The Formyl-HIST1H1D (K140) Antibody (PACO65094) is a rabbit-derived polyclonal antibody that specifically recognizes the formylated lysine 140 modification on histone H1.3. This modification is linked to chromatin remodeling and transcriptional regulation, with implications in cellular differentiation and disease mechanisms .
Histone H1.3 (encoded by HIST1H1D) is a linker histone critical for:
Chromatin compaction: Stabilizes higher-order chromatin structures by binding linker DNA .
Transcriptional regulation: Modulates nucleosome spacing and DNA methylation .
Epigenetic signaling: Formylation at K140 alters chromatin accessibility, influencing gene expression in processes like DNA repair and differentiation .
This antibody is pivotal for identifying formylation events associated with:
Chromatin remodeling: Mapping PTMs that regulate chromatin fiber dynamics .
Gene silencing/activation: Correlating formylation with transcriptional states in cancer or developmental models .
Specificity: Validated for minimal cross-reactivity with non-formylated HIST1H1D .
Sensitivity: Detects endogenous levels of formylated H1.3 in immunofluorescence (IF) and ELISA .
Suppliers: Available through Assay Genie (PACO65094), Afigen, and GlycoTech Corporation .
Formats: Liquid formulation with 0.03% Proclin 300 preservative in PBS-glycerol buffer .
Research leveraging this antibody could explore:
Disease mechanisms: Role of HIST1H1D formylation in cancer progression or neurodegenerative disorders.
Therapeutic targeting: Small-molecule inhibitors modulating formylation-dependent chromatin states.
The Formyl-HIST1H1D (K140) Antibody is a polyclonal antibody produced in rabbits that specifically targets the formylated lysine 140 residue of histone H1.3 (HIST1H1D). This antibody serves as an essential tool for researchers investigating post-translational modifications of linker histones and their functional implications.
Histones play a vital role in packaging and organizing DNA within the nucleus, and histone variants like Formyl-HIST1H1D K140 are implicated in regulating various nuclear processes. By targeting this specific modification, researchers can investigate its role in transcriptional regulation, DNA repair mechanisms, chromatin remodeling, and developmental processes . The high specificity of this antibody makes it particularly valuable for studying how formylation of K140 affects chromatin structure and gene expression patterns.
The Formyl-HIST1H1D (K140) Antibody has been extensively characterized with the following specifications and validated applications:
| Parameter | Specification |
|---|---|
| Product Code | PACO65094 |
| Host Species | Rabbit |
| Clonality | Polyclonal |
| Target Species Reactivity | Human |
| Isotype | IgG |
| Antigen | Synthesized peptide derived from Human Histone H1.3 protein (137-149aa) |
| Tested Applications | ELISA, ICC |
| Recommended Dilutions | ICC: 1:10-1:100 |
| Form | Liquid |
| Buffer | Preservative: 0.03% Proclin 300, Constituents: 50% Glycerol, 0.01M PBS, pH 7.4 |
| Purification Method | Antigen Affinity Purified |
The antibody has been validated primarily for ELISA and immunocytochemistry (ICC) applications, allowing researchers to detect the presence and cellular localization of formylated HIST1H1D . Though not explicitly validated in the provided data, experienced researchers often test such antibodies in additional applications such as Western blotting and ChIP assays after appropriate optimization.
HIST1H1D (Histone H1.3) is one of several histone H1 variants that function as linker histones in chromatin organization. Unlike core histones, linker histones like H1 variants bind to DNA between nucleosomes, helping to stabilize higher-order chromatin structures. The key differences between HIST1H1D and other H1 variants include:
Expression patterns: Different H1 variants exhibit tissue-specific and development-specific expression profiles
Protein sequence variations: Particularly in the N-terminal and C-terminal domains
Post-translational modification sites: Each variant has unique modification patterns
Nuclear distribution: Studies using subtype-specific antibodies have shown that H1 variants like H1-2 and H1-4 exhibit distinct punctate staining patterns within the nucleus, suggesting association with specific genome regions
Studying formylation of HIST1H1D at K140 is particularly important because post-translational modifications of histones serve as epigenetic markers that influence chromatin accessibility and gene expression. Formylation is a relatively less-studied modification compared to acetylation or methylation, and understanding its functional significance on HIST1H1D can provide insights into novel regulatory mechanisms in chromatin dynamics and gene regulation . This knowledge contributes to our understanding of both normal cellular processes and disease mechanisms involving epigenetic dysregulation.
Designing robust experimental controls is essential when using the Formyl-HIST1H1D (K140) Antibody in epigenetic research to ensure data validity and interpretability. Critical considerations include:
Antibody specificity controls:
Peptide competition assays: Pre-incubating the antibody with the immunizing peptide should abolish specific signal
Knockout/knockdown verification: Using HIST1H1D-depleted cells should show significantly reduced signal
Comparison with pan-H1 antibodies: To distinguish variant-specific from general H1 patterns
Post-translational modification controls:
Treatment with deformylase enzymes to remove the modification and verify signal specificity
Comparison with antibodies targeting other modifications at the same or nearby residues
Mass spectrometry validation of the formylation site when possible
Cell-type and condition-specific controls:
Include multiple cell types known to express different levels of HIST1H1D
Compare formylation patterns under different cellular conditions that affect chromatin states (proliferation, differentiation, stress responses)
Use synchronized cells to account for cell cycle-dependent variations in histone modifications
Technical controls for specific applications:
For immunofluorescence: Include secondary antibody-only controls and pre-immune serum controls
For ChIP experiments: Include IgG controls and input samples
For Western blots: Include recombinant HIST1H1D proteins with and without the formylation
Implementation of these controls is crucial as they help distinguish between true biological signals and technical artifacts, particularly important when studying subtle epigenetic modifications like formylation that may be present at relatively low abundance in the genome.
Optimizing immunocytochemistry (ICC) protocols for the Formyl-HIST1H1D (K140) Antibody requires careful attention to several parameters:
Fixation optimization:
Test multiple fixatives: While 4% paraformaldehyde is standard, methanol or a combination of paraformaldehyde and glutaraldehyde may better preserve certain histone modifications
Fixation time: Generally 10-15 minutes at room temperature, but this should be empirically determined
Post-fixation washes: Use PBS with 0.1% Triton X-100 to remove excess fixative
Antigen retrieval considerations:
Heat-induced epitope retrieval in citrate buffer (pH 6.0) or Tris-EDTA (pH 9.0)
Enzymatic retrieval using proteinase K may improve accessibility to nuclear antigens
Test different retrieval times (10-30 minutes) to determine optimal conditions
Antibody concentration optimization:
Blocking and permeabilization refinement:
Blocking with 5-10% normal serum from the same species as the secondary antibody
Add 0.2-0.5% Triton X-100 for nuclear permeabilization
Include 0.1% BSA in antibody diluent to reduce non-specific binding
Signal amplification strategies:
Consider tyramide signal amplification for low-abundance targets
Use biotin-streptavidin systems for enhanced sensitivity
Optimize secondary antibody concentration independently
Counterstaining optimization:
Use DAPI for nuclear visualization (1μg/ml for 5 minutes)
Consider additional markers for nuclear compartments to correlate formylation with functional domains
Methodologically, researchers should systematically test each parameter while keeping others constant, documenting the signal-to-noise ratio under each condition. For quantitative analysis, it's advisable to use image analysis software to objectively measure nuclear signal intensity across different conditions.
Investigating the functional significance of HIST1H1D K140 formylation requires a multi-faceted approach combining biochemical, molecular, and cellular techniques:
Chromatin structure analysis:
Micrococcal nuclease (MNase) sensitivity assays to determine if formylation alters chromatin accessibility
ATAC-seq to map genome-wide changes in chromatin accessibility in response to manipulating formylation levels
Hi-C or similar chromosome conformation capture techniques to assess if formylation affects higher-order chromatin organization
Genome localization studies:
Chromatin immunoprecipitation followed by sequencing (ChIP-seq) using the Formyl-HIST1H1D (K140) Antibody to map genomic locations enriched for this modification
Compare with distribution patterns of other histone variants to identify unique vs. shared chromatin domains
Correlate formylation patterns with transcriptionally active/repressive regions
Protein interaction studies:
Identify "readers" of the formyl-K140 mark using techniques like formyl-peptide pulldowns coupled with mass spectrometry
Perform co-immunoprecipitation experiments to identify proteins that specifically interact with formylated HIST1H1D
Use proximity ligation assays to visualize interactions in situ
Functional manipulation approaches:
Generate K140 mutants (K140R or K140Q) that cannot be formylated or mimic formylation
Use CRISPR/Cas9 to introduce these mutations into the endogenous HIST1H1D gene
Assess phenotypic consequences on cell proliferation, differentiation, and stress responses
Transcriptional impact analysis:
Perform RNA-seq in cells with wild-type vs. mutant HIST1H1D to identify affected genes
Use nascent RNA sequencing techniques like NET-seq to directly measure transcriptional changes
Correlate transcriptional changes with altered chromatin accessibility in specific genomic regions
These approaches together can provide comprehensive insights into how formylation of HIST1H1D at K140 contributes to chromatin structure regulation and subsequent effects on nuclear processes like transcription, replication, and DNA repair .
Comparing and integrating data from different histone H1 variant-specific antibodies requires careful methodological considerations to ensure accurate interpretation:
Standardization of experimental conditions:
Use identical sample preparation methods across all antibodies
Process and analyze samples in parallel when possible
Standardize imaging parameters for microscopy or detection settings for blotting techniques
Cross-validation approaches:
Perform reciprocal verification using antibodies raised against different epitopes of the same variant
Validate variant specificity using genetic approaches (overexpression, knockdown)
Use mass spectrometry to independently confirm variant identity and modification status
Data normalization strategies:
Normalize signals to total histone H1 levels using pan-H1 antibodies
Use internal loading controls appropriate for the technique (housekeeping proteins for Western blots, DAPI for immunofluorescence)
Consider global chromatin markers as normalization controls for ChIP experiments
Integrated analysis frameworks:
Create comprehensive maps of different H1 variants' distribution patterns
Use bioinformatic approaches to identify unique and overlapping genomic regions
Perform correlation analyses between different H1 variants and functional genomic elements
Systematic comparison with published datasets:
For visualization and quantitative comparison, researchers should consider data integration tables that summarize findings across different H1 variants:
| Histone Variant | Nuclear Distribution Pattern | Genomic Enrichment | Association with Chromatin States | Cell Cycle Dynamics |
|---|---|---|---|---|
| HIST1H1D (H1.3) | Variable based on formylation status | Gene regulatory regions | Active chromatin when formylated | Cell cycle-dependent |
| H1.0 | Broadly distributed | Heterochromatin | Repressive chromatin | Cell cycle-independent |
| H1.1 | Parallel with DNA concentration | Various genomic regions | Mixed states | Cell cycle-regulated |
| H1.2 | Punctate staining pattern | Specific functional domains | Context-dependent | Cell cycle-regulated |
| H1.4 | Punctate staining pattern | Specific functional domains | Context-dependent | Cell cycle-regulated |
This integrated approach allows researchers to build a comprehensive understanding of how different H1 variants, including formylated HIST1H1D, contribute to chromatin organization and nuclear function .
Although the Formyl-HIST1H1D (K140) Antibody has not been explicitly validated for ChIP applications in the provided data, researchers can adapt standard ChIP protocols with specific considerations for this antibody:
Sample preparation:
Cross-link cells with 1% formaldehyde for 10 minutes at room temperature
Quench with 125mM glycine for 5 minutes
Lyse cells in appropriate buffers to isolate nuclei
Sonicate chromatin to 200-500bp fragments (verify size by gel electrophoresis)
Antibody-specific considerations:
Perform antibody titration experiments (1-10μg per reaction) to determine optimal concentration
Include overnight incubation at 4°C with rotation to maximize antibody-antigen interaction
Use magnetic protein A/G beads for efficient immunoprecipitation of rabbit IgG
Include appropriate controls: IgG control, input sample, and ideally a non-formylatable mutant control
Washing and elution optimization:
Implement stringent washing conditions to reduce background: low-salt, high-salt, LiCl, and TE washes
Elute chromatin complexes with freshly prepared elution buffer (1% SDS, 0.1M NaHCO₃)
Reverse cross-links overnight at 65°C
Treat with RNase A and Proteinase K before DNA purification
Analysis approaches:
qPCR validation at candidate loci before proceeding to genome-wide analysis
For ChIP-seq library preparation, ensure adequate depth of sequencing (>20 million reads)
Use appropriate bioinformatic pipelines for peak calling (MACS2) and annotation (HOMER)
Validation and interpretation strategies:
Confirm enrichment at expected genomic regions based on existing knowledge of H1 variants
Compare with published datasets of histone modifications and chromatin states
Integrate with RNA-seq data to correlate with gene expression levels
For researchers specifically interested in the genome-wide distribution of formylated HIST1H1D, ChIP-seq protocols similar to those used for studying histone H1.0 could be adapted , with additional optimization for the specific characteristics of the Formyl-HIST1H1D (K140) Antibody. Successful ChIP experiments would provide valuable insights into the genomic locations where this modification is present and potentially functioning in chromatin regulation.
Troubleshooting non-specific binding or weak signals with the Formyl-HIST1H1D (K140) Antibody requires a systematic approach addressing several potential issues:
Non-specific binding problems:
Symptom: Multiple bands in Western blot or diffuse nuclear/cytoplasmic staining in ICC
Solutions:
Increase blocking reagent concentration to 5-10% normal serum or BSA
Add 0.1-0.3% Triton X-100 to reduce hydrophobic interactions
Pre-absorb antibody with cell/tissue lysate from non-relevant species
Increase washing stringency with higher salt concentration buffers
Verify antibody specificity with peptide competition assays
Weak signal issues:
Symptom: Faint or barely detectable signal in target applications
Solutions:
Decrease antibody dilution within recommended range (try 1:10 instead of 1:100)
Increase incubation time to overnight at 4°C
Optimize antigen retrieval methods for fixed tissues/cells
Consider signal amplification systems (biotin-streptavidin or tyramide)
Verify target protein expression levels in your samples
Test different detection systems with increased sensitivity
Technical optimization approaches:
For Western blot:
Test different transfer conditions and membrane types
Increase primary antibody incubation time to overnight at 4°C
Use enhanced chemiluminescence (ECL) substrates with higher sensitivity
For ICC/IHC:
Optimize fixation methods to preserve epitope accessibility
Test different permeabilization conditions
Use high-sensitivity fluorescent secondary antibodies
Optimize image acquisition settings (exposure time, gain)
Sample-specific considerations:
Verify that your experimental conditions promote formylation of HIST1H1D
Consider cell cycle synchronization as histone modifications can vary during the cell cycle
Test positive control samples with known high levels of the modification
Antibody storage and handling:
When troubleshooting, it's advisable to change only one parameter at a time and document the results systematically. Creating a troubleshooting decision tree can help guide the optimization process efficiently. Additionally, consulting published protocols for similar histone modification antibodies can provide valuable insights into potential optimization strategies.
Formylation of HIST1H1D at K140 represents one of several post-translational modifications (PTMs) that can occur on histone H1 variants. Comparing this modification with others provides insights into its unique functional significance:
Comparison with phosphorylation:
Phosphorylation of H1 variants (including HIST1H1D at T17) is well-documented to increase during mitosis and associated with chromatin condensation
While phosphorylation is typically dynamic and reversible, formylation may represent a more stable, long-term modification
Functionally, phosphorylation reduces H1's affinity for DNA, whereas formylation's effects on DNA binding remain under investigation
Both modifications may alter the interaction surface of H1 for binding partner proteins
Comparison with methylation and acetylation:
Methylation of H1 variants typically occurs on lysine residues and is associated with transcriptional repression
Acetylation generally promotes chromatin accessibility by neutralizing positive charges
Formylation, like acetylation, neutralizes the positive charge of lysine, potentially reducing DNA binding affinity
Unlike acetylation and methylation, enzymatic "erasers" for formylation are less well-characterized
Site-specific functional impacts:
K140 in HIST1H1D is located in the globular domain, which is crucial for nucleosome binding
Modifications in this region likely alter nucleosome interaction dynamics compared to modifications in the N-terminal or C-terminal tails
The specificity of formylation at K140 suggests a targeted regulatory mechanism rather than global chromatin effects
Evolutionary conservation and specificity:
H1 variants exhibit different degrees of conservation across species
The specificity of formylation at K140 may indicate a specialized function that has been selected for during evolution
Cross-talk between formylation and other modifications may create a complex regulatory code
From a methodological perspective, researchers investigating the unique aspects of HIST1H1D formylation should consider:
Developing mass spectrometry approaches to quantify the stoichiometry of formylation compared to other modifications
Using biochemical assays to directly compare how different modifications alter H1-DNA and H1-nucleosome interactions
Employing genetic approaches to introduce modification-specific mutations and observe phenotypic consequences
Current evidence suggests that formylation of HIST1H1D at K140 may represent a specialized regulatory mechanism that complements the better-characterized modifications like phosphorylation and acetylation, potentially functioning in specific genomic contexts or cellular conditions.
Several cutting-edge technologies and methodological approaches are poised to significantly advance our understanding of HIST1H1D formylation in chromatin biology:
Single-cell epigenomics technologies:
Single-cell ChIP-seq to map formylated HIST1H1D distribution with cellular resolution
Single-cell ATAC-seq to correlate formylation with chromatin accessibility at individual cell level
Single-cell multi-omics approaches that simultaneously measure histone modifications, chromatin accessibility, and gene expression
These approaches would reveal cell-to-cell heterogeneity in formylation patterns not detectable in bulk analyses
Advanced imaging techniques:
Super-resolution microscopy (STORM, PALM) to visualize the nanoscale distribution of formylated HIST1H1D
Live-cell imaging using formylation-specific nanobodies or mintbodies
Correlative light and electron microscopy to relate formylation patterns to ultrastructural features
These techniques would provide spatial context for formylation within the nuclear architecture
Proximity-based mapping approaches:
BioID or APEX2 fusion proteins to identify proteins in proximity to formylated HIST1H1D
ChIP-SICAP to identify proteins that directly interact with formylated HIST1H1D on chromatin
Proximity ligation assays to visualize and quantify interactions between formylated HIST1H1D and other nuclear proteins
These methods would help construct the interaction network surrounding this modification
CRISPR-based technologies:
CRISPR activation/inhibition systems targeting enzymes involved in formylation
Base editing to introduce or remove specific formylation sites
CRISPR screens to identify genes affecting HIST1H1D formylation
These approaches would allow precise manipulation of formylation for functional studies
Advanced biochemical approaches:
Development of formylation-specific "reader" domains for chromatin profiling
Protein engineering to create formylation-specific biosensors
In vitro reconstitution of chromatin with defined formylation status to study biophysical properties
These tools would enable more precise biochemical characterization of formylation effects
Computational and AI-based approaches:
Machine learning algorithms to predict formylation sites and their functional consequences
Molecular dynamics simulations to model how formylation alters H1-nucleosome interactions
Integrative multi-omics data analysis to place formylation in broader epigenetic context
These computational methods would help interpret and predict the functional significance of observed patterns
The integration of these emerging technologies with established approaches would create a comprehensive framework for understanding the role of HIST1H1D formylation in chromatin regulation, potentially revealing new principles of epigenetic control and identifying novel therapeutic targets for diseases involving chromatin dysregulation.
Researchers considering using the Formyl-HIST1H1D (K140) Antibody should keep in mind several key points that will help them design robust experiments and interpret their results accurately:
Antibody characteristics and applications:
The Formyl-HIST1H1D (K140) Antibody is a rabbit polyclonal antibody that specifically recognizes the formylated lysine 140 residue of human histone H1.3 (HIST1H1D)
It has been validated primarily for ELISA and immunocytochemistry applications, with recommended dilutions between 1:10-1:100 for ICC
The antibody recognizes a specific post-translational modification that may be present at relatively low abundance, requiring careful optimization of detection protocols
Experimental design considerations:
Include appropriate controls to verify antibody specificity, including peptide competition assays and non-formylated controls
Optimize fixation and permeabilization methods to ensure nuclear accessibility while preserving the epitope
Consider the biological context of formylation, including cell cycle stage and cellular stress conditions that might affect modification levels
Integration with broader research contexts:
HIST1H1D is one of several histone H1 variants with distinct nuclear distribution patterns , making comparative studies valuable
Different histone variants appear to be concentrated in distinct regions of the nucleus and may be associated with specific genomic regions
The functional significance of formylation should be interpreted in the context of other histone modifications and chromatin regulatory mechanisms
Technical and methodological awareness:
Store the antibody properly at -20°C or -80°C and avoid repeated freeze-thaw cycles
Be aware that optimization may be required for applications beyond those explicitly validated by the manufacturer
Consider that signal intensity may vary depending on the expression level and formylation status of HIST1H1D in different cell types and conditions
Research significance context:
Studying HIST1H1D formylation contributes to our understanding of epigenetic regulation of chromatin structure and gene expression
This research area connects to broader questions in cell biology, development, and disease mechanisms
Findings may have implications for understanding how histone modifications contribute to cellular identity and response to environmental signals
By considering these key points, researchers can effectively incorporate the Formyl-HIST1H1D (K140) Antibody into their experimental workflows, generate reliable data, and contribute meaningful insights to the field of chromatin biology and epigenetics.