The HIST1H1C (Ab-62) Antibody recognizes the Lys62 site of histone H1.2, a linker histone critical for chromatin compaction and gene expression regulation . Unlike acetylation-specific variants (e.g., acLys62), this antibody targets the non-acetylated form of Lys62, enabling studies of histone dynamics in diverse cellular contexts .
This antibody is validated for multiple techniques:
The antibody has been instrumental in uncovering HIST1H1C’s roles in pathophysiology:
In diabetic rodent retinas, HIST1H1C overexpression correlates with increased autophagy, inflammation, and neuron loss . The antibody confirmed these findings via WB and IHC:
Autophagy flux: HIST1H1C knockdown reduced LC3B-II conversion and SQSTM1 degradation .
Inflammation: Overexpression upregulated Ccl2 and Il6 mRNAs, validated by qPCR .
HIST1H1C interacts with IRF3 to regulate interferon-β (IFN-β) production . Using the Ab-62 antibody:
HIST1H1C (Ab-62) Antibody is a polyclonal antibody raised in rabbits that specifically recognizes the acetylated lysine at position 62 (acLys62) of the Histone Cluster 1, H1c protein (HIST1H1C), also known as Histone H1.2. The antibody is typically generated using a peptide sequence surrounding the acetylated Lys62 site derived from human Histone H1.2 as the immunogen . This site-specific recognition makes it particularly valuable for studying post-translational modifications of histone proteins in epigenetic research. The antibody is generally supplied in an unconjugated form and purified through antigen affinity methods to ensure specificity .
The HIST1H1C (Ab-62) Antibody has been validated for multiple experimental applications in molecular biology and cell research. These applications include:
| Application | Validated | Recommended Dilution |
|---|---|---|
| ELISA | Yes | Variable by manufacturer |
| Immunofluorescence (IF) | Yes | 1:50-1:500 |
| Chromatin Immunoprecipitation (ChIP) | Yes | Variable by protocol |
| Immunocytochemistry (ICC) | Yes | Variable by protocol |
The antibody shows specific reactivity with human samples across these applications . When designing experiments, it's important to note that optimal dilutions may need to be determined empirically for each specific experimental setup and sample type.
HIST1H1C, or Histone H1.2, is a linker histone that plays crucial roles in chromatin architecture and gene expression regulation. It binds to linker DNA between nucleosomes, facilitating the formation of higher-order chromatin structures known as chromatin fibers . Through this structural role, HIST1H1C contributes to genome compaction and organization.
Beyond its structural function, HIST1H1C also serves as a regulator of gene transcription through multiple mechanisms, including:
Recent research has revealed that while H1 variants are generally distributed across the genome, H1.2 (HIST1H1C) exhibits specific distribution patterns, particularly at promoter regions, suggesting specialized regulatory functions distinct from other H1 variants .
HIST1H1C (H1.2) is one of several somatic H1 histone variants that exist in human cells. Research has demonstrated that while all H1 variants share the fundamental function of binding to linker DNA between nucleosomes, they exhibit distinct genomic distribution patterns and potentially specialized functions:
Notably, mapping studies in breast cancer cells have uncovered specific features for H1.2 distribution both at promoters and genome-wide that distinguish it from other H1 variants . This suggests that H1.2 may play unique roles in gene regulation and chromatin organization in specific cellular contexts.
The acetylation of lysine 62 in HIST1H1C represents an important post-translational modification that can significantly affect the protein's function. While the specific impacts of this modification are still being investigated, histone acetylation generally:
Reduces the positive charge of histones, potentially weakening their interaction with negatively charged DNA
Creates binding sites for proteins containing bromodomains that specifically recognize acetylated lysines
Contributes to a more open chromatin structure that is typically associated with active gene transcription
The HIST1H1C (Ab-62) Antibody specifically recognizes this acetylated form, making it a valuable tool for investigating how this particular modification correlates with chromatin states and gene expression patterns . Understanding the dynamics of Lys62 acetylation can provide insights into regulatory mechanisms governing chromatin accessibility and transcriptional control.
For optimal ChIP experiments using HIST1H1C (Ab-62) Antibody, careful attention to protocol optimization is essential:
Crosslinking Conditions: Standard formaldehyde crosslinking (1% for 10 minutes at room temperature) works for most histone ChIP applications, but H1 histones may benefit from an optimized crosslinking time due to their more dynamic association with DNA.
Chromatin Fragmentation: Aim for fragments between 200-500bp for optimal resolution. This can be achieved through:
Sonication: 10-15 cycles (30 seconds on/30 seconds off) at medium power
Enzymatic digestion: Using micrococcal nuclease with titrated concentrations
Antibody Concentration: Start with 2-5 μg of HIST1H1C (Ab-62) Antibody per ChIP reaction and optimize based on signal-to-noise ratio.
Washing Stringency: Include high-salt washes (up to 500mM NaCl) to reduce non-specific binding, but determine optimal washing conditions empirically as excessive stringency may reduce signal.
Controls: Always include:
Input chromatin (pre-immunoprecipitation sample)
IgG antibody control (same host species as the HIST1H1C antibody)
Positive control (antibody against a histone mark known to be present in your samples)
When analyzing ChIP data, compare the distribution of HIST1H1C with other H1 variants or core histones to understand its specific enrichment patterns, especially given that research has demonstrated specific genomic distribution patterns for H1.2 compared to other H1 variants .
Rigorous validation of antibody specificity is crucial for reliable experimental results. For HIST1H1C (Ab-62) Antibody, consider implementing these validation approaches:
Peptide Competition Assay: Pre-incubate the antibody with:
Acetylated Lys62 peptide (should abolish signal)
Unmodified peptide (should not affect signal)
Peptides with other modifications at Lys62 (should not affect signal)
Knockdown/Knockout Controls:
Use siRNA/shRNA to reduce HIST1H1C expression
Use CRISPR/Cas9 to generate HIST1H1C knockout cell lines
Compare antibody signal between wild-type and manipulated cells
Recombinant Protein Controls:
Test reactivity against recombinant HIST1H1C with and without Lys62 acetylation
Include closely related H1 variants to assess cross-reactivity
Mass Spectrometry Validation:
Perform IP followed by mass spectrometry
Confirm the presence of HIST1H1C and the Lys62 acetylation modification
Check for potential cross-reactive proteins
Multiple Antibody Comparison:
Compare with other validated HIST1H1C antibodies targeting different epitopes
Use antibodies from different manufacturers or raised in different host species
Results should demonstrate specific detection of the acetylated form at the expected molecular weight (observed molecular weight may be 32-33 kDa, though the calculated molecular weight is 21 kDa, due to the influence of post-translational modifications and the highly charged nature of histones on gel migration) .
When facing contradictory results regarding HIST1H1C genomic distribution, consider these methodological approaches to resolve discrepancies:
Technical Considerations:
Compare fixation methods (formaldehyde vs. DSG+formaldehyde for more stable crosslinking)
Assess chromatin preparation methods (sonication vs. enzymatic digestion)
Evaluate antibody specificity (using validation methods described in FAQ 2.2)
Consider sequencing depth requirements for detecting H1 enrichment
Analytical Approaches:
Apply multiple peak calling algorithms and compare results
Use spike-in normalization to control for technical variation
Implement more sensitive normalization methods for H1 histones, which may show broader distribution patterns
Biological Variables:
Document cell cycle stage (H1 binding patterns may vary throughout the cell cycle)
Account for cell type specificity (H1.2 distribution patterns may differ between cell types)
Consider the impact of chromatin compaction states in different experimental systems
Orthogonal Validation:
Research has shown that while H1 variants occur across the genome, H1.2 exhibits specific features both at promoters and genome-wide . Contradictory results may reflect biological realities rather than technical issues, as HIST1H1C distribution may genuinely differ between cell types or physiological conditions.
The acetylation of lysine 62 in HIST1H1C introduces significant functional consequences for chromatin structure and protein interactions:
Chromatin Binding Dynamics:
Acetylation neutralizes the positive charge at Lys62, potentially reducing the electrostatic interaction with negatively charged DNA
This modification may increase the mobility and exchange rate of H1.2 on chromatin
Studies suggest acetylated H1.2 associates preferentially with more accessible, transcriptionally active chromatin regions
Protein-Protein Interactions:
Acetylated Lys62 creates a binding platform for bromodomain-containing proteins
This modification may disrupt interactions with repressive chromatin modifiers
It may facilitate recruitment of chromatin remodeling complexes that promote more open chromatin structures
Relationship to Other PTMs:
The presence of Lys62 acetylation may influence other nearby modifications on H1.2
The interplay between acetylation and other modifications (methylation, phosphorylation) creates a complex regulatory code
Functional Outcomes:
Gene expression changes associated with Lys62 acetylation likely reflect altered chromatin compaction
The modification may influence higher-order chromatin structure formation
Preliminary research suggests connections to specific transcriptional programs
While the physical properties of charged disordered regions in H1 are likely crucial for processes involving liquid-liquid phase separation , the specific impact of Lys62 acetylation on these properties remains an active area of investigation. The HIST1H1C (Ab-62) Antibody serves as a critical tool for further elucidating these functional relationships.
Distinguishing between highly similar H1 variants requires specialized methodological approaches:
Variant-Specific Antibody Selection:
Use antibodies targeting unique regions or specific modifications like the HIST1H1C (Ab-62) Antibody for acetylated Lys62
Validate antibody specificity against all H1 variants using recombinant proteins
Consider developing custom antibodies for highly specific epitopes
Mass Spectrometry-Based Identification:
Employ targeted proteomics methods like parallel reaction monitoring (PRM)
Utilize variant-specific peptides with unique sequences for identification
Quantify relative abundance of different variants using labeled peptide standards
Genetic Engineering Approaches:
Express tagged versions of specific H1 variants (e.g., HA-tagged H1.2)
Use CRISPR/Cas9 to introduce endogenous tags or create variant-specific knockouts
Implement variant-specific RNA interference for functional studies
Chromatin Distribution Analysis:
Compare genome-wide distribution using ChIP-seq with variant-specific antibodies
Look for variant-enriched regions that distinguish H1.2 from other variants
Analyze co-localization with specific chromatin marks or genomic features
Biophysical Characterization:
Research comparing distribution patterns of different H1 variants has revealed that while all variants are found throughout the genome, H1.2 (HIST1H1C) shows specific distribution patterns at promoters and genome-wide that distinguish it from other variants . These unique patterns can be leveraged to develop more precise experimental approaches.
Researchers often encounter specific challenges when using HIST1H1C (Ab-62) Antibody for immunofluorescence studies:
High Background Signal:
Poor Signal Intensity:
Problem: Weak or undetectable signal from acetylated HIST1H1C
Solutions:
Optimize fixation (4% paraformaldehyde for 10-15 minutes)
Include antigen retrieval step (10mM sodium citrate buffer, pH 6.0)
Pre-treat cells with HDAC inhibitors (e.g., TSA, sodium butyrate) to preserve acetylation
Increase antibody incubation time (overnight at 4°C)
Inconsistent Nuclear Localization:
Problem: Variable or unexpected subcellular distribution
Solutions:
Use gentle permeabilization conditions to preserve nuclear structure
Co-stain with DAPI or other nuclear markers for precise localization
Compare with distribution of other histone proteins as controls
Verify cell cycle stage, as H1 distribution may vary throughout the cell cycle
Epitope Accessibility Issues:
Problem: Antibody cannot access acetylated Lys62 in compact chromatin
Solutions:
Implement more stringent permeabilization (0.5% Triton X-100 for 10 minutes)
Test different fixatives (methanol vs. paraformaldehyde)
Consider enzymatic antigen retrieval methods
Use thinner tissue sections (5μm or less) for tissue samples
For optimal results, follow validated protocols with recommended dilutions (1:50-1:500 for IF/ICC applications) and include appropriate positive controls (cell lines known to express acetylated HIST1H1C) and negative controls (blocking peptide, HIST1H1C knockdown cells).
Detecting acetylated HIST1H1C by Western blot requires specific optimization strategies:
Sample Preparation Considerations:
Harvest cells directly in SDS lysis buffer containing HDAC inhibitors (5mM sodium butyrate, 1μM TSA)
Include phosphatase inhibitors to preserve all PTM states
Add protease inhibitors to prevent degradation
Use either acid extraction methods (specialized for histones) or direct lysis in Laemmli buffer
Gel Electrophoresis Parameters:
Use 15% or gradient gels (4-20%) for optimal resolution of histone proteins
Load appropriate amount of protein (10-20μg of acid-extracted histones or 30-50μg of whole cell lysate)
Include molecular weight marker covering low range (10-50 kDa)
Note that observed molecular weight may be 32-33 kDa despite calculated MW of 21 kDa
Transfer and Detection Optimization:
Validation and Troubleshooting:
Include positive controls (cells treated with HDAC inhibitors)
Run acetylation-deficient mutants or deacetylated samples as negative controls
Test detection sensitivity with increasing protein loads
If detection is difficult, consider using enhanced chemiluminescence substrates or fluorescent secondary antibodies
For consistent results, maintain recommended protocol parameters and storage conditions (store antibody at -20°C in aliquots to avoid freeze-thaw cycles) . When analyzing results, confirm the molecular weight matches reported observations (32-33 kDa) and verify that signal strength correlates with expected acetylation levels across experimental conditions.
Achieving high-quality ChIP-seq data for HIST1H1C requires specialized considerations beyond standard ChIP protocols:
Optimized Crosslinking Strategy:
Implement dual crosslinking with DSG (disuccinimidyl glutarate) followed by formaldehyde
This captures both protein-protein and protein-DNA interactions, critical for linker histones
Optimize crosslinking time to balance efficient capture and DNA recovery
Chromatin Preparation Refinements:
Titrate nuclease digestion or sonication carefully to avoid over-fragmentation
Aim for slightly larger fragments (200-700bp) than in standard histone ChIP
Pre-clear chromatin thoroughly to reduce background
Immunoprecipitation Enhancements:
Use higher antibody amounts (3-5μg per reaction) than standard ChIP
Extend incubation time (overnight at 4°C with rotation)
Consider combining traditional ChIP with modern approaches like CUT&RUN for complementary data
Next-Generation Sequencing Considerations:
Increase sequencing depth (40-60 million reads) to capture broad distribution patterns
Implement paired-end sequencing for more precise mapping
Include spike-in controls for accurate normalization
Advanced Data Analysis Approaches:
Use specialized peak callers suitable for broad domains rather than sharp peaks
Compare HIST1H1C distribution with other histone variants and marks
Implement differential binding analysis to identify condition-specific patterns
Correlate binding with gene expression data to establish functional relationships
Research has demonstrated that while H1 variants occur across the genome, H1.2 (HIST1H1C) shows specific distribution patterns both at promoters and genome-wide . When analyzing ChIP-seq data, look specifically for these unique distribution patterns to distinguish H1.2 from other variants and understand its specific regulatory roles in your experimental system.
The HIST1H1C (Ab-62) Antibody offers powerful approaches for investigating the functional relationship between site-specific histone acetylation and transcriptional regulation:
Integrated Multi-Omics Approaches:
Combine ChIP-seq using HIST1H1C (Ab-62) Antibody with RNA-seq to correlate acetylation patterns with gene expression
Implement ATAC-seq in parallel to assess chromatin accessibility changes
Add CUT&RUN for other histone marks to build comprehensive epigenetic profiles
Analyze data using integrated computational frameworks to identify statistically significant associations
Perturbation Studies:
Apply HDAC inhibitors to increase global acetylation and assess HIST1H1C Lys62 acetylation dynamics
Use HAT inhibitors to determine which acetyltransferases target Lys62
Create lysine-to-arginine mutants (K62R) to prevent acetylation and assess functional consequences
Implement inducible expression systems to establish temporal relationships
Single-Cell Applications:
Develop single-cell ChIP protocols to assess cell-to-cell variability in acetylation patterns
Combine with single-cell RNA-seq to correlate at individual cell resolution
Implement imaging approaches like STORM with HIST1H1C (Ab-62) Antibody for spatial resolution
Functional Validation Experiments:
Use CRISPR/dCas9 systems with acetyltransferase domains to target Lys62 specifically
Implement reporter assays to measure transcriptional outcomes
Conduct nucleosome occupancy assays to determine effects on chromatin structure
This research direction builds on findings that H1.2 shows specific distribution patterns at promoters and genome-wide , suggesting it may play unique roles in transcriptional regulation through its acetylation state. Using the HIST1H1C (Ab-62) Antibody in these approaches can reveal how acetylation at Lys62 specifically contributes to these regulatory functions.
Several cutting-edge technologies can be integrated with HIST1H1C (Ab-62) Antibody to advance epigenetic research:
Advanced Genomic Mapping Technologies:
CUT&Tag: Offers improved signal-to-noise ratio over traditional ChIP by tethering the tagmentation enzyme directly to the antibody
CUT&RUN: Provides high resolution mapping with lower cell input requirements
HiChIP/PLAC-seq: Combines chromatin conformation capture with ChIP to map long-range interactions involving acetylated HIST1H1C
Spatial and Temporal Resolution Methods:
MERFISH/seqFISH: Allows visualization of chromatin states in intact cells with spatial context
Live-cell imaging with mintbodies: Modified antibody fragments for tracking acetylation dynamics in real-time
Optogenetic tools: Control acetylation/deacetylation with light-inducible enzymes
Single-Molecule and Biophysical Approaches:
Single-molecule tracking: Measure dynamics of acetylated vs. non-acetylated HIST1H1C
Optical tweezers combined with FRET: Assess how acetylation alters chromatin fiber mechanics
Liquid-liquid phase separation assays: Investigate how acetylation affects condensate formation, building on findings about charged disordered regions in histones
Multi-modal Single-Cell Technologies:
scCUT&Tag: Assess HIST1H1C acetylation patterns in individual cells
CITE-seq adaptations: Combine antibody detection with transcriptomics
Spatial transcriptomics with protein detection: Map acetylation patterns in tissue context
Computational Integration Frameworks:
Deep learning approaches for predicting acetylation patterns and functional outcomes
Multi-omics data integration methods to correlate acetylation with other epigenetic marks
Network analysis to identify regulatory hubs involving acetylated HIST1H1C
These technologies can help address key questions about how HIST1H1C acetylation contributes to chromatin structure regulation and gene expression control, potentially revealing mechanisms underlying H1.2's specific distribution patterns at promoters and across the genome .
The dynamic regulation of HIST1H1C acetylation during cellular transitions and pathological states represents an important research frontier:
Developmental Transitions:
Stem Cell Differentiation:
HIST1H1C acetylation patterns shift during lineage commitment
Changes correlate with alterations in chromatin accessibility at developmental genes
Time-course studies can reveal sequential epigenetic changes during differentiation
Tissue Development:
Tissue-specific patterns of HIST1H1C acetylation emerge during organogenesis
Cell-type specific acetylation signatures develop in mature tissues
Spatial variations in acetylation correlate with functional domains in tissues
Disease-Associated Alterations:
Cancer Progression:
Global changes in H1.2 acetylation occur during transformation
Aberrant patterns correlate with altered gene expression profiles
Specific tumor types show characteristic HIST1H1C modification signatures
Neurological Disorders:
Altered HIST1H1C acetylation in neurodegenerative conditions
Changes in acetylation precede clinical manifestations in some disorders
Potential for biomarker development based on modification patterns
Response to Environmental Signals:
Stress Responses:
Rapid changes in HIST1H1C acetylation following cellular stress
Different stressors induce distinct modification patterns
Acetylation changes may contribute to stress adaptation mechanisms
Metabolic Regulation:
Nutritional status influences HIST1H1C acetylation through metabolite availability
Circadian rhythms correlate with cyclic changes in modification patterns
Metabolic disorders show disrupted acetylation profiles
Therapeutic Implications:
HDAC Inhibitor Effects:
Different HDAC inhibitor classes have distinct effects on HIST1H1C Lys62 acetylation
Correlation between acetylation changes and therapeutic outcomes
Potential for monitoring treatment efficacy through modification levels
Targeted Interventions:
Specificity of different epigenetic modulators for HIST1H1C
Development of site-specific approaches to modify Lys62 acetylation
Combination strategies targeting multiple epigenetic marks
The HIST1H1C (Ab-62) Antibody provides a critical tool for investigating these dynamic changes, especially given that H1.2 shows specific distribution patterns at promoters and genome-wide that distinguish it from other H1 variants , suggesting specialized regulatory functions that may be particularly important during cellular transitions and disease states.