The formyl-HIST1H3A (K122) recombinant monoclonal antibody generation typically starts with the insertion of the HIST1H3A antibody-encoding gene into expression vectors. These vectors are subsequently delivered into host cells through polyethyleneimine-mediated transfection methods. The host cells containing these vectors are cultured to produce and release the antibodies. After purification using affinity chromatography, the antibodies undergo testing through ELISA and IHC assays to confirm their recognition of the human HIST1H3A protein formylated at K122.
The generation of formyl-HIST1H3A (K122) recombinant monoclonal antibody typically involves the insertion of the HIST1H3A antibody-encoding gene into expression vectors. These vectors are then delivered into host cells using polyethyleneimine-mediated transfection methods. The host cells containing these vectors are cultured to produce and release the antibodies. Following purification using affinity chromatography, the antibodies are rigorously tested through ELISA and IHC assays to confirm their specific recognition of the human HIST1H3A protein formylated at K122.
Formyl-HIST1H3A (K122) antibody specifically recognizes histone H3.1 that has been formylated at lysine residue 122. Histone H3.1 is a core component of nucleosomes, which wrap and compact DNA into chromatin, thereby regulating DNA accessibility to cellular machinery involved in transcription, replication, and repair processes . The antibody targets the peptide sequence surrounding the formyl-Lys(122) site derived from Human Histone H3, with UniProt ID P68431 . This specific post-translational modification plays a critical role in epigenetic regulation and has implications in various research areas including cancer biology and stem cell research .
Both monoclonal and polyclonal versions of Formyl-HIST1H3A (K122) antibodies are available, each with distinct characteristics:
The monoclonal antibody (such as clone 32C1) was raised against a specific peptide IMP-(Fo)K-DI formylated at K122 , providing highly reproducible results with minimal batch variation. The polyclonal version recognizes multiple epitopes around the K122 formylation site, potentially offering higher sensitivity but with greater batch-to-batch variation .
To maintain optimal antibody activity, the following storage recommendations should be followed:
Avoid repeated freeze-thaw cycles by dividing into single-use aliquots
The antibody is typically supplied in buffers containing glycerol (50%), PBS (pH 7.4), and preservatives
Some formulations contain 0.03% Proclin 300 as a preservative , while others may contain 0.035% sodium azide , which is highly toxic and should be handled with appropriate precautions.
The Formyl-HIST1H3A (K122) antibody has been validated for multiple experimental applications with different recommended protocols:
While the antibody has been validated for the above applications, researchers should perform their own validation with appropriate controls when using the antibody in new experimental systems or conditions .
For optimal detection of formylated HIST1H3A at K122, sample preparation is critical:
For cellular/tissue extracts (Western blot):
Use acid extraction methods (such as HeLa acid extract) to efficiently isolate histones
Include protease inhibitors and deacetylase inhibitors to prevent modification loss
Add formylation stabilizing agents if available to preserve the modification
Maintain cold temperature throughout extraction to minimize enzymatic activity
For fixed samples (IF/IHC):
Optimal fixation with 4% paraformaldehyde preserves epitope accessibility
Permeabilization conditions should be optimized (typically 0.1-0.5% Triton X-100)
Antigen retrieval methods may be necessary for tissue sections (citrate or EDTA buffer)
Blocking with appropriate agents (BSA, normal serum) to reduce background
For all applications:
Positive controls:
Negative controls:
Primary antibody omission
Isotype control (non-specific rabbit IgG)
Samples treated with deformylase enzymes
Peptide competition assay using the immunizing peptide
Specificity controls:
Comparison with antibodies recognizing total H3
Use of genetic knockouts/knockdowns where feasible
Peptide competition with formylated versus non-formylated peptides
Heterogeneous staining patterns in immunofluorescence using Formyl-HIST1H3A (K122) antibody may occur for several research-relevant reasons:
Cell cycle-dependent formylation: Histone modifications can vary throughout the cell cycle, particularly during DNA replication and mitosis. Cells in different cycle phases may show differential formylation patterns .
Transcriptional activity variation: Since histone modifications regulate gene expression, heterogeneity may reflect differential transcriptional states among cells.
Newly incorporated versus existing histones: The antibody may preferentially recognize newly incorporated formylated H3.1 versus older histone proteins . This is particularly relevant as one researcher specifically asked whether the antibody recognizes only newly incorporated H3.1 .
Technical factors:
Incomplete permeabilization
Inconsistent fixation
Variable antibody penetration
Epitope masking due to chromatin compaction differences
To address this heterogeneity, researchers should:
Synchronize cells if cell cycle effects are suspected
Compare staining patterns with total H3 antibodies
Optimize fixation and permeabilization protocols
Consider chromatin decompaction methods if accessibility is an issue
To optimize signal-to-noise ratio in Western blots using Formyl-HIST1H3A (K122) antibody:
Sample preparation optimization:
Use acid extraction methods specifically designed for histones
Include appropriate modification-preserving inhibitors
Consider enrichment of histone fractions before loading
Blocking optimization:
Test different blocking agents (5% non-fat milk, 3-5% BSA)
Optimize blocking time (typically 1-2 hours at room temperature)
Consider specialized blocking reagents for phospho-specific antibodies
Antibody incubation parameters:
Test different dilutions (starting with manufacturer recommendations)
Optimize incubation time and temperature (4°C overnight versus room temperature)
Add 0.05-0.1% Tween-20 to reduce background
Washing optimization:
Increase number and duration of washes (typically 3-5 washes of 5-10 minutes each)
Use appropriate wash buffer (TBST or PBST with 0.05-0.1% Tween-20)
Detection method considerations:
Compare ECL substrates of different sensitivities
Consider fluorescent secondary antibodies for greater linear range
Optimize exposure time to prevent overexposure
Lysine 122 formylation (K122fo) in histone H3.1 represents an important but less-studied post-translational modification with significant implications for epigenetic regulation:
Structural implications: K122 is located in the globular domain of H3, at the interface between DNA and the histone octamer. Formylation at this site likely alters DNA-histone interactions, potentially affecting nucleosome stability and chromatin accessibility .
Functional consequences:
Altered transcriptional regulation due to changed DNA-histone affinity
Potential modulation of other histone modifications in the vicinity
Possible recruitment of specific reader proteins that recognize formylated lysine
Interplay with other modifications: K122 formylation may affect or be affected by other nearby modifications, creating a complex regulatory network within the histone code. This includes potential crosstalk with acetylation and methylation marks.
Biological contexts: Formylation at K122 may play roles in:
Further research is needed to fully elucidate the writers (enzymes that add formyl groups), erasers (enzymes that remove them), and readers (proteins that recognize formylated lysines) in this pathway.
Formyl-HIST1H3A (K122) has emerging significance in cancer research with several key applications:
Biomarker potential: Altered formylation patterns may serve as cancer biomarkers. The antibody enables detection of these alterations in patient samples using IHC or other diagnostic techniques .
Epigenetic dysregulation: Cancer cells often display epigenetic abnormalities. Studying K122 formylation may reveal specific mechanisms of gene expression dysregulation in tumor development .
Research applications in oncology:
Comparing formylation levels between normal and tumor tissues
Correlating formylation patterns with clinical outcomes
Studying changes in formylation during treatment response
Investigating drug resistance mechanisms related to histone modifications
Therapeutic implications: Understanding the role of K122 formylation may reveal:
Novel druggable targets in the formylation/deformylation pathways
Potential for combination therapies targeting formylation alongside other treatments
Predictive biomarkers for treatment response to epigenetic therapies
Current research indicates interest in breast cancer applications specifically, as noted in the antibody product specifications , suggesting particular relevance in this cancer type.
Integration of Formyl-HIST1H3A (K122) antibody into multi-omics research frameworks enables comprehensive understanding of the biological significance of this modification:
Epigenomics approaches:
ChIP-seq to map genomic distribution of K122 formylation
CUT&RUN or CUT&Tag for higher resolution localization
Integration with other histone modification maps to understand the histone code
Proteomics integration:
Immunoprecipitation followed by mass spectrometry (IP-MS) to identify proteins interacting with formylated H3K122
Proximity labeling approaches to capture the molecular neighborhood of formylated histones
Analysis of co-occurring modifications using specialized MS approaches
Transcriptomics correlation:
Integration of formylation ChIP-seq data with RNA-seq to correlate with gene expression patterns
Single-cell approaches to understand heterogeneity in formylation and expression
Functional genomics:
CRISPR screens targeting enzymes involved in formylation/deformylation
Perturbation studies altering metabolic pathways that influence formylation levels
Suggested experimental workflow:
Phase | Approach | Outcome |
---|---|---|
Discovery | ChIP-seq with Formyl-HIST1H3A (K122) antibody | Genome-wide K122fo distribution |
Integration | Correlate with RNA-seq, other histone marks | Functional associations |
Validation | Site-directed mutagenesis, enzyme perturbation | Causal relationships |
Mechanism | IP-MS, reader domain screening | Molecular partners |
Function | Phenotypic assays after perturbation | Biological significance |
This integrated approach would provide a comprehensive understanding of the biological role of H3K122 formylation beyond what single-technique approaches could achieve.
The production of Formyl-HIST1H3A (K122) recombinant monoclonal antibodies involves a sophisticated multi-step process:
Immunization and gene extraction: Rabbits are immunized with a synthetic peptide containing formylated K122 derived from human HIST1H3A. Genes encoding antibodies specific to this modification are then extracted from the rabbit immune cells .
Recombinant expression: The extracted antibody genes are integrated into specialized expression vectors, which are then introduced into host suspension cells .
Culture and production: The modified host cells are cultured under optimized conditions to stimulate expression and secretion of the antibody .
Purification: Affinity chromatography techniques are employed to isolate the antibody from the cell culture supernatant . This typically involves protein A/G affinity purification followed by additional purification steps if needed.
Quality control: The purified antibody undergoes comprehensive validation through multiple assays including ELISA, Western blotting, immunohistochemistry, and immunofluorescence to confirm specificity and functionality .
This recombinant approach offers advantages over traditional hybridoma-based monoclonal antibody production, including better reproducibility, reduced batch-to-batch variation, and elimination of hybridoma instability issues.
Understanding the buffer composition is essential for optimal antibody handling and application compatibility:
When designing experiments, researchers should consider buffer components that might interfere with particular applications. For example:
Sodium azide can inhibit peroxidase activity in HRP-based detection systems
High glycerol content may affect loading in some applications
Salt concentration may need adjustment for certain enzymatic reactions