Formyl-HIST1H1C (K84) Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to Formyl-HIST1H1C (K84) Antibody

The Formyl-HIST1H1C (K84) Antibody is a polyclonal rabbit-derived antibody specifically targeting the formylated lysine residue at position 84 (K84) of histone H1.2, encoded by HIST1H1C. This post-translational modification (PTM) is critical for studying chromatin dynamics, epigenetic regulation, and cellular processes involving linker histones. The antibody is validated for applications such as immunofluorescence (IF) and enzyme-linked immunosorbent assay (ELISA), with a focus on detecting formylation at this site in human cells .

Immunofluorescence (IF) and Cellular Localization

The antibody enables visualization of formylated H1C in nuclear compartments. For example:

  • Protocol: Fixed cells (e.g., HeLa) are permeabilized, blocked, and incubated with the primary antibody overnight at 4°C. Detection uses Alexa Fluor 488-conjugated secondary antibodies .

  • Expected Localization: Nuclear staining, consistent with histone H1's role in chromatin structure .

Potential Role in Immune Modulation

While not directly studied for K84 formylation, anti-histone H1 antibodies broadly inhibit dendritic cell (DC) maturation by blocking signaling pathways (e.g., p38 MAPK and NF-κB) . This suggests that formylation at K84 could influence H1's extracellular signaling roles, such as modulating immune cell activity.

Comparison with Related Antibodies

AntibodyTargetApplicationsKey Differences
Formyl-HIST1H1C (K84)Formyl-Lys84 of H1.2IF, ELISASpecific to formylated K84; limited cross-reactivity
HIST1H1C (Ab-109, Ab-167) Acetylated or methylated residuesWB, ChIPTargets distinct PTMs (e.g., acetylation, methylation)
Histone H1.2 (19649-1-AP) Pan-H1.2 (non-site-specific)WB, IHC, IF, IP, ChIPBroad reactivity; lacks PTM specificity

Research Gaps and Future Directions

  1. Mechanistic Studies:

    • Elucidate how K84 formylation affects H1's DNA-binding affinity or chromatin compaction.

    • Explore its role in diseases linked to chromatin dysregulation (e.g., cancer, diabetic retinopathy) .

  2. Therapeutic Implications:

    • Investigate whether targeting K84 formylation could modulate autophagy or immune responses, as seen with other H1 PTMs .

  3. Cross-Species Reactivity:

    • Validate the antibody in non-human models (e.g., mouse, rat) to broaden its utility in preclinical studies .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we are able to ship products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchasing method and location. Please consult 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 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 findings define a network of E2F target genes that are susceptible to the regulatory influence of H1.2. H1.2 enhances the global association of pRb with chromatin, strengthens 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. The 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. Additionally, 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 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 are distinct 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. These data suggest 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 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. Studies demonstrate that the recruitment of YB1, PURalpha, and H1.2 to the p53 target gene Bax is required for the 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 Formyl-HIST1H1C (K84) Antibody and what does it detect?

Formyl-HIST1H1C (K84) Antibody is a polyclonal antibody that specifically recognizes the formylated lysine at position 84 of human Histone H1.2 (HIST1H1C). This antibody is designed to detect post-translational modifications (PTMs) on histone proteins that may have significant regulatory functions in chromatin structure and gene expression. The antibody (catalog number orb417591) is raised in rabbits against a peptide sequence surrounding the formyl-Lys (84) site derived from Human Histone H1.2 . This specific formylation is an important epigenetic marker that may regulate chromatin accessibility and gene expression patterns.

How does HIST1H1C differ from other histone variants?

HIST1H1C (Histone H1.2) is one of several H1 histone variants that functions as a linker histone. Unlike core histones (H2A, H2B, H3, and H4) that form nucleosome octamers, H1 histones bind to the nucleosome at the entry and exit sites of DNA, facilitating higher-order chromatin structure. HIST1H1C has a calculated molecular weight of 21 kDa, though it typically runs at 32-33 kDa on SDS-PAGE gels due to its highly basic nature and post-translational modifications . It is distinguished from other H1 variants by its specific amino acid sequence, temporal expression patterns during the cell cycle, and tissue distribution. Research indicates HIST1H1C may have specialized functions in certain cellular contexts, particularly in nuclear processes including hepatocarcinogenesis as evidenced by knockout studies .

What are the validated applications for Formyl-HIST1H1C (K84) Antibody?

The Formyl-HIST1H1C (K84) Antibody has been validated for several research applications:

ApplicationValidatedRecommended DilutionNotes
ELISAYesAccording to protocolFor quantitative detection of formylated HIST1H1C
Immunofluorescence (IF)Yes1:50-200Successfully used in Hela cells

Other HIST1H1C antibodies (not specifically formyl-K84) have been validated for additional applications including Western Blot (1:500-1:3000), Immunoprecipitation (0.5-4.0 μg for 1.0-3.0 mg protein), Immunohistochemistry (1:100-1:600), and ChIP assays . Researchers should carefully select the appropriate antibody based on their specific experimental needs and target modification.

How does formylation at K84 affect HIST1H1C function in chromatin regulation?

Lysine formylation represents an important but less-studied histone post-translational modification compared to methylation and acetylation. Formylation of HIST1H1C at K84 likely impacts its binding affinity to DNA and interaction with chromatin remodeling complexes. Current research suggests that histone formylation may result from oxidative stress conditions, potentially serving as a cellular stress response mechanism.

The specific K84 position on HIST1H1C appears within a region that interacts with linker DNA, suggesting that formylation at this site could modulate the ability of H1.2 to compact chromatin or regulate nucleosome spacing. Researchers investigating this modification should consider combining ChIP-seq with formyl-specific antibodies to identify genomic regions where this modification predominates, and correlate with gene expression data to determine functional consequences.

What is the relationship between HIST1H1C and STAT3 signaling in cancer progression?

Research indicates a significant relationship between HIST1H1C and STAT3 signaling pathways, particularly relevant to cancer progression. Studies using HIST1H1C knockout mice have identified its role in hepatocarcinogenesis through regulation of signal transducer and activator of transcription 3 (STAT3) signaling . The H1C promoter contains multiple STAT3 binding sites (including positions -1423/-1413, -701/-691, and -162/-152) that have been confirmed through ChIP assays .

This interaction suggests that HIST1H1C expression may be regulated by STAT3 activation, creating a potential feedback loop in cancer progression. Researchers investigating cancer mechanisms should consider examining how formylation at K84 might influence this pathway, potentially altering HIST1H1C's interaction with STAT3 or affecting downstream gene expression patterns in cancer cells.

How can HIST1H1C formylation be distinguished from other post-translational modifications in multiplexed epigenetic studies?

Distinguishing histone formylation from other similar post-translational modifications presents a significant technical challenge. Researchers conducting multiplexed epigenetic studies should implement a multi-methodological approach:

  • Antibody specificity validation: Perform competitive binding assays with peptides containing formyl-K84, acetyl-K84, and unmodified K84 to confirm the Formyl-HIST1H1C antibody's specificity.

  • Mass spectrometry verification: Use high-resolution MS/MS to distinguish formyl (mass shift +28 Da) from acetyl modifications (mass shift +42 Da).

  • Chemical labeling strategies: Employ formaldehyde-specific chemical probes that react differentially with formyl groups versus acetyl groups.

  • Orthogonal techniques: Combine antibody-based detection with enzymatic assays that specifically remove formyl groups but not acetyl groups.

  • Context-dependent controls: Include oxidative stress conditions known to increase formylation rates versus HDAC inhibitors that increase acetylation as experimental controls.

This comprehensive approach enables researchers to confidently identify and quantify formylation in complex epigenetic landscapes.

What are the optimal conditions for immunofluorescence assays using Formyl-HIST1H1C (K84) Antibody?

For optimal immunofluorescence results with Formyl-HIST1H1C (K84) Antibody:

ParameterRecommended ConditionNotes
Fixation4% paraformaldehyde, 10 minutes at RTPreserves nuclear architecture and epitope accessibility
Permeabilization0.1% Triton X-100, 5 minutesSufficient for nuclear penetration without excessive extraction
Blocking5% BSA in PBS, 1 hour at RTReduces non-specific binding
Primary antibody dilution1:50-1:200 in blocking bufferHas been validated in HeLa cells
IncubationOvernight at 4°CAllows complete epitope binding
Secondary antibodyAnti-rabbit IgG conjugated with appropriate fluorophoreSelection based on imaging system specifications
CounterstainDAPI (1 μg/ml)For nuclear visualization
Mounting mediumAnti-fade reagentPrevents photobleaching during imaging

Researchers should optimize these conditions based on their specific cell type and sample preparation methods. For challenging samples, consider antigen retrieval methods or alternative fixation protocols if initial results are suboptimal.

How should HIST1H1C knockout models be validated for studying histone H1.2 functions?

Proper validation of HIST1H1C knockout models is critical for accurate interpretation of experimental results. A comprehensive validation strategy includes:

  • Genomic verification: Confirm the knockout at the DNA level using PCR with primers flanking the targeted region. The CRISPR/Cas9 system targeting the 5'-UTR and 3'-UTR of Hist1h1c exon 1 has been successfully used to generate knockout mice .

  • Transcript analysis: Perform RT-PCR and qPCR to verify the absence of Hist1h1c mRNA.

  • Protein verification: Use Western blotting with validated HIST1H1C antibodies (such as 19649-1-AP, which has been tested in multiple tissues and cell lines) to confirm the absence of H1.2 protein. The expected molecular weight is 32-33 kDa despite a calculated weight of 21 kDa.

  • Compensatory mechanisms assessment: Evaluate the expression levels of other H1 variants (H1.1, H1.3, H1.4, H1.5) to identify potential compensatory upregulation.

  • Phenotypic characterization: Document all observable phenotypes, including changes in chromatin compaction (using MNase assays), cell cycle progression, and tissue-specific alterations.

  • Functional rescue: Perform rescue experiments by reintroducing wild-type HIST1H1C to verify that observed phenotypes are directly attributable to H1.2 deficiency.

This comprehensive validation approach ensures reliable experimental outcomes when using these models for studying histone H1.2 functions in various biological contexts.

What controls should be included when studying formylation of HIST1H1C in cellular stress responses?

When investigating HIST1H1C formylation in cellular stress responses, include these essential controls:

  • Negative controls:

    • Unstressed cells to establish baseline formylation levels

    • HIST1H1C knockout or knockdown cells to confirm antibody specificity

    • Immunoprecipitation with non-specific IgG of matching species and isotype

  • Positive controls:

    • Cells treated with known formylation inducers (e.g., high glucose, hypoxia)

    • Recombinant formylated HIST1H1C peptides at known concentrations

  • Specificity controls:

    • Competitive inhibition with excess formylated HIST1H1C peptides

    • Parallel detection with antibodies against other HIST1H1C modifications

    • Mass spectrometry validation of formylation sites

  • Procedural controls:

    • Treatment with deformylase enzymes to remove the modification

    • Time-course experiments to capture dynamic changes in formylation

    • Dose-response studies to establish threshold levels for stress-induced formylation

  • Biological context controls:

    • Multiple cell types to assess tissue-specific responses

    • Inhibitors of stress-response pathways to determine mechanism specificity

    • Alternative stress inducers to test context-dependent formylation patterns

These comprehensive controls ensure robust data interpretation and minimize experimental artifacts when studying this sensitive post-translational modification.

How can researchers troubleshoot weak or non-specific signals in immunofluorescence using Formyl-HIST1H1C (K84) Antibody?

When encountering weak or non-specific signals, implement this systematic troubleshooting approach:

ProblemPossible CausesSolutions
Weak signalInsufficient antibody concentrationIncrease antibody concentration (try 1:50 dilution instead of 1:200)
Epitope masking during fixationTry alternative fixation methods (methanol or acetone)
Low abundance of formylated proteinInduce formylation with oxidative stress treatments
Improper storage of antibodyStore at -20°C with glycerol to prevent freeze-thaw damage
High backgroundInsufficient blockingExtend blocking time to 2 hours; try 5% normal serum instead of BSA
Excessive antibody concentrationDilute antibody further; 1:200-1:500 range
Non-specific secondary antibody bindingInclude additional blocking steps; try different secondary antibody
AutofluorescenceInclude quenching steps; use different fluorophores
Non-nuclear stainingCross-reactivity with other formylated proteinsPerform peptide competition assays to verify specificity
Cytoplasmic localization of HIST1H1CVerify with cellular fractionation and Western blot
No signalIncorrect application of antibodyVerify the antibody is suitable for IF (confirmed for Formyl-HIST1H1C (K84))
Incompatible cell typeTest alternative cell lines; HeLa cells have been validated

For optimal results, researchers should titrate the antibody concentration for each specific cell type and ensure appropriate image acquisition parameters on their microscopy systems.

How should researchers interpret discrepancies between predicted and observed molecular weights of HIST1H1C in Western blots?

Researchers commonly observe HIST1H1C running at 32-33 kDa despite its calculated molecular weight of 21 kDa . This significant discrepancy requires careful interpretation:

  • Post-translational modifications: Histone proteins frequently undergo extensive PTMs (phosphorylation, acetylation, methylation, formylation) that can substantially alter their electrophoretic mobility. The formylation at K84 and other potential modifications contribute to this shift.

  • Intrinsic protein properties: HIST1H1C is highly basic (high isoelectric point), which affects SDS binding and results in anomalous migration in SDS-PAGE systems. This is common for histone proteins.

  • Verification approaches:

    • Use recombinant HIST1H1C protein as size reference

    • Perform mass spectrometry to confirm the actual molecular weight

    • Include HIST1H1C knockout samples as negative controls

    • Test multiple antibodies targeting different epitopes of HIST1H1C

    • Use gradient gels to improve resolution in the relevant size range

  • Interpretation guidelines:

    • Report both the observed and calculated molecular weights in publications

    • Document running conditions carefully (gel percentage, buffer system)

    • Consider using specialized gel systems optimized for basic proteins

    • Validate identity through immunoprecipitation followed by mass spectrometry

This mobility shift is a consistent feature of HIST1H1C and should not be interpreted as non-specific binding if other validation criteria are met.

How can researchers quantify changes in HIST1H1C formylation levels across different experimental conditions?

For accurate quantification of HIST1H1C formylation across experimental conditions, implement this multi-faceted approach:

  • Western blot quantification:

    • Use formyl-specific antibodies alongside total HIST1H1C antibodies

    • Normalize formylated signal to total HIST1H1C protein

    • Include loading controls (β-actin, GAPDH) for total protein normalization

    • Apply densitometric analysis with appropriate software (ImageJ, Image Studio)

  • Immunofluorescence quantification:

    • Measure nuclear intensity of formyl-HIST1H1C staining

    • Co-stain with total HIST1H1C antibody from different species

    • Calculate the ratio of formylated/total signal per nucleus

    • Analyze >100 cells per condition for statistical robustness

  • Mass spectrometry approaches:

    • Use SILAC or TMT labeling for direct comparison between conditions

    • Calculate stoichiometry of formylation at K84 relative to unmodified peptide

    • Monitor multiple HIST1H1C peptides to control for protein level changes

    • Employ parallel reaction monitoring (PRM) for highest sensitivity

  • ChIP-based quantification:

    • Perform ChIP-seq with formyl-specific antibodies

    • Compare genomic distribution of formylated HIST1H1C between conditions

    • Normalize to total HIST1H1C occupancy from parallel ChIP experiments

    • Identify condition-specific changes in genomic localization

These complementary approaches provide comprehensive assessment of both global and site-specific changes in HIST1H1C formylation levels across different experimental conditions.

How can Formyl-HIST1H1C (K84) Antibody be used to investigate the role of histone formylation in cancer progression?

The Formyl-HIST1H1C (K84) Antibody offers valuable insights into cancer research through multiple methodological applications:

  • Tumor tissue analysis:

    • Perform immunohistochemistry on cancer tissue microarrays to correlate formylation levels with clinical outcomes

    • Compare formylation in tumor versus adjacent normal tissue

    • Studies with HIST1H1C antibodies have successfully detected the protein in human thyroid cancer tissue and ovarian tumor tissue

  • Mechanistic investigation:

    • Evaluate the relationship between HIST1H1C formylation and STAT3 signaling, as HIST1H1C has demonstrated involvement in hepatocarcinogenesis through STAT3 pathway regulation

    • Perform ChIP-seq to identify genomic regions where formylated HIST1H1C binds preferentially in cancer cells

    • Correlate with gene expression data to identify formylation-regulated oncogenes or tumor suppressors

  • Cancer model systems:

    • Compare formylation levels across cancer cell lines with varying aggressiveness

    • Use HIST1H1C knockout models (generated using CRISPR/Cas9 targeting the 5′-UTR and 3′-UTR of Hist1h1c exon 1) to assess the requirement for H1.2 in tumorigenesis

    • Examine how oxidative stress conditions alter formylation patterns in cancer progression

  • Therapeutic implications:

    • Test whether cancer treatments affect HIST1H1C formylation status

    • Investigate whether formylation levels predict treatment response

    • Explore the potential of targeting enzymes responsible for histone formylation

This multifaceted approach leverages the Formyl-HIST1H1C (K84) Antibody to comprehensively investigate the emerging role of histone formylation in cancer biology.

What methodologies are recommended for studying HIST1H1C in liver disease and hepatocarcinogenesis?

Based on established research showing HIST1H1C's role in hepatocarcinogenesis , the following methodologies are recommended:

  • Animal models:

    • Utilize HIST1H1C knockout mice generated via CRISPR/Cas9 targeting of the 5′-UTR and 3′-UTR of Hist1h1c exon 1

    • Employ diethylnitrosamine (DEN)-induced hepatocarcinogenesis model to study tumor development

    • Monitor liver tumor formation, size, and multiplicity in wild-type versus knockout animals

    • Analyze liver function parameters (ALT, AST, bilirubin) throughout disease progression

  • Histological and immunohistochemical analysis:

    • Perform H&E staining to assess liver architecture and tumor pathology

    • Use immunohistochemistry with antibodies against:

      • HIST1H1C (recommended dilution 1:100-1:600)

      • Ki-67 for proliferation assessment

      • F4/80 for macrophage infiltration

      • CD3 for T-cell infiltration

      • Ly6G for neutrophil infiltration

      • p-STAT3 Y705 to evaluate STAT3 pathway activation

  • Molecular mechanisms investigation:

    • Perform ChIP assays to identify HIST1H1C binding sites in liver tissue

    • Analyze STAT3 binding to the HIST1H1C promoter at key sites (-1423/-1413, -701/-691, and -162/-152)

    • Conduct luciferase reporter assays with wild-type and mutant HIST1H1C promoters

    • Evaluate formylation status of HIST1H1C at K84 during disease progression

  • Clinical translation:

    • Analyze HIST1H1C expression and formylation in human HCC tissue microarrays

    • Correlate with clinical parameters and patient outcomes

    • Investigate potential as a biomarker for disease progression or treatment response

This comprehensive methodology leverages established techniques while focusing on the specific role of HIST1H1C in liver disease pathogenesis.

How can epigenetic cross-talk between HIST1H1C formylation and other histone modifications be investigated?

Investigating epigenetic cross-talk between HIST1H1C formylation and other modifications requires sophisticated methodological approaches:

  • Sequential ChIP (Re-ChIP):

    • First immunoprecipitate with Formyl-HIST1H1C (K84) Antibody

    • Elute and perform second immunoprecipitation with antibodies against other modifications

    • Sequence resulting DNA to identify genomic regions with co-occurrence

    • Alternatively, perform in reverse order to confirm bidirectional relationship

  • Mass spectrometry-based approaches:

    • Perform top-down mass spectrometry on intact HIST1H1C to identify combinatorial modification patterns

    • Use middle-down approaches focusing on larger histone fragments

    • Employ electron transfer dissociation (ETD) for improved PTM identification

    • Quantify co-occurrence frequencies of formylation with other modifications

  • Proximity ligation assays (PLA):

    • Use Formyl-HIST1H1C (K84) Antibody together with antibodies against other modifications

    • Visualize and quantify co-localization at the single-cell level

    • Perform under different cellular conditions to assess dynamic relationships

  • Biochemical interaction studies:

    • Identify proteins that specifically recognize formylated HIST1H1C

    • Determine how these readers are affected by adjacent modifications

    • Test whether formylation affects the activity of enzymes that modify neighboring residues

  • Genetic manipulation approaches:

    • Create targeted mutations that prevent specific modifications

    • Assess the impact on formylation levels and distribution

    • Use CRISPR/Cas9-based approaches to target writer enzymes for each modification

  • Computational integration:

    • Develop machine learning algorithms to identify patterns in multi-modal epigenetic data

    • Predict functional consequences of specific modification combinations

    • Model the hierarchical relationships between modifications

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.