Formyl-HIST1H4A (K12) Antibody

Shipped with Ice Packs
In Stock

Description

Target and Immunogen

  • Target: Formylated lysine 12 (K12) on histone H4 (HIST1H4A), a post-translational modification (PTM) involving the addition of a formyl group to the ε-amino group of lysine .

  • Immunogen: Synthetic peptide sequence around formyl-Lys12 derived from human histone H4 .

  • Host: Rabbit .

  • Reactivity: Primarily human, with cross-reactivity reported in rat in some formulations .

Key Features

AttributeDetail
ClonalityPolyclonal
ConjugationUnconjugated
PurificationAntigen affinity purification
Storage-20°C or -80°C (long-term); 2–8°C (short-term)
Application Buffer50% glycerol, 0.01M PBS, pH 7.4

Applications and Dilution Guidelines

The antibody is validated for multiple techniques, with dilutions optimized for sensitivity and specificity:

ApplicationRecommended DilutionNotes
Western Blot (WB)1:100–1:1,000 Detects formylated H4 in whole-cell lysates or nuclear extracts .
Immunocytochemistry (ICC)1:10–1:100 Stains nuclear regions in fixed cells; compatible with paraformaldehyde fixation .
Immunofluorescence (IF)1:50–1:200 Visualizes chromatin-bound formyl-H4 in permeabilized cells .
ELISAVaries by protocol Quantitative assessment of formyl-H4 levels in cell lysates .

Suppliers and Pricing

Multiple vendors offer this antibody with varying sizes and prices:

SupplierSizePrice (USD)ApplicationsCitations
Cusabio100 µl$175WB, ICC, IF, ELISA N/A
Biomatik100 µl$335WB, ELISA, ICC, IF, IP, ChIP N/A
Qtonics50 µl$200
100 µl
$319ELISA, WB, ICC, IF
EpiGentek50 µl/100 µl$249WB, ELISA, ICC, IF N/A

Role of Formylation in Epigenetics

  • Chromatin Dynamics: Formylation may modulate histone-DNA interactions, affecting gene expression .

  • DNA Repair: PTMs on histones are critical for recruiting repair machinery; formylation might play a role in this process .

  • Cancer and Disease: Dysregulation of histone modifications is implicated in oncogenesis; formylation could serve as a biomarker .

Comparison with Acetylation

While antibodies for acetylated H4 (e.g., H4K12ac) are widely used, the Formyl-HIST1H4A (K12) Antibody fills a gap in detecting formylation. Key distinctions include:

ModificationEnzymatic RegulationBiological ContextDetection Tools
FormylationUnknown (likely specific enzymes)Niche PTM; potential role in repairPolyclonal antibodies (e.g., this tool)
AcetylationHATs and HDACsBroad chromatin remodelingMonoclonal/polyclonal antibodies

Challenges and Future Directions

  • Limited Validation: Direct studies using this antibody are sparse, though its specificity is inferred from immunogen design and WB/ICC data .

  • Cross-Reactivity: Reactivity in non-human species (e.g., rat) requires cautious interpretation .

  • Research Potential: This antibody could enable high-throughput screening for formylation in epigenetic studies, particularly in contexts where acetylation is masked or irrelevant .

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 after receiving it. Delivery times may vary depending on your location and shipping method. For specific delivery information, please consult with your local distributor.
Synonyms
dJ160A22.1 antibody; dJ160A22.2 antibody; dJ221C16.1 antibody; dJ221C16.9 antibody; FO108 antibody; H4 antibody; H4.k antibody; H4/a antibody; H4/b antibody; H4/c antibody; H4/d antibody; H4/e antibody; H4/g antibody; H4/h antibody; H4/I antibody; H4/j antibody; H4/k antibody; H4/m antibody; H4/n antibody; H4/p antibody; H4_HUMAN antibody; H4F2 antibody; H4F2iii antibody; H4F2iv antibody; H4FA antibody; H4FB antibody; H4FC antibody; H4FD antibody; H4FE antibody; H4FG antibody; H4FH antibody; H4FI antibody; H4FJ antibody; H4FK antibody; H4FM antibody; H4FN antibody; H4M antibody; HIST1H4A antibody; HIST1H4B antibody; HIST1H4C antibody; HIST1H4D antibody; HIST1H4E antibody; HIST1H4F antibody; HIST1H4H antibody; HIST1H4I antibody; HIST1H4J antibody; HIST1H4K antibody; HIST1H4L antibody; HIST2H4 antibody; HIST2H4A antibody; Hist4h4 antibody; Histone 1 H4a antibody; Histone 1 H4b antibody; Histone 1 H4c antibody; Histone 1 H4d antibody; Histone 1 H4e antibody; Histone 1 H4f antibody; Histone 1 H4h antibody; Histone 1 H4i antibody; Histone 1 H4j antibody; Histone 1 H4k antibody; Histone 1 H4l antibody; Histone 2 H4a antibody; histone 4 H4 antibody; Histone H4 antibody; MGC24116 antibody
Target Names
HIST1H4A
Uniprot No.

Target Background

Function
Histone H4 is a core component of the nucleosome, which plays a vital role in packaging and compacting DNA into chromatin. This compact structure limits DNA accessibility to cellular machinery, such as those involved in transcription, DNA repair, replication, and chromosomal stability. Histones, including Histone H4, regulate DNA accessibility through intricate post-translational modifications, often referred to as the histone code. These modifications influence various cellular processes by altering chromatin structure and function.
Gene References Into Functions
  1. Studies have demonstrated that PP32 and SET/TAF-Ibeta proteins inhibit HAT1-mediated H4 acetylation. PMID: 28977641
  2. Research suggests that post-translational modifications of histones, specifically trimethylation of lysine 36 in H3 (H3K36me3) and acetylation of lysine 16 in H4 (H4K16ac), play crucial roles in DNA damage repair. H3K36me3 stimulates H4K16ac upon DNA double-strand breaks, and this process is dependent on SETD2, LEDGF, and KAT5 (SETD2 = SET domain containing 2; LEDGF = lens epithelium-derived growth factor; KAT5 = lysine acetyltransferase 5). PMID: 28546430
  3. Data indicates that Omomyc protein colocalizes with proto-oncogene protein c-myc (c-Myc), protein arginine methyltransferase 5 (PRMT5), and histone H4 H4R3me2s-enriched chromatin domains. PMID: 26563484
  4. H4K12ac is regulated by estrogen receptor-alpha and is associated with BRD4 function and inducible transcription. PMID: 25788266
  5. Systemic lupus erythematosus appears to be associated with an imbalance in histone acetyltransferases and histone deacetylase enzymes, leading to pathologic H4 acetylation. PMID: 25611806
  6. Sumoylated human histone H4 inhibits chromatin compaction by disrupting long-range internucleosomal interactions. PMID: 25294883
  7. Acetylation at lysine 5 of histone H4 is associated with lytic gene promoters during reactivation of Kaposi's sarcoma-associated herpesvirus. PMID: 25283865
  8. Increased histone H4 acetylation caused by hypoxia in human neuroblastoma cell lines is correlated with elevated levels of N-myc transcription factor. PMID: 24481548
  9. Research suggests that G1-phase histone assembly is restricted to CENP-A and H4. PMID: 23363600
  10. This study focused on the distribution of a specific histone modification, namely H4K12ac, in human sperm and characterized its specific enrichment sites in promoters throughout the whole human genome. PMID: 22894908
  11. SRP68/72 heterodimers are identified as major nuclear proteins whose binding of histone H4 tail is inhibited by H4R3 methylation. PMID: 23048028
  12. TNF-alpha inhibition of AQP5 expression in human salivary gland acinar cells is attributed to an epigenetic mechanism involving suppression of acetylation of histone H4. PMID: 21973049
  13. Research indicates that global histone H3 and H4 modification patterns may serve as potential markers for tumor recurrence and disease-free survival in non-small cell lung cancer. PMID: 22360506
  14. HAT1 differentially impacts nucleosome assembly of H3.1-H4 and H3.3-H4. PMID: 22228774
  15. Phosphorylation of histone H4 Ser 47, catalyzed by the PAK2 kinase, promotes nucleosome assembly of H3.3-H4 and inhibits nucleosome assembly of H3.1-H4 by enhancing the binding affinity of HIRA to H3.3-H4 and reducing the association of CAF-1 with H3.1-H4. PMID: 21724829
  16. The imatinib-induced hemoglobinization and erythroid differentiation in K562 cells are associated with global histone H4 modifications. PMID: 20949922
  17. Findings reveal the molecular mechanisms by which DNA sequences within specific gene bodies initiate monomethylation of histone H4 lysine 200, leading to a reduction in gene expression by half. PMID: 20512922
  18. Histone H4 expression is downregulated by zinc and upregulated by docosahexaenoate in a neuroblastoma cell line. PMID: 19747413
  19. Low levels of histone acetylation are linked to the development and progression of gastric carcinomas, potentially through alterations in gene expression. PMID: 12385581
  20. Overexpression of MTA1 protein is closely associated with the acetylation level of histone H4 protein. PMID: 15095300
  21. Peptidylarginine deiminase 4 (PAD4) regulates histone Arg methylation by converting methyl-Arg to citrulline and releasing methylamine. Data suggests that PAD4 influences gene expression by modulating Arg methylation and citrullination in histones. PMID: 15345777
  22. The lack of biotinylation of K12 in histone H4 is an early signaling event in response to double-strand breaks. PMID: 16177192
  23. The incorporation of acetylated histone H4-K16 into nucleosomal arrays inhibits the formation of compact 30-nanometer-like fibers and hinders the ability of chromatin to form cross-fiber interactions. PMID: 16469925
  24. Apoptosis is associated with global DNA hypomethylation and histone deacetylation events in leukemia cells. PMID: 16531610
  25. BTG2 contributes to retinoic acid activity by promoting differentiation through gene-specific modifications of histone H4 arginine methylation and acetylation levels. PMID: 16782888
  26. There is a relationship between histone H4 modification, epigenetic regulation of BDNF gene expression, and long-term memory for extinction of conditioned fear. PMID: 17522015
  27. The H4 tail and its acetylation play novel roles in mediating the recruitment of multiple regulatory factors that can modify chromatin states for transcription regulation. PMID: 17548343
  28. Brd2 bromodomain 2 exists as a monomer in solution and dynamically interacts with H4-AcK12. Additional secondary elements in the long ZA loop may be a common feature of BET bromodomains. PMID: 17848202
  29. Spermatids Hypac-H4 impairment in mixed atrophy is not further worsened by AZFc region deletion. PMID: 18001726
  30. The interaction between SET8 and PCNA couples H4-K20 methylation with DNA replication. PMID: 18319261
  31. H4K20 monomethylation and PR-SET7 are essential for L3MBTL1 function. PMID: 18408754
  32. High expression of acetylated H4 is more prevalent in aggressive than indolent cutaneous T-cell lymphoma. PMID: 18671804
  33. Findings indicate a significant role of histone H4 modifications in bronchial carcinogenesis. PMID: 18974389
  34. Results show that acetylation of histone H4 K16 during S-phase leads to early replicating chromatin domains acquiring the H4K16ac-K20me2 epigenetic label, which persists throughout mitosis and is deacetylated in early G1-phase of the next cell cycle. PMID: 19348949
  35. Acetylated H4 is overexpressed in diffuse large B-cell lymphoma and peripheral T-cell lymphoma compared to normal lymphoid tissue. PMID: 19438744
  36. The release of histone H4 by holocrine secretion from the sebaceous gland may play a crucial role in innate immunity. PMID: 19536143
  37. Histone modification, including PRC2-mediated repressive histone marker H3K27me3 and active histone marker acH4, may be involved in CD11b transcription during HL-60 leukemia cells reprogramming to terminal differentiation. PMID: 19578722
  38. A role of Cdk7 in regulating elongation is further suggested by enhanced histone H4 acetylation and reduced histone H4 trimethylation on lysine 36, two marks of elongation, within genes when the kinase was inhibited. PMID: 19667075
  39. Data reveals the dynamic fluctuation of histone H4 acetylation levels during mitosis, as well as acetylation changes in response to structurally distinct histone deacetylase inhibitors. PMID: 19805290
  40. Data directly implicates BBAP in the monoubiquitylation and additional posttranslational modification of histone H4 and an associated DNA damage response. PMID: 19818714

Show More

Hide All

Database Links

HGNC: 4781

OMIM: 142750

KEGG: hsa:121504

STRING: 9606.ENSP00000367034

UniGene: Hs.143080

Involvement In Disease
Chromosomal aberrations involving HISTONE H4 is a cause of B-cell non-Hodgkin lymphomas (B-cell NHL). Translocation t(3;6)(q27;p21), with BCL6.
Protein Families
Histone H4 family
Subcellular Location
Nucleus. Chromosome.

Q&A

What is the Formyl-HIST1H4A (K12) antibody and what does it detect?

The Formyl-HIST1H4A (K12) antibody is a polyclonal antibody that specifically recognizes formylated lysine at position 12 of human histone H4 (HIST1H4A). This antibody detects a specific post-translational modification (PTM) that occurs on histone H4, which is part of the nucleosome core particle in chromatin structure. The antibody is typically raised in rabbits using a peptide sequence around the site of Formyl-Lys (12) derived from Human Histone H4 as the immunogen . This specific antibody provides researchers with a tool to investigate formylation at K12, which represents an important epigenetic mark that may influence chromatin organization and gene expression.

What are the common applications for Formyl-HIST1H4A (K12) antibody?

The Formyl-HIST1H4A (K12) polyclonal antibody has been validated for several standard laboratory applications in epigenetics and molecular biology research:

  • Enzyme-Linked Immunosorbent Assay (ELISA): For quantitative detection of formylated H4K12 in cell or tissue lysates

  • Western Blotting (WB): For detecting the presence and relative abundance of formylated H4K12 in protein samples

  • Immunocytochemistry (ICC): For visualizing the cellular distribution of formylated H4K12 in fixed cells

  • Immunofluorescence (IF): For fluorescent visualization of formylated H4K12 localization in cells or tissues

When designing experiments, researchers should consider that the antibody specifically recognizes human (Homo sapiens) histone H4 with formylation at the K12 position, which may limit cross-reactivity with other species or modifications.

What is the biological significance of histone H4 K12 formylation compared to other modifications?

Histone H4 can be acetylated at N-terminal lysines K5, K8, K12, and K16, with each modification potentially serving distinct biological functions . While acetylation at K12 has been extensively studied, formylation represents a distinct modification with potentially different roles. Newly synthesized histone H4 is typically diacetylated at K5/K12 in diverse organisms, a pattern historically associated with histone deposition during DNA replication .

How should researchers optimize experimental conditions when using Formyl-HIST1H4A (K12) antibody for chromatin immunoprecipitation (ChIP) assays?

While the product information indicates the antibody has been validated for ELISA, WB, ICC, and IF applications , researchers interested in using it for ChIP should first validate its specificity in this context. For optimal ChIP protocol development:

  • Crosslinking optimization: Start with standard formaldehyde crosslinking (1% for 10 minutes at room temperature), but consider titrating concentration and time for histone formylation studies.

  • Sonication parameters: Aim for chromatin fragments of 200-500bp. This typically requires optimization with your specific sonicator settings.

  • Antibody concentration: Begin with 2-5μg of antibody per ChIP reaction and adjust based on preliminary results. Include appropriate controls:

    • Input chromatin control (pre-immunoprecipitation)

    • Negative control using non-specific IgG

    • Positive control using an antibody against a known abundant histone mark

  • Washing stringency: Balance between minimizing background and preserving specific interactions. Consider a titration of salt concentrations in wash buffers.

  • Validation: Confirm specificity using:

    • Western blot of input and immunoprecipitated material

    • qPCR of regions known to be enriched or depleted for this mark

    • Peptide competition assays with formylated and unmodified peptides

The success of ChIP experiments with this antibody will depend on the abundance of the formyl-K12 mark and potential epitope masking within chromatin complexes.

What are the key considerations when designing experiments to investigate the relationship between histone H4 K12 formylation and nucleosome assembly?

Research on the analogous acetylation at K12 has shown that this modification alone is not required for nucleosome assembly in yeast . When designing experiments to investigate whether formylation at K12 influences nucleosome assembly, consider these methodological approaches:

  • In vivo superhelicity assays: Measure plasmid superhelical density in whole cells as an indicator of nucleosome formation, similar to methods used to demonstrate that K5/K12 mutations do not prevent nucleosome assembly in yeast . This approach requires:

    • Construction of cellular systems with mutations at the K12 site that prevent formylation

    • Extraction of plasmid DNA under conditions that preserve supercoiling

    • Analysis by agarose gel electrophoresis with chloroquine to resolve topoisomers

  • In vitro nucleosome assembly assays: Using purified components to assess whether formylation at K12 influences assembly kinetics or structural outcomes:

    • Compare assembly efficiency using histone H4 with and without formylation at K12

    • Analyze by native gel electrophoresis, MNase digestion patterns, and salt-dependent stability assays

  • Combined mutation approaches: Based on findings that K5, K8, and K12 function redundantly in nucleosome assembly , design experiments that systematically examine combinations of modifications:

    • Create systems with mutations preventing formylation at multiple sites

    • Test for synthetic phenotypes that appear only when multiple sites are mutated

    • Consider the influence of the histone H3 N-terminus, which has functions redundant with H4 in histone deposition

How can researchers differentiate between formylation and acetylation at H4K12 in their experimental analyses?

Distinguishing between formylation and acetylation at the same residue presents significant technical challenges. A methodological approach should include:

  • Antibody specificity validation:

    • Perform peptide competition assays using synthetic peptides containing either formylated or acetylated K12

    • Conduct Western blot analysis comparing known formylated and acetylated samples

    • Consider dot blot analysis with modified and unmodified peptides at varying concentrations

  • Mass spectrometry approaches:

    • Utilize high-resolution mass spectrometry to distinguish between formylation (mass shift of +28 Da) and acetylation (mass shift of +42 Da)

    • Implement targeted MS/MS analysis focusing specifically on the K12-containing peptide

    • Consider chemical derivatization strategies that can selectively react with either formyl or acetyl groups

  • Sequential immunoprecipitation:

    • First immunoprecipitate with anti-acetyl-K12 antibody

    • Then immunoprecipitate the unbound fraction with anti-formyl-K12 antibody

    • Analyze both fractions to assess relative abundance and distribution

  • Enzymatic manipulation:

    • Treat samples with histone deacetylases (HDACs) that remove acetyl but not formyl groups

    • Compare antibody reactivity before and after HDAC treatment

How should researchers interpret conflicting results between formylation detection at H4K12 and functional outcomes in nucleosome assembly?

When facing contradictory results between the detection of formylation at H4K12 and functional outcomes in nucleosome assembly experiments, consider these analytical approaches:

  • Assess modification abundance: The functional impact of a modification often depends on its stoichiometry. Quantitative approaches like mass spectrometry can determine what percentage of H4K12 is actually formylated in your experimental system.

  • Consider context dependency: The research on H4K5/K12 acetylation demonstrates that these modifications function redundantly with K8 in nucleosome assembly . Similarly, the functional significance of H4K12 formylation might depend on the modification state of neighboring residues. Analyze:

    • Co-occurrence patterns with other modifications

    • Cell cycle phase-specific effects

    • Cell type-specific differences

  • Evaluate kinetics: The timing of formylation relative to other cellular processes may be crucial:

    • Is formylation occurring before, during, or after nucleosome assembly?

    • Does the formylation persist or is it transient?

    • How does the kinetics of formylation compare with that of acetylation at the same site?

  • Reexamine experimental controls: Based on findings that multiple lysine residues (K5, K8, K12) function redundantly , ensure your experimental design adequately controls for compensation by other sites. Consider:

    • Using combined mutations or modifications

    • Implementing acute rather than chronic interventions

    • Employing conditional systems to minimize adaptation

What statistical approaches are recommended for analyzing ChIP-seq data generated using Formyl-HIST1H4A (K12) antibody?

For robust statistical analysis of ChIP-seq data using Formyl-HIST1H4A (K12) antibody, implement this methodological framework:

  • Quality control metrics:

    • Assess library complexity (PCR duplicates, unique fragment count)

    • Evaluate read mapping statistics (% mapped, % uniquely mapped)

    • Calculate signal-to-noise ratios (fraction of reads in peaks)

    • Perform cross-correlation analysis to verify enrichment

  • Peak calling considerations:

    • Use appropriate peak calling algorithms (MACS2, SICER) with parameters adjusted for histone modifications

    • Implement stringent fold-enrichment thresholds (typically >4-fold over input)

    • Apply false discovery rate (FDR) correction (q < 0.05 or q < 0.01)

    • Consider broader domains rather than sharp peaks, as histone modifications often span regions

  • Differential binding analysis:

    • Utilize specialized tools (DiffBind, MAnorm, or DESeq2 with appropriate normalization)

    • Apply batch effect correction if samples were processed in different batches

    • Consider quantile normalization to account for global differences in modification levels

  • Biological replication:

    • Implement irreproducible discovery rate (IDR) analysis between replicates

    • Calculate Pearson correlation coefficients between normalized replicate profiles

    • Consider consensus peak sets requiring detection in multiple replicates

  • Integration with other data types:

    • Correlate formylation patterns with transcriptome data

    • Compare with other histone modifications, particularly acetylation at K12

    • Analyze enrichment in different genomic annotations (promoters, enhancers, gene bodies)

How can researchers address potential artifacts in detection of H4K12 formylation during sample preparation?

Formylation can potentially occur as an artifact during sample preparation, particularly during fixation processes. To distinguish genuine biological formylation from technical artifacts:

  • Modified fixation protocols:

    • Compare formaldehyde-fixed samples with alternative fixation methods

    • Implement gradient fixation protocols with varying formaldehyde concentrations

    • Test native (non-crosslinked) sample preparation when possible

  • Chemical protection strategies:

    • Include formylation-quenching agents (e.g., glycine) during sample processing

    • Test the effect of antioxidants in buffers to prevent oxidative formylation

    • Use heavy isotope-labeled reagents to track potential artificial modifications

  • Control samples for background estimation:

    • Process matched samples without fixation steps

    • Include negative controls from organisms or cell types known to lack the specific formylation

    • Implement spike-in normalization with standards containing known formylation levels

  • Orthogonal validation:

    • Confirm findings using alternative detection methods (e.g., mass spectrometry)

    • Validate by manipulating enzymes responsible for formylation/deformylation

    • Compare results from multiple antibody clones or sources

How does the performance of Formyl-HIST1H4A (K12) antibody compare across different application techniques?

Based on available information, the Formyl-HIST1H4A (K12) antibody has been validated for ELISA, Western Blotting, Immunocytochemistry, and Immunofluorescence applications . When comparing performance across these techniques:

ApplicationSensitivitySpecificityKey Optimization FactorsCommon Challenges
ELISAHigh (pg-ng range)Moderate-HighAntibody concentration, blocking conditions, incubation timeCross-reactivity with acetylated K12
Western BlotModerate (ng range)HighTransfer efficiency, blocking conditions, antibody dilutionSize similarity with other histones, detection of free vs. nucleosomal H4
ICCVariableModerateFixation method, permeabilization, antibody penetrationBackground signal, epitope masking by chromatin structure
IFModerate-HighModerateSignal amplification, confocal settings, counterstainingAutofluorescence, signal-to-noise ratio, preservation of nuclear architecture

For optimal results across all applications, consider:

  • Validating specificity using peptide competition assays

  • Including appropriate positive and negative controls

  • Optimizing blocking conditions to minimize background signal

  • Testing multiple antibody concentrations to identify the optimal signal-to-noise ratio

What methodological differences should researchers consider when investigating formylation at H4K12 versus acetylation or other modifications?

When designing experiments to study formylation at H4K12 compared to other modifications like acetylation, implement these methodological considerations:

  • Antibody selection and validation:

    • Confirm specificity against formylated vs. acetylated peptides

    • Test for cross-reactivity with other modified forms of H4K12

    • Consider the influence of neighboring modifications on epitope recognition

  • Sample preparation differences:

    • Formylation may be more sensitive to oxidative conditions during extraction

    • Acetylation is affected by HDAC activity; consider HDAC inhibitors during sample preparation

    • Different fixation protocols may preferentially preserve certain modifications

  • Experimental controls:

    • Include specific enzyme inhibitors:

      • For acetylation: HDAC inhibitors (e.g., TSA, sodium butyrate)

      • For formylation: Relevant enzyme inhibitors if known

    • Consider in vitro modified recombinant histones as standards

    • Compare wild-type samples with mutants where K12 is replaced with non-modifiable residues

  • Data analysis considerations:

    • Formylation may have different genomic distribution patterns than acetylation

    • Temporal dynamics may differ (stability, enzyme kinetics)

    • Correlation with other histone marks may reveal distinct functional contexts

  • Functional validation approaches:

    • Research on K5/K12 acetylation suggests redundancy with K8 in nucleosome assembly

    • Consider similar redundancy testing for formylation

    • Examine the effects under conditions where K5, K8, or other sites are also modified or mutated

When should researchers combine Formyl-HIST1H4A (K12) antibody with other techniques for comprehensive epigenetic analysis?

For comprehensive epigenetic analysis, the Formyl-HIST1H4A (K12) antibody should be combined with other techniques in these research scenarios:

  • When establishing modification patterns across the genome:

    • Combine ChIP-seq using the Formyl-HIST1H4A (K12) antibody with similar analyses of:

      • Other histone modifications (particularly acetylation at K5, K8, K12, K16)

      • Histone variants

      • Chromatin accessibility (ATAC-seq, DNase-seq)

      • DNA methylation patterns

  • When investigating functional consequences:

    • Correlate formylation patterns with:

      • Transcriptome data (RNA-seq)

      • Translation efficiency measurements

      • Protein-DNA interaction maps (other ChIP-seq data)

      • Chromosome conformation capture techniques (Hi-C, 4C)

  • When studying temporal dynamics:

    • Implement time-course experiments combining:

      • ChIP-seq at multiple time points

      • Pulse-chase labeling of histones

      • Live-cell imaging with modification-specific sensors

      • Single-cell approaches to capture heterogeneity

  • When analyzing mechanism:

    • Complement antibody-based detection with:

      • Mass spectrometry to identify co-occurring modifications

      • Enzyme activity assays for relevant writers/erasers

      • Protein-protein interaction studies to identify readers

      • CRISPR screens to identify regulatory factors

The research on H4 K5/K12 acetylation demonstrates that understanding the true biological function requires multiple complementary approaches, including both in vivo and in vitro assays, and consideration of potential redundancy with modifications at other sites .

How might the pattern of H4K12 formylation differ functionally from the well-established K5/K12 diacetylation pattern associated with histone deposition?

While newly synthesized histone H4 is typically diacetylated at K5/K12 in many organisms, research has shown that this specific pattern is not strictly required for nucleosome assembly in yeast . When investigating how H4K12 formylation might function differently:

  • Contextual redundancy: Research has demonstrated that K5, K8, and K12 function redundantly in histone deposition, with assembly strongly impaired only when all three sites are mutated . This suggests that formylation at K12 might similarly participate in redundant regulatory systems, possibly serving as an alternative pathway when acetylation is compromised.

  • Timing differences: Unlike acetylation, which is well-established as occurring on newly synthesized histones, formylation might occur post-incorporation into chromatin, potentially serving regulatory functions beyond deposition.

  • Reader protein specificity: Different modifications recruit distinct reader proteins. While acetylation typically recruits bromodomain-containing proteins, formylation likely engages a different set of reader molecules, potentially activating distinct downstream pathways.

  • Stability and reversibility: The enzymatic machinery for adding and removing formyl groups likely differs from that controlling acetylation, potentially resulting in different kinetic properties and response to cellular signals.

  • Metabolic connections: Formylation may connect histone regulation to specific metabolic pathways distinct from those influencing acetylation, potentially serving as a mechanism to integrate metabolic state with chromatin structure.

Future research should explore these functional differences by comparing the genomic distribution, temporal dynamics, and protein interactions of formylated versus acetylated H4K12.

What experimental approaches would best elucidate whether H4K12 formylation participates in the redundancy observed with K5, K8, and K12 acetylation in nucleosome assembly?

Building on the finding that K5, K8, and K12 function redundantly in histone deposition , a comprehensive experimental strategy to investigate whether formylation at K12 participates in this redundancy would include:

  • Mutation studies with formylation-specific controls:

    • Generate yeast strains with lysine-to-arginine (K→R) mutations that prevent acetylation but potentially allow formylation

    • Compare with lysine-to-glutamine (K→Q) mutations that mimic acetylation

    • Include controls that specifically prevent formylation while permitting acetylation

  • In vivo nucleosome assembly assessment:

    • Measure plasmid superhelicity as an indicator of nucleosome formation in vivo

    • Compare assembly efficiency in strains with different mutation combinations

    • Analyze under conditions that promote or inhibit formylation

  • In vitro reconstitution:

    • Perform nucleosome assembly assays using purified components

    • Compare assembly kinetics and stability using H4 with different modification states:

      • Unmodified at K5, K8, K12

      • Acetylated at various combinations of these sites

      • Formylated at K12 (with or without acetylation at K5/K8)

    • Analyze by native gel electrophoresis, MNase digestion patterns, and biophysical techniques

  • Enzymatic manipulation:

    • Identify and manipulate enzymes responsible for formylation/deformylation

    • Assess the impact on nucleosome assembly when these enzymes are inhibited or overexpressed

    • Examine genetic interactions with known histone acetyltransferases/deacetylases

  • Combined modification analysis:

    • Implement mass spectrometry approaches to quantify the co-occurrence of formylation and acetylation

    • Assess whether formylation increases when acetylation is blocked

    • Determine if there are conditions where formylation becomes critical for assembly

What are the emerging research directions regarding the potential relationship between histone H4K12 formylation and pathological conditions?

While direct evidence linking H4K12 formylation to pathological conditions is limited in the provided search results, several promising research directions emerge based on our understanding of histone modifications and disease:

  • Neurodegenerative disorders:

    • Investigate H4K12 formylation patterns in models of Alzheimer's, Parkinson's, and other neurodegenerative diseases

    • Compare with known dysregulation of histone acetylation in these conditions

    • Assess correlation with cognitive deficits and disease progression

  • Cancer epigenetics:

    • Profile H4K12 formylation across cancer types and stages

    • Correlate patterns with oncogene expression and tumor suppressor silencing

    • Explore potential as a biomarker for specific cancer subtypes or progression states

  • Inflammatory conditions:

    • Examine changes in H4K12 formylation during acute and chronic inflammation

    • Investigate connection to oxidative stress, which can promote formylation

    • Assess relationship with inflammatory gene expression programs

  • Metabolic disorders:

    • Study how metabolic perturbations affect H4K12 formylation levels

    • Investigate in models of diabetes, obesity, and metabolic syndrome

    • Explore connection to mitochondrial dysfunction and altered cellular metabolism

  • Therapeutic targeting:

    • Identify enzymes responsible for adding/removing formyl groups

    • Develop small molecule inhibitors or activators of these enzymes

    • Assess therapeutic potential in relevant disease models

Future studies should employ advanced techniques such as genome-wide profiling of H4K12 formylation in patient samples, CRISPR-mediated manipulation of formylation levels, and development of specific inhibitors/activators for formylation regulatory enzymes.

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.