Formyl-HIST1H3A (K122) Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Target Specificity

The antibody recognizes the formylated lysine at position 122 of HIST1H3A, a subtype of histone H3. Formylation at this site is distinct from other modifications (e.g., acetylation, methylation) and plays a role in chromatin remodeling. The antibody is raised against a synthetic peptide derived from human HIST1H3A, specifically targeting the formylated K122 residue .

Production Methodology

StepDetailsReferences
Gene CloningInsertion of HIST1H3A antibody-encoding gene into expression vectors.
TransfectionHost cells transfected using polyethyleneimine-mediated methods.
PurificationAffinity chromatography to isolate antibodies.
ValidationELISA and IHC assays confirm specificity for formylated K122.

Key Validation Data

  • Immunoblotting: Detects a 15 kDa band in NIH/3T3 (mouse) and HeLa (human) cell lysates .

  • Peptide Array: Demonstrated high affinity for formylated K122 peptides, with negligible cross-reactivity to unmodified or other modified histones .

Primary Techniques

ApplicationRecommended DilutionKey ObservationsReferences
Immunohistochemistry (IHC)1:50–1:200Nuclear staining in human colon, mouse/rat kidney tissues
ELISANot specifiedQuantification of formylated H3 in cell lysates
Western Blot (WB)1:500–1:2000Detection in human and murine samples

Case Studies

  • Cancer Research: Used to study formylation in breast cancer and other malignancies .

  • Stem Cell Biology: Investigates chromatin states in embryonic stem cells .

Specificity and Cross-Reactivity

SampleObservationMethodReference
Human Colon TissueNuclear staining with formylated K122IHC
Mouse/Rat KidneyNucleus-specific stainingIHC
HeLa Cells15 kDa band in WB, confirming target recognitionWB

Mechanistic Insights

  • Epigenetic Regulation: Formylation at K122 may influence histone-DNA interactions or recruitment of reader proteins .

  • DNA Repair: Potential role in chromatin accessibility during repair processes .

Future Research Directions

  1. Combination with Other Modifications: Study interplay between formylation and acetylation/methylation at K122 .

  2. Cancer Drug Development: Explore formylation as a biomarker for targeted therapies .

  3. Epigenetic Profiling: Map formylation patterns in pluripotent vs. differentiated cells .

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 orders within 1-3 business days of receipt. Delivery times may vary depending on the purchase method and location. Please consult your local distributor for specific delivery timelines.
Synonyms
H3 histone family member E pseudogene antibody; H3 histone family; member A antibody; H3/A antibody; H31_HUMAN antibody; H3F3 antibody; H3FA antibody; Hist1h3a antibody; HIST1H3B antibody; HIST1H3C antibody; HIST1H3D antibody; HIST1H3E antibody; HIST1H3F antibody; HIST1H3G antibody; HIST1H3H antibody; HIST1H3I antibody; HIST1H3J antibody; HIST3H3 antibody; histone 1; H3a antibody; Histone cluster 1; H3a antibody; Histone H3 3 pseudogene antibody; Histone H3.1 antibody; Histone H3/a antibody; Histone H3/b antibody; Histone H3/c antibody; Histone H3/d antibody; Histone H3/f antibody; Histone H3/h antibody; Histone H3/i antibody; Histone H3/j antibody; Histone H3/k antibody; Histone H3/l antibody
Target Names
Uniprot No.

Target Background

Function
Histone H3 is a core component of nucleosomes. Nucleosomes wrap and compact DNA into chromatin, limiting DNA accessibility to cellular machinery that requires DNA as a template. Histones play a crucial role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is regulated by a complex interplay of post-translational modifications of histones, also known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Research suggests that epigenetic regulation in cancer may occur through the induction of E3 ubiquitin ligase NEDD4-dependent histone H3 ubiquitination. PMID: 28300060
  2. Elevated expression of H3K27me3 during a patient's clinical course can be helpful in determining whether tumors are heterochronous. PMID: 29482987
  3. Recent studies have revealed that JMJD5, a Jumonji C (JmjC) domain-containing protein, acts as a Cathepsin L-type protease that mediates histone H3 N-tail proteolytic cleavage under stress conditions inducing a DNA damage response. PMID: 28982940
  4. Data indicate that the Ki-67 antigen proliferative index has significant limitations and phosphohistone H3 (PHH3) presents an alternative proliferative marker. PMID: 29040195
  5. These findings identify cytokine-induced histone 3 lysine 27 trimethylation as a mechanism that stabilizes gene silencing in macrophages. PMID: 27653678
  6. This data indicates that HIST1H3B constitutes the largest proportion of H3.1 transcripts among H3.1 isoforms in the early developing human brain. PMID: 27251074
  7. In a series of 47 diffuse midline gliomas, histone H3-K27M mutation was found to be mutually exclusive with IDH1-R132H mutation and EGFR amplification. It rarely co-occurred with BRAF-V600E mutation and was commonly associated with p53 overexpression, ATRX loss, and monosomy 10. PMID: 26517431
  8. Research demonstrates that histone chaperone HIRA co-localizes with viral genomes, binds to incoming viral and deposits histone H3.3 onto these. PMID: 28981850
  9. These experiments have shown that PHF13 binds specifically to DNA and to two types of histone H3 methyl tags (lysine 4-tri-methyl or lysine 4-di-methyl), where it functions as a transcriptional co-regulator. PMID: 27223324
  10. Hemi-methylated CpGs DNA recognition activates UHRF1 ubiquitylation towards multiple lysines on the H3 tail adjacent to the UHRF1 histone-binding site. PMID: 27595565
  11. This study describes, for the first time, the MR imaging features of pediatric diffuse midline gliomas with histone H3 K27M mutation. PMID: 28183840
  12. Approximately 30% of pediatric high-grade gliomas (pedHGG), including GBM and DIPG, harbor a lysine 27 mutation (K27M) in histone 3.3 (H3.3), which is correlated with poor outcome and was shown to influence EZH2 function. PMID: 27135271
  13. H3F3A K27M mutation in adult cerebellar HGG is not uncommon. PMID: 28547652
  14. Data show that lysyl oxidase-like 2 (LOXL2) is a histone modifier enzyme that removes trimethylated lysine 4 (K4) in histone H3 (H3K4me3) through an amino-oxidase reaction. PMID: 27735137
  15. Histone H3 lysine 9 (H3K9) acetylation was most prevalent when the Dbf4 transcription level was highest, while the H3K9me3 level was greatest during and just after replication. PMID: 27341472
  16. The SPOP-containing complex regulates SETD2 stability and H3K36me3-coupled alternative splicing. PMID: 27614073
  17. These data suggest that binding of the helical tail of histone 3 (H3) with PHD ('plant homeodomain') fingers of BAZ2A or BAZ2B (bromodomain adjacent to zinc finger domain 2A or 2B) requires molecular recognition of secondary structure motifs within the H3 tail and could represent an additional layer of regulation in epigenetic processes. PMID: 28341809
  18. The results demonstrate a novel mechanism by which Kdm4d regulates DNA replication by reducing the H3K9me3 level to facilitate formation of the preinitiation complex. PMID: 27679476
  19. Histone H3 modifications have been observed in leukocytes due to exposures to traffic-derived airborne particulate matter. PMID: 27918982
  20. Persistent histone H3 serine 10 or serine 28 phosphorylation plays a key role in chemical carcinogenesis by regulating gene transcription of DNA damage response genes. PMID: 27996159
  21. hTERT promoter mutations are frequent in medulloblastoma and are associated with older patients, prone to recurrence and located in the right cerebellar hemisphere. However, histone 3 mutations do not appear to be present in medulloblastoma. PMID: 27694758
  22. AS1eRNA-driven DNA looping and activating histone modifications promote the expression of DHRS4-AS1 to economically control the DHRS4 gene cluster. PMID: 26864944
  23. Research suggests that nuclear antigen Sp100C is a multifaceted histone H3 methylation and phosphorylation sensor. PMID: 27129259
  24. The authors propose that histone H3 threonine 118 phosphorylation via Aurora-A alters the chromatin structure during specific phases of mitosis to promote timely condensin I and cohesin disassociation, which is essential for effective chromosome segregation. PMID: 26878753
  25. Hemi-methylated DNA opens a closed conformation of UHRF1 to facilitate its H3 histone recognition. PMID: 27045799
  26. H3K9me3 plays a significant role in hypoxia, apoptosis, and repression of APAK. PMID: 25961932
  27. The authors confirmed that histone H3 is a genuine substrate for GzmA in vivo in Raji cells treated with staurosporin. PMID: 26032366
  28. Circulating H3 levels correlate with mortality in sepsis patients and inversely correlate with antithrombin levels and platelet counts. PMID: 26232351
  29. Data show that double mutations on the residues in the interface (L325A/D328A) decrease the histone H3 H3K4me2/3 demethylation activity of lysine (K)-specific demethylase 5B (KDM5B). PMID: 24952722
  30. MCM2 binding is not required for the incorporation of histone H3.1-H4 into chromatin but is important for the stability of H3.1-H4. PMID: 26167883
  31. Histone H3 lysine methylation (H3K4me3) plays a critical mechanistic role in leukemia stem cell (LSC) maintenance. PMID: 26190263
  32. PIP5K1A modulates ribosomal RNA gene silencing through its interaction with histone H3 lysine 9 trimethylation and heterochromatin protein HP1-alpha. PMID: 26157143
  33. Lower-resolution mass spectrometry instruments can be used for histone post-translational modifications (PTMs) analysis. PMID: 25325711
  34. Inhibition of lysine-specific demethylase 1 activity prevented IL-1beta-induced histone H3 lysine 9 (H3K9) demethylation at the microsomal prostaglandin E synthase 1 (mPGES-1) promoter. PMID: 24886859
  35. De novo CENP-A assembly and kinetochore formation on human centromeric alphoid DNA arrays are regulated by a histone H3K9 acetyl/methyl balance. PMID: 22473132

Show More

Hide All

Database Links

HGNC: 4766

OMIM: 137800

KEGG: hsa:8350

STRING: 9606.ENSP00000444823

UniGene: Hs.132854

Involvement In Disease
Glioma (GLM)
Protein Families
Histone H3 family
Subcellular Location
Nucleus. Chromosome.

Q&A

What is Formyl-HIST1H3A (K122) Antibody and what does it detect?

Formyl-HIST1H3A (K122) Antibody is a specialized immunological reagent that specifically recognizes and binds to histone H3.1 (HIST1H3A) when it contains a formyl group modification at lysine 122. This antibody enables researchers to study this specific post-translational modification of histone proteins . The antibody targets a core component of nucleosomes, which are fundamental units of chromatin structure that wrap and compact DNA, thereby regulating DNA accessibility to cellular machinery involved in transcription, replication, and repair .

The target protein (Histone H3.1) is identified by UniProt ID P68431 and has multiple synonyms including Histone H3/a, Histone H3/b, Histone H3/c, and others . The antibody is typically raised against synthetic peptides derived from human HIST1H3A containing the formylated lysine at position 122, such as the sequence IMP-(Fo)K-DI .

What are the common applications for Formyl-HIST1H3A (K122) Antibody?

Formyl-HIST1H3A (K122) Antibody has been validated for several key research applications:

ApplicationRecommended DilutionNotes
ELISA1:50-1:200For quantitative detection of formylated H3K122
Immunohistochemistry (IHC)1:50-1:200For tissue localization studies
Western Blot (WB)1:500-1:2000For protein expression analysis
Peptide Array (PepArr)Varies by protocolFor epitope mapping and specificity testing

These applications enable researchers to investigate the presence, abundance, and localization of H3K122 formylation in various experimental contexts . The antibody has demonstrated reactivity with human samples, and some versions may cross-react with mouse and rat samples due to the high conservation of histone sequences across species .

What is the biological significance of histone H3.1 formylation at lysine 122?

Lysine 122 is located in a critically important region of histone H3.1, positioned at the nucleosome dyad axis where DNA makes contact with the histone octamer. Formylation at this site has significant implications for chromatin structure and function:

  • Chromatin Structure Modulation: Formylation at K122 can directly affect nucleosome stability by altering the electrostatic interactions between histones and DNA .

  • Transcriptional Regulation: As part of the "histone code," this modification plays a role in regulating gene expression by influencing the accessibility of DNA to transcription factors and other regulatory proteins .

  • Cancer and Disease Associations: Altered patterns of histone formylation have been implicated in various pathological conditions, particularly cancer, where they may contribute to dysregulated gene expression .

Understanding the presence and dynamics of this modification provides insights into fundamental epigenetic mechanisms that control cellular processes and may be dysregulated in disease states .

How should I store and handle Formyl-HIST1H3A (K122) Antibody to maintain its activity?

Proper storage and handling are crucial for maintaining antibody performance:

  • Storage Temperature: Store at -20°C to -70°C for long-term preservation .

  • Avoid Freeze-Thaw Cycles: Minimize repeated freezing and thawing which can degrade antibody quality .

  • Working Aliquots: Upon receipt, divide into small aliquots to prevent repeated freeze-thaw cycles.

  • Buffer Conditions: Formyl-HIST1H3A (K122) Antibodies are typically provided in buffers containing PBS, pH 7.4, with 150mM NaCl and often 50% glycerol as a cryoprotectant .

  • Handling: Work in clean environments to prevent contamination, and use proper aseptic techniques when accessing antibody solutions.

Following these storage and handling guidelines will help ensure consistent experimental results and extend the useful life of the antibody preparation.

How can I validate the specificity of Formyl-HIST1H3A (K122) Antibody in my experimental system?

Comprehensive validation of antibody specificity is essential for generating reliable research data. The following methodological approach is recommended:

  • Peptide Competition Assay: Pre-incubate the antibody with increasing concentrations of the immunizing peptide (formylated K122 peptide) before application in your detection method. Signal reduction confirms specificity for the formylated epitope .

  • Cross-Reactivity Assessment: Test against both formylated and unmodified peptides, as well as peptides with other modifications at K122 (acetylation, methylation) to ensure the antibody discriminates between different modifications .

  • Knockout/Knockdown Controls: When possible, use genetic models where the histone variant is depleted or where enzymes responsible for formylation are knocked out.

  • Mass Spectrometry Correlation: Validate antibody-based detection with mass spectrometry analysis of histone modifications to confirm the presence of formylation at K122.

  • Multiple Antibody Comparison: When available, compare results using different antibody clones (such as comparing monoclonal 32C1 with other antibodies targeting the same modification) .

A comprehensive validation plan incorporates multiple complementary approaches to ensure antibody specificity before proceeding with extensive experimental work.

What are the optimal protocols for immunohistochemistry using Formyl-HIST1H3A (K122) Antibody?

For successful immunohistochemistry with Formyl-HIST1H3A (K122) Antibody, consider the following protocol optimizations:

  • Tissue Preparation:

    • Fix tissues promptly (preferably in 10% neutral buffered formalin)

    • Limit fixation time to prevent epitope masking

    • Use positively charged slides for better tissue adherence

  • Antigen Retrieval:

    • Heat-induced epitope retrieval in citrate buffer (pH 6.0) or EDTA buffer (pH 8.0-9.0)

    • Optimize retrieval time (typically 15-20 minutes at 95-100°C)

  • Blocking and Permeabilization:

    • Block with 5-10% normal serum from the species of the secondary antibody

    • Include 0.1-0.3% Triton X-100 for nuclear permeabilization

  • Antibody Incubation:

    • Dilute antibody 1:50-1:200 in blocking buffer

    • Incubate overnight at 4°C or 1-2 hours at room temperature

    • Include positive controls (tissues known to express formylated H3K122)

    • Include negative controls (primary antibody omission and competitive blocking)

  • Detection System:

    • Use high-sensitivity detection systems (e.g., polymer-based)

    • Optimize counterstaining to visualize nuclear morphology without obscuring specific signals

  • Signal Evaluation:

    • Assess nuclear localization of staining

    • Document staining intensity and distribution patterns

    • Consider digital image analysis for quantification

These methodological details will help researchers achieve optimal staining results when using Formyl-HIST1H3A (K122) Antibody for IHC applications.

How does histone H3K122 formylation relate to other histone modifications in the epigenetic landscape?

Histone H3K122 formylation exists within a complex network of histone modifications that collectively regulate chromatin structure and function:

  • Spatial Context: K122 is located at the nucleosome dyad axis, a region where DNA makes direct contact with the histone octamer. Modifications at this site can directly influence nucleosome stability and DNA accessibility, distinct from modifications on histone tails .

  • Functional Relationship with Other Modifications:

    • Acetylation at K122: While K122 acetylation promotes transcriptional activation by destabilizing nucleosomes, formylation may have distinct effects

    • Crosstalk with Tail Modifications: H3K122 formylation likely functions in concert with modifications on histone tails (e.g., H3K4me3, H3K27ac) to fine-tune gene expression

    • Competitive Modifications: Formylation, acetylation, and other modifications at K122 are mutually exclusive, suggesting regulatory competition

  • Regulatory Enzymes:

    • Unlike well-characterized "writers" and "erasers" for acetylation and methylation, the enzymatic machinery responsible for formylation/deformylation is less well characterized

    • Potential links to metabolic state and oxidative stress have been proposed

  • Temporal Dynamics:

    • Evidence suggests that formylation may represent a more stable modification compared to the more dynamic acetylation, potentially serving as a longer-term epigenetic mark

Understanding how H3K122 formylation integrates with other histone modifications provides crucial context for interpreting experimental results and designing studies to elucidate specific functional consequences.

What research areas are currently exploring H3K122 formylation using this antibody?

Formyl-HIST1H3A (K122) Antibody has been utilized in several cutting-edge research areas:

  • Cancer Biology: Investigations into altered histone formylation patterns in various cancer types, particularly breast cancer, where this modification may contribute to oncogenic gene expression programs .

  • Stem Cell Research: Studies examining the role of histone formylation in maintaining pluripotency and during cellular differentiation processes .

  • Chromatin Architecture: Research exploring how formylation at the nucleosome dyad affects higher-order chromatin structure and nuclear organization.

  • Transcriptional Regulation: Investigations into the impact of H3K122 formylation on RNA polymerase accessibility and activity at specific genomic loci.

  • Metabolic Regulation of Epigenetics: Studies examining how cellular metabolic state influences histone formylation through changes in formyl-donor availability.

  • DNA Damage Response: Research into potential roles of H3K122 formylation in DNA repair processes and genomic stability maintenance.

These diverse research directions highlight the importance of having reliable antibodies against formylated H3K122 to advance understanding of this epigenetic modification's biological significance.

What are effective troubleshooting strategies for experiments using Formyl-HIST1H3A (K122) Antibody?

When encountering challenges with Formyl-HIST1H3A (K122) Antibody experiments, consider the following methodological troubleshooting approaches:

IssuePotential CausesSolutions
Weak or No Signal- Insufficient antibody concentration
- Epitope masking during fixation
- Inadequate antigen retrieval
- Sample degradation
- Optimize antibody concentration (try 1:50 dilution)
- Test different fixation protocols
- Enhance antigen retrieval (extend time, try different buffers)
- Ensure proper sample handling and storage
High Background- Excessive antibody concentration
- Insufficient blocking
- Cross-reactivity
- Non-specific binding
- Increase antibody dilution
- Extend blocking time/concentration
- Add additional washing steps
- Try different blocking reagents (BSA, serum)
Inconsistent Results- Batch-to-batch antibody variation
- Variable fixation conditions
- Biological variation in modification levels
- Use same antibody lot when possible
- Standardize all protocol steps
- Include consistent positive controls
- Normalize to total H3 levels
Unexpected Localization- Cross-reactivity with other formylated proteins
- Detection of alternative histone variants
- Technical artifacts
- Validate with additional antibodies
- Perform peptide competition assays
- Verify with non-antibody methods (e.g., mass spectrometry)
Poor Reproducibility- Variable experimental conditions
- Inconsistent cell/tissue preparation
- Antibody degradation
- Document detailed protocols
- Standardize all reagents and conditions
- Store antibody appropriately
- Include technical replicates

Implementing these targeted troubleshooting strategies can help researchers overcome technical challenges and generate reliable data when working with Formyl-HIST1H3A (K122) Antibody.

How should I design ChIP-seq experiments using Formyl-HIST1H3A (K122) Antibody?

Chromatin immunoprecipitation followed by sequencing (ChIP-seq) with Formyl-HIST1H3A (K122) Antibody requires careful experimental design:

  • Starting Material Optimization:

    • Use 1-5 million cells per immunoprecipitation

    • Ensure high cell viability before fixation

    • Consider cell type-specific optimization (cancer vs. normal cells)

  • Crosslinking and Chromatin Preparation:

    • Optimize formaldehyde concentration (typically 1%) and fixation time (8-10 minutes)

    • Ensure efficient sonication to generate 200-500 bp fragments

    • Verify fragment size distribution by agarose gel electrophoresis

  • Immunoprecipitation Protocol:

    • Pre-clear chromatin with protein A/G beads

    • Determine optimal antibody amount (typically 2-5 μg per IP)

    • Include appropriate controls:

      • IgG negative control

      • Input DNA control

      • Total H3 antibody for normalization

      • Positive control targeting known abundant modifications (e.g., H3K4me3)

  • Library Preparation and Sequencing Considerations:

    • Use spike-in normalization standards for quantitative comparisons

    • Sequence to adequate depth (minimum 20-30 million reads)

    • Include biological replicates (minimum 2-3)

  • Data Analysis Approach:

    • Normalize to input and IgG controls

    • Consider integrative analysis with RNA-seq and other histone marks

    • Analyze genomic distribution relative to transcriptional elements

This methodological framework provides researchers with a starting point for designing rigorous ChIP-seq experiments to investigate the genomic distribution of H3K122 formylation.

What considerations are important when comparing different histone H3 modification antibodies in multiplex experiments?

When designing multiplex experiments to examine relationships between H3K122 formylation and other histone modifications, consider these methodological aspects:

  • Antibody Compatibility Assessment:

    • Verify host species compatibility for multi-color immunofluorescence

    • Test for potential cross-reactivity between antibodies

    • Validate each antibody individually before combining

  • Sequential ChIP Design (Re-ChIP):

    • Consider epitope accessibility in sequential immunoprecipitations

    • Optimize elution conditions between IPs to preserve epitopes

    • Verify recovery efficiency at each step

  • Multiplexed Detection Systems:

    • For imaging applications:

      • Select fluorophores with minimal spectral overlap

      • Include appropriate single-stain controls

      • Perform sequential staining when using same-species antibodies

    • For flow cytometry:

      • Establish compensation controls

      • Validate signal specificity with blocking peptides

      • Consider fixation/permeabilization effects on epitope detection

  • Control for Biological Variables:

    • Account for cell cycle effects on histone modification patterns

    • Consider treatment timing when studying dynamic modifications

    • Include synchronization protocols when appropriate

  • Data Normalization Strategies:

    • Normalize modification-specific signals to total histone H3

    • Use spike-in controls for quantitative comparisons

    • Implement batch correction for experiments performed across multiple days

These methodological considerations enable researchers to generate reliable comparative data on H3K122 formylation in relation to other histone modifications.

How can I quantitatively assess H3K122 formylation levels in different experimental conditions?

For quantitative analysis of H3K122 formylation across experimental conditions, implement these methodological approaches:

  • Western Blot Quantification:

    • Extract histones using acid extraction protocols

    • Separate using SDS-PAGE with 15-18% gels for optimal histone resolution

    • Transfer to PVDF membranes (recommended over nitrocellulose for histones)

    • Probe with Formyl-HIST1H3A (K122) Antibody at 1:500-1:2000 dilution

    • Normalize to total H3 levels using a modification-insensitive H3 antibody

    • Use fluorescent secondary antibodies for wider linear detection range

    • Include standard curves with recombinant histones when possible

  • ELISA-Based Approaches:

    • Develop sandwich ELISA with capture antibody against H3 and detection with Formyl-HIST1H3A (K122) Antibody

    • Generate standard curves using synthetic formylated peptides

    • Normalize to total histone content

    • Consider commercial histone modification ELISA kits with compatible antibodies

  • Mass Spectrometry Analysis:

    • Perform histone propionylation to improve peptide properties for LC-MS/MS

    • Use antibody-based enrichment before MS analysis for low-abundance modifications

    • Implement parallel reaction monitoring (PRM) or selected reaction monitoring (SRM) for targeted quantification

    • Calculate modification stoichiometry by comparing modified to unmodified peptides

  • Imaging-Based Quantification:

    • Standardize image acquisition parameters across all samples

    • Implement nuclear segmentation for single-cell analysis

    • Normalize formylation signal to DAPI or total H3 staining

    • Use automated image analysis pipelines to reduce bias

These quantitative methodologies provide researchers with reliable approaches to measure changes in H3K122 formylation levels under different experimental conditions.

What is known about the relationship between histone H3K122 formylation and chromatin remodeling?

The relationship between H3K122 formylation and chromatin remodeling represents an emerging area of research:

  • Structural Implications:

    • H3K122 is located at the nucleosome dyad axis where DNA makes crucial contacts with the histone octamer

    • Formylation neutralizes the positive charge of lysine, potentially weakening histone-DNA interactions

    • This modification may create a more permissive chromatin structure for remodeling enzyme access

  • Interplay with Chromatin Remodeling Complexes:

    • Emerging evidence suggests that H3K122 formylation may influence the recruitment or activity of ATP-dependent chromatin remodeling complexes

    • The precise mechanisms of this interaction remain to be fully elucidated through biochemical and structural studies

  • Functional Consequences:

    • Altered nucleosome positioning and stability

    • Modified higher-order chromatin structure

    • Potentially increased accessibility for transcription factors and the transcriptional machinery

  • Methodological Approaches to Study This Relationship:

    • In vitro nucleosome reconstitution with formylated H3K122

    • Nucleosome remodeling assays comparing unmodified vs. formylated templates

    • Genome-wide nucleosome mapping in contexts with altered H3K122 formylation

These insights provide a foundation for designing experiments to further elucidate the functional significance of H3K122 formylation in chromatin dynamics and gene regulation.

How can I design experiments to investigate the writers and erasers of H3K122 formylation?

Unlike many histone modifications with well-characterized enzymatic machinery, the specific writers and erasers of H3K122 formylation remain less defined. Consider these experimental approaches:

  • Candidate Enzyme Screening:

    • Generate a list of potential formyltransferases based on structural similarities to known histone-modifying enzymes

    • Perform siRNA/CRISPR knockdown screens of candidate enzymes

    • Quantify changes in H3K122 formylation levels using the antibody in Western blot or immunofluorescence assays

  • Metabolic Manipulation Experiments:

    • Modulate cellular one-carbon metabolism (potential source of formyl groups)

    • Trace experiments with isotope-labeled metabolic precursors

    • Monitor H3K122 formylation levels under various metabolic states

  • Mass Spectrometry-Based Enzyme Discovery:

    • Implement affinity purification using modified and unmodified H3K122 peptides

    • Identify differentially bound proteins by mass spectrometry

    • Validate candidates through biochemical assays

  • In Vitro Enzymatic Assays:

    • Develop assays using recombinant histone H3 as substrate

    • Test candidate enzymes for formyltransferase or deformylase activity

    • Confirm site specificity using mutated histones (K122A, K122R)

  • Temporal Dynamics Analysis:

    • Establish cell systems with inducible expression of tagged histones

    • Track modification appearance and disappearance kinetics

    • Correlate with cellular conditions or stimuli

These systematic approaches provide a framework for identifying and characterizing the enzymatic machinery responsible for regulating H3K122 formylation.

What are the latest findings on the role of H3K122 formylation in disease pathogenesis?

Research into the role of H3K122 formylation in disease contexts is still developing, with several emerging areas of investigation:

  • Cancer Associations:

    • Altered H3K122 formylation patterns have been observed in various cancer types, particularly breast cancer

    • Studies suggest potential associations with oncogenic gene expression programs

    • Initial evidence links formylation changes to tumor progression and metastatic potential

  • Inflammatory Conditions:

    • Emerging evidence suggests connections between oxidative stress, inflammation, and histone formylation

    • Changes in one-carbon metabolism during inflammation may influence formylation levels

    • Potential role in inflammatory gene regulation programs

  • Neurodegenerative Disorders:

    • Preliminary studies exploring connections between altered histone formylation and neurodegenerative processes

    • Potential links to mitochondrial dysfunction and oxidative stress in neuronal cells

  • Metabolic Disorders:

    • Initial investigations into how metabolic dysregulation affects histone formylation patterns

    • Possible connections to insulin resistance and diabetes pathogenesis

  • Research Methodologies in Disease Contexts:

    • Patient-derived tissue analysis using Formyl-HIST1H3A (K122) Antibody

    • Integration of genomic and epigenomic data from disease cohorts

    • Disease model systems to study functional consequences of altered formylation

This evolving understanding provides opportunities for researchers to explore H3K122 formylation as both a biomarker and potential therapeutic target in various disease contexts.

What are appropriate positive and negative controls when using Formyl-HIST1H3A (K122) Antibody?

Implementing robust controls is essential for reliable interpretation of results with Formyl-HIST1H3A (K122) Antibody:

Positive Controls:

  • Cell Line Standards:

    • Cell lines with known high levels of H3K122 formylation (based on literature)

    • Cells treated with inducers of histone formylation (e.g., oxidative stress agents)

  • Recombinant/Synthetic Standards:

    • Formylated H3K122 peptides (such as the immunogen peptide)

    • Formylated recombinant histone H3

    • Control protein microarrays containing formylated and unmodified peptides

  • Tissue Standards:

    • Tissues with documented H3K122 formylation (based on literature)

    • Serial sections of positive control tissues for IHC optimization

Negative Controls:

  • Antibody Controls:

    • Primary antibody omission

    • Isotype control antibody (matching host species and isotype)

    • Pre-immune serum (for polyclonal antibodies)

  • Peptide Competition:

    • Antibody pre-incubated with excess formylated H3K122 peptide

    • Comparison with unformylated peptide pre-incubation

  • Genetic Controls:

    • Cells with H3.1 knockdown/knockout (when available)

    • K122 mutant constructs (K122A, K122R) expressed in cells

  • Technical Controls:

    • Secondary antibody-only controls

    • Blocking peptide controls

    • Cross-reactivity assessment with other modified peptides

Implementing these control strategies provides crucial validation of signal specificity and technical reliability in experiments using Formyl-HIST1H3A (K122) Antibody.

How do different sample preparation methods affect the detection of H3K122 formylation?

Sample preparation significantly impacts the detection of histone modifications, including H3K122 formylation:

  • Fixation Effects:

    • Formaldehyde Fixation: Common for IHC and IF, but may mask epitopes; requires optimization of fixation time (typically 10-15 minutes)

    • Methanol Fixation: Preserves some histone epitopes better than formaldehyde but can cause protein precipitation

    • Fresh-Frozen Samples: May preserve native epitopes but compromise morphology

    • FFPE Tissues: Require robust antigen retrieval methods to expose the H3K122 formylation epitope

  • Antigen Retrieval Methods:

    • Heat-Induced Epitope Retrieval (HIER): Most effective for histone modifications in FFPE tissues

    • Citrate Buffer (pH 6.0): Standard starting point for histone modifications

    • EDTA Buffer (pH 8-9): Alternative when citrate buffer yields suboptimal results

    • Enzymatic Retrieval: Generally less effective for histone modifications

  • Histone Extraction Protocols:

    • Acid Extraction: Gold standard for histone preparation in biochemical analyses

    • Triton Extraction: Preserves nuclear architecture for imaging applications

    • Direct Lysis: May result in lower sensitivity due to dilution of histones in whole cell lysates

  • Storage Considerations:

    • Freeze-Thaw Effects: Multiple cycles can degrade histone modifications

    • Long-term Storage: Stability of formylation during prolonged storage requires investigation

    • Stability in Paraffin Blocks: Epitope may deteriorate in very old archived samples

Optimizing sample preparation protocols is essential for reliable detection of H3K122 formylation and should be carefully considered in experimental design.

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.