences:

    • Compare antibody specificity and validation between studies

    • Assess differences in experimental conditions (cell types, stimulation protocols)

    • Evaluate knockout/knockdown efficiency across studies

    • Different ChIP-seq or analysis protocols may yield varying results

  • Cell type and context dependence:

    • HIST1H1C functions differently in breast cancer cells versus lung epithelial cells

    • Results from unstimulated cells may differ from those under stress conditions (viral infection, high glucose)

    • Healthy versus diseased tissue may show distinct HIST1H1C functions

  • Post-translational modification status:

    • Different modification patterns explain apparently contradictory functions

    • For example, phosphorylated versus methylated HIST1H1C has opposite effects on IFN-β production

    • Assess whether studies examined the same modifications

  • Interaction partners:

    • HIST1H1C functions through interaction with different partners (IRF3, NS2, etc.)

    • Presence or absence of these partners may explain varying results

    • Assess the protein complexes being studied in different experimental systems

  • Integration of multi-omics data:

    • Combining ChIP-seq, RNA-seq, proteomics, and functional assays provides a more complete picture

    • This comprehensive approach often resolves apparent contradictions by revealing condition-specific mechanisms

By systematically analyzing these factors, researchers can often identify the underlying reasons for discrepant results and develop more unified models of HIST1H1C function.

What are emerging techniques for studying HIST1H1C dynamics and interactions?

Several cutting-edge technologies are advancing HIST1H1C research:

  • Proximity labeling techniques:

    • BioID or TurboID fused to HIST1H1C can identify proximal interacting proteins

    • APEX2 fusion proteins allow temporal mapping of interaction networks

    • These approaches reveal previously unknown HIST1H1C binding partners in living cells

  • Live-cell imaging of HIST1H1C dynamics:

    • FRAP (Fluorescence Recovery After Photobleaching) with fluorescently-tagged HIST1H1C

    • Single-molecule tracking to visualize HIST1H1C movement and residence time on chromatin

    • These techniques reveal how HIST1H1C mobility changes during immune responses or stress conditions

  • Single-cell approaches:

    • Single-cell CUT&Tag or CUT&RUN for HIST1H1C binding with cellular resolution

    • Combined with single-cell RNA-seq to correlate binding with gene expression

    • Reveals cell-to-cell heterogeneity in HIST1H1C function during disease progression

  • Structural studies:

    • Cryo-EM of HIST1H1C-containing nucleosomes with transcription factors like IRF3

    • Hydrogen-deuterium exchange mass spectrometry to map interaction interfaces

    • These methods provide mechanistic insights into how HIST1H1C regulates target gene accessibility

These emerging methodologies will provide unprecedented insights into the dynamic functions of HIST1H1C in both physiological and pathological contexts.

How might HIST1H1C be targeted therapeutically in disease conditions?

Based on current research, HIST1H1C represents a promising therapeutic target with several potential approaches:

  • For viral infections (including influenza):

    • Develop small molecules that disrupt NS2-HIST1H1C interaction

    • Design peptide mimetics that stabilize HIST1H1C-IRF3 interaction

    • Target specific modifications of HIST1H1C (inhibit phosphorylation at T146 or enhance methylation at K34/K187)

    • These approaches could enhance antiviral immunity by preserving HIST1H1C's IFN-β promoting function

  • For diabetic retinopathy:

    • RNA interference approaches targeting HIST1H1C in the retina

    • Small molecules that inhibit HIST1H1C-mediated upregulation of autophagy

    • Agents that prevent HIST1H1C-induced inflammation without affecting beneficial functions

    • These strategies could reduce pathological autophagy and inflammation in diabetic retinopathy

  • Delivery considerations:

    • For retinal conditions, intravitreal injection of siRNA or AAV-delivered shRNA has shown efficacy

    • For systemic applications, nanoparticle-mediated delivery of HIST1H1C modulators

    • Cell-type specific promoters in viral vectors for targeted expression

  • Biomarker development:

    • HIST1H1C levels or modification patterns as diagnostic or prognostic markers

    • Monitoring HIST1H1C status to predict therapeutic responses

These therapeutic strategies represent promising avenues based on the emerging understanding of HIST1H1C's role in disease processes.

What unresolved questions remain in HIST1H1C biology?

Despite significant advances, several important questions about HIST1H1C remain unanswered:

  • Specificity vs. redundancy:

    • To what extent are HIST1H1C functions unique among H1 variants?

    • Are there genomic regions or cellular processes where HIST1H1C cannot be replaced by other variants?

    • How is variant-specific binding achieved despite high sequence similarity?

  • Regulatory mechanisms:

    • What factors control HIST1H1C expression in different cell types and conditions?

    • How is the balance between different post-translational modifications regulated?

    • What determines the genome-wide binding pattern of HIST1H1C?

  • Functional interactions:

    • Beyond IRF3 and NS2, what other proteins directly interact with HIST1H1C?

    • How does HIST1H1C communicate with core histones and chromatin remodeling complexes?

    • What is the full spectrum of genes and pathways regulated by HIST1H1C?

  • Evolution and conservation:

    • How conserved are HIST1H1C functions across species?

    • Does HIST1H1C play similar roles in antiviral immunity and autophagy regulation in diverse organisms?

    • Have pathogens other than influenza evolved mechanisms to target HIST1H1C?

  • Disease associations:

    • Beyond diabetic retinopathy and viral infections, what other diseases involve HIST1H1C dysregulation?

    • Are there naturally occurring HIST1H1C variants that affect disease susceptibility?

    • How does HIST1H1C contribute to cancer biology beyond its presence in breast cancer cells?

Addressing these questions will require integrated approaches combining structural biology, genomics, proteomics, and advanced imaging techniques applied across diverse experimental systems.

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 2024 Thebiotek. All Rights Reserved.