Acetyl-HIST1H1C (K84) Antibody is a polyclonal rabbit antibody specifically targeting the acetylated lysine 84 (K84) residue of histone H1.2 (HIST1H1C), a linker histone critical for chromatin structure and gene regulation . This antibody enables researchers to study post-translational modifications (PTMs) at this site, which are implicated in chromatin remodeling, transcriptional activation, and cellular processes such as apoptosis and immune response modulation .
Acetylation of histone H1 reduces its interaction with DNA, promoting chromatin decondensation and facilitating access to transcriptional machinery . At K84, acetylation may influence:
Chromatin compaction: Altered nucleosome spacing and DNA methylation patterns .
Gene expression: Regulation of genes involved in cell cycle control and differentiation .
Immune function: Modulation of dendritic cell (DC) maturation and T-cell activation, as seen in studies using histone H1 blockade .
Detection: Recognizes acetylated HIST1H1C in whole-cell lysates (e.g., HeLa, 293, A549 cells) .
Example: Sodium butyrate treatment induces acetylation, detected as a 22 kDa band .
Target regions: Enriched at regulatory regions (e.g., beta-globin promoter) .
Protocol: Cross-linked chromatin is immunoprecipitated with the antibody, followed by qPCR validation .
Localization: Stains nuclear regions with acetylated HIST1H1C, particularly in cells treated with HDAC inhibitors .
Mechanism: Histone H1 acetylation (including K84) is required for DC maturation via activation of p38 MAPK and IκBα pathways .
Impact: Blocking acetylation with anti-H1 antibodies suppresses CD80/CD86 expression and T-cell proliferation .
H1cTKO mice: Depletion of H1C/D/E variants reduces B/T cell proliferation and survival, linked to chromatin structure defects .
Acetylation dynamics: Acetyl-HIST1H1C may counteract H1-mediated repression of genes critical for cell fate decisions .
PTM interference: Multiple acetylation sites on H1 tails (e.g., K62, K74, K84) necessitate site-specific antibodies to avoid cross-reactivity .
Evolutionary variability: Low conservation of H1 sequences complicates antibody design .
Specificity concerns: MS-based approaches may be preferred over antibodies for complex PTM analysis .
Functional studies: Limited data on K84 acetylation’s role in cancer or metabolic disorders.
Therapeutic potential: Targeting H1 acetylation could modulate immune responses in transplantation or autoimmune diseases .
HIST1H1C, also known as Histone H1.2, is a member of the linker histone H1 family that plays crucial roles in chromatin organization and gene expression regulation. Unlike core histones (H2A, H2B, H3, and H4) that form the nucleosome, H1 histones bind to the linker DNA between nucleosomes, facilitating higher-order chromatin structure formation . The protein has a calculated molecular weight of 21 kDa but is often observed at 32-33 kDa in experimental conditions . HIST1H1C contains multiple lysine residues that can undergo post-translational modifications, including acetylation at K84, which can alter its binding properties and functional activities in chromatin regulation . H1 histones, including HIST1H1C, are essential not only for maintaining higher-order chromatin structure but also for regulating gene expression through controlling chromatin accessibility to transcription factors and other regulatory proteins .
Acetyl-HIST1H1C (K84) antibodies can be employed in multiple research applications depending on the experimental question. Based on data from similar HIST1H1C antibodies, recommended applications include:
Application | Technique | Recommended Dilution | Sample Types |
---|---|---|---|
Protein Detection | Western Blot (WB) | 1:500-1:3000 | Cell lysates, tissue extracts |
Cellular Localization | Immunofluorescence (IF) | 1:50-1:500 | Fixed cells, tissue sections |
Tissue Studies | Immunohistochemistry (IHC) | 1:100-1:600 | FFPE or frozen tissue sections |
Protein Interactions | Immunoprecipitation (IP) | 0.5-4.0 μg for 1-3 mg protein | Cell/tissue lysates |
Chromatin Analysis | Chromatin Immunoprecipitation (ChIP) | Application-specific | Cross-linked chromatin |
It is crucial to validate the specific Acetyl-HIST1H1C (K84) antibody for each application as modification-specific antibodies may have different optimal conditions compared to pan-HIST1H1C antibodies . For IHC applications, antigen retrieval with TE buffer (pH 9.0) or citrate buffer (pH 6.0) is recommended to maximize epitope accessibility .
Proper storage is critical for maintaining antibody functionality. For Acetyl-HIST1H1C (K84) antibodies, the following storage guidelines should be followed:
Store at -20°C for long-term preservation. Most HIST1H1C antibodies are stable for at least one year when properly stored .
Use appropriate storage buffer - typically PBS with 0.02% sodium azide and 50% glycerol at pH 7.3 provides optimal stability .
Avoid repeated freeze-thaw cycles that can degrade antibody quality. For antibodies in glycerol formulations (≥50%), aliquoting is often unnecessary for -20°C storage .
For working solutions, store at 4°C for up to one month. Beyond this timeframe, degradation may affect antibody performance.
Check the manufacturer's specific recommendations, as formulations may vary among suppliers .
Proper storage conditions are essential for preserving specificity and reactivity, particularly for modification-specific antibodies that must distinguish subtle epitope differences.
Validating antibody specificity is crucial for accurate interpretation of experimental results, particularly for post-translational modification-specific antibodies. For Acetyl-HIST1H1C (K84) antibodies, a multi-step validation approach is recommended:
Peptide Competition Assays: Pre-incubate the antibody with acetylated and non-acetylated peptides containing the K84 position. Only the acetylated peptide should block antibody binding in subsequent applications.
Testing on Recombinant Proteins: Evaluate reactivity against recombinant HIST1H1C proteins with defined modifications:
Wild-type HIST1H1C
HIST1H1C with K84 acetylation
HIST1H1C with acetylation at other lysine residues
HIST1H1C with K84R mutation (mimicking non-acetylated state)
ELISA-Based Validation: Similar to approaches used for other histone modifications, perform ELISA using synthetic peptides with specific modifications to assess cross-reactivity with neighboring modifications .
Lysine Deacetylase (KDAC) Treatment: Treat samples with KDACs and confirm reduced antibody reactivity as acetyl groups are removed.
Knockout/Knockdown Controls: Use HIST1H1C knockout cells or cells treated with CRISPR-Cas9 to create K84R mutations as negative controls.
Mass Spectrometry Correlation: Validate antibody-based findings using mass spectrometry to confirm the presence of acetylation at K84 in immunoprecipitated samples.
The comprehensive validation method established for histone H4 modification antibodies described in the literature provides an excellent framework that can be adapted for Acetyl-HIST1H1C (K84) antibodies .
Chromatin immunoprecipitation followed by sequencing (ChIP-seq) with Acetyl-HIST1H1C (K84) antibodies requires careful optimization:
Chromatin Preparation:
Optimize fixation time (typically 5-15 minutes with 1% formaldehyde)
Use appropriate sonication conditions to achieve fragments of 200-500 bp
Verify fragmentation efficiency by gel electrophoresis
Antibody Validation for ChIP:
Perform preliminary ChIP-qPCR at known target loci
Include appropriate controls (IgG, input chromatin)
Optimize antibody concentration (typically 2-5 μg per ChIP reaction)
Sequential ChIP Considerations:
To study co-occupancy of acetylated HIST1H1C with other modifications, sequential ChIP can be performed
Ensure elution conditions from first IP do not denature the epitope for the second IP
Data Analysis Parameters:
Use appropriate peak calling algorithms (MACS2 is commonly used)
Consider using a spike-in normalization approach for comparative analyses
Validate findings at selected loci using ChIP-qPCR
Biological Interpretation:
Compare acetylated HIST1H1C K84 profiles with other histone modifications (e.g., H3K27ac for active enhancers)
Correlate with transcriptome data to establish functional relationships
Consider cell-type specific patterns of HIST1H1C acetylation
Drawing from experiences with other histone modification ChIP-seq studies, successful experiments typically require 10^6-10^7 cells per immunoprecipitation and antibodies with high specificity for the target modification .
Post-translational modifications (PTMs) on histone proteins can significantly impact antibody recognition through several mechanisms:
To address these challenges, researchers should:
Test antibody reactivity against peptide arrays containing combinations of modifications
Use complementary techniques (mass spectrometry) to verify modification patterns
Consider developing antibodies that recognize specific combinatorial PTM patterns
Validate findings using recombinant HIST1H1C proteins with defined modification states
These considerations highlight the importance of comprehensive antibody characterization before application in complex experimental systems .
Developing highly specific antibodies against acetylated HIST1H1C presents several challenges that researchers should be aware of:
Limited Immunogenicity: Modified histone peptides may have limited immunogenicity, making it difficult to generate robust immune responses in host animals.
Sequence Conservation: High sequence conservation of histones across species can limit the diversity of immune responses and potentially lead to cross-reactivity with other H1 variants.
Epitope Design Complexities: The design of immunogens must carefully consider:
Peptide length (typically 10-15 amino acids surrounding K84)
Carrier protein selection for immunization
Position of acetylated lysine within the immunogen (central positioning generally yields better specificity)
Validation Challenges: As noted in the literature, the availability of specific immunological reagents for histone H1 is "drastically lacking," creating a significant obstacle for research progress .
Cross-Reactivity Issues: The acetylated lysine epitope may be recognized in multiple contexts, requiring extensive negative selection to ensure specificity.
Batch-to-Batch Variability: Polyclonal antibodies against PTMs often show significant batch-to-batch variation, necessitating extensive validation for each lot.
To address these challenges, advanced antibody development approaches include:
Using multiple host species to generate diverse antibody repertoires
Implementing negative selection strategies during screening
Employing recombinant antibody technology to ensure consistency
Developing monoclonal antibodies using modified hybridoma techniques
These strategies have been successfully employed for other histone modifications and could be adapted for Acetyl-HIST1H1C (K84) antibody development .
Proper experimental controls are critical for accurate interpretation of results with Acetyl-HIST1H1C (K84) antibodies:
Positive Controls:
Cell lines or tissues with documented high levels of K84 acetylation
Recombinant HIST1H1C protein acetylated at K84
Samples treated with histone deacetylase inhibitors (HDACi) like trichostatin A (TSA) or sodium butyrate, which increase global histone acetylation
Negative Controls:
Samples treated with lysine acetyltransferase inhibitors
CRISPR-engineered cell lines with K84R mutation (preventing acetylation)
Peptide competition controls where antibody is pre-incubated with acetylated peptide
Technical Controls:
For Western blotting: Loading controls (total HIST1H1C or other histones)
For IF/IHC: Secondary antibody-only controls
For ChIP: IgG controls from the same species as the primary antibody
Input samples (representing starting material before immunoprecipitation)
Validation Controls:
Analysis with a second antibody against the same modification
Correlation with enzymatic activity assays for writers and erasers of K84 acetylation
Mass spectrometry validation of acetylation status
Including these comprehensive controls will significantly enhance data reliability and facilitate proper interpretation of experimental outcomes .
Optimizing immunohistochemistry (IHC) protocols for detecting Acetyl-HIST1H1C (K84) requires attention to several critical parameters:
Tissue Preparation and Fixation:
Fixation time affects epitope accessibility; over-fixation can mask epitopes
For formalin-fixed paraffin-embedded (FFPE) tissues, 12-24 hours fixation in 10% neutral buffered formalin is typically optimal
Fresh frozen tissues may provide better epitope preservation for some applications
Antigen Retrieval Methods:
Blocking and Antibody Incubation:
Detection Systems:
For chromogenic detection, HRP-based systems work well with DAB substrate
For fluorescent detection, select fluorophores that minimize tissue autofluorescence
Consider amplification systems (tyramide signal amplification) for low-abundance targets
Counterstaining and Mounting:
For nuclear targets like HIST1H1C, hematoxylin provides good nuclear contrast
Use mounting media with DAPI for fluorescent applications
Consider antifade reagents to preserve signal in fluorescent applications
The validation data available for HIST1H1C antibodies demonstrates successful IHC staining of human thyroid cancer tissue at 1:70 dilution , which can serve as a starting point for optimization with acetylation-specific antibodies.
Achieving reproducible Western blot results with Acetyl-HIST1H1C (K84) antibodies requires careful consideration of several factors:
Sample Preparation:
Include histone deacetylase inhibitors (e.g., sodium butyrate, TSA) in lysis buffers
Add protease inhibitors to prevent degradation
Use acidic extraction methods optimized for histone proteins
Ensure consistent protein quantification between experiments
Gel Electrophoresis Parameters:
Select appropriate gel percentage (12-15% for histones)
Load equal amounts of protein (typically 10-30 μg total protein)
Include molecular weight markers appropriate for low molecular weight proteins
Consider using specialized gel systems optimized for histone separation
Transfer Conditions:
Use PVDF membranes (0.2 μm pore size) for optimal binding of small proteins
Optimize transfer conditions (voltage/time) for small proteins
Consider using transfer buffers with lower methanol content for histones
Antibody Incubation:
Detection and Analysis:
Select appropriate exposure times to avoid signal saturation
Use quantification standards for comparing acetylation levels
Normalize to total HIST1H1C levels when comparing acetylation between samples
Consider using fluorescent secondary antibodies for more quantitative analysis
Despite the calculated molecular weight of 21 kDa, HIST1H1C is typically observed at 32-33 kDa in SDS-PAGE , likely due to the charged nature of histones affecting their migration. This should be considered when interpreting Western blot results.
Investigating chromatin dynamics during cell differentiation using Acetyl-HIST1H1C (K84) antibodies can provide valuable insights into epigenetic regulation mechanisms:
Time-Course Experiments:
Design experiments to capture acetylation changes at key differentiation timepoints
Correlate acetylation patterns with expression of lineage-specific genes
Compare with other histone modifications to establish temporal relationships
ChIP-seq Integration Approaches:
Perform ChIP-seq with Acetyl-HIST1H1C (K84) antibodies at multiple differentiation stages
Integrate data with transcriptome profiling (RNA-seq)
Analyze genomic distribution patterns (promoters, enhancers, gene bodies)
Compare with maps of other histone modifications and chromatin accessibility data
Locus-Specific Analysis:
Use ChIP-qPCR to examine acetylation changes at specific regulatory elements
Implement genome editing to mutate K84 and assess functional consequences
Perform reporter assays to determine the impact of K84 acetylation on gene expression
Single-Cell Approaches:
Apply CUT&Tag or other single-cell compatible techniques
Correlate HIST1H1C acetylation with cell fate decisions
Identify cell subpopulations with distinct epigenetic signatures
Manipulation of K84 Acetylation:
Identify and modulate the activity of enzymes responsible for K84 acetylation/deacetylation
Observe consequences on differentiation efficiency and lineage specification
Investigate mechanisms linking HIST1H1C acetylation to transcriptional regulation
Similar approaches have been successfully employed to study histone H4 modifications , and could be adapted for HIST1H1C acetylation studies in developmental contexts.
Identifying proteins that specifically interact with acetylated HIST1H1C at K84 requires specialized proteomic approaches:
Acetylation-Specific Protein Pulldown:
Synthesize biotinylated peptides containing acetylated or unacetylated K84
Immobilize peptides on streptavidin beads
Incubate with nuclear extracts under physiological conditions
Identify bound proteins by mass spectrometry
Compare binding profiles between acetylated and unacetylated peptides
SILAC-Based Quantitative Proteomics:
Culture cells in media containing light or heavy isotope-labeled amino acids
Perform pulldowns with acetylated and unacetylated baits using differently labeled extracts
Combine samples and analyze by mass spectrometry
Calculate heavy/light ratios to identify preferential binders
Proximity Labeling Approaches:
Generate fusion proteins of HIST1H1C with promiscuous biotin ligases (BioID or TurboID)
Create K84Q (acetylation mimetic) and K84R (non-acetylatable) mutants
Express in cells and activate biotin labeling
Purify biotinylated proteins and identify by mass spectrometry
Compare interactomes between wildtype and mutant proteins
Cross-Linking Mass Spectrometry:
Perform in vivo chemical cross-linking
Immunoprecipitate with Acetyl-HIST1H1C (K84) antibodies
Analyze cross-linked peptides by specialized mass spectrometry
Identify direct protein-protein interactions involving acetylated K84
Validation Methods:
Confirm interactions by co-immunoprecipitation with Acetyl-HIST1H1C (K84) antibodies
Test direct binding using recombinant proteins or peptides
Assess functional relevance through depletion or overexpression studies
These approaches can reveal "readers" of HIST1H1C acetylation, providing insights into downstream functional consequences of this modification.
Quantitative analysis of HIST1H1C K84 acetylation requires rigorous methodological approaches:
Mass Spectrometry-Based Quantification:
Extract histones using specialized protocols (acid extraction)
Perform propionylation of unmodified lysines
Digest with trypsin to generate peptides containing K84
Use multiple reaction monitoring (MRM) or parallel reaction monitoring (PRM) for targeted quantification
Calculate stoichiometry of K84 acetylation relative to unmodified peptide
Western Blot Quantification:
Use dual detection systems (fluorescent secondary antibodies)
Probe simultaneously for Acetyl-HIST1H1C (K84) and total HIST1H1C
Calculate the ratio of acetylated to total protein
Include standard curves using recombinant proteins or synthetic peptides
Employ image analysis software for densitometry with background correction
ELISA-Based Approaches:
Develop sandwich ELISA using capture antibodies against HIST1H1C
Detect with Acetyl-HIST1H1C (K84) antibodies
Compare to standard curves generated with synthetic peptides
Normalize to total HIST1H1C levels measured in parallel assays
ChIP-seq Quantification:
Include spike-in controls (e.g., Drosophila chromatin) for normalization
Calculate normalized read density at regions of interest
Compare acetylation levels between conditions using appropriate statistical methods
Validate changes at selected loci by ChIP-qPCR
Single-Cell Analysis:
Apply immunofluorescence with Acetyl-HIST1H1C (K84) antibodies
Perform quantitative image analysis to measure nuclear signal intensity
Normalize to total HIST1H1C or DNA content
Analyze cell-to-cell variability and population distributions
These quantitative approaches enable precise measurement of changes in HIST1H1C K84 acetylation levels in response to experimental manipulations, drug treatments, or during biological processes.
Understanding potential artifacts is crucial for accurate data interpretation:
Sources of False Positives:
Cross-reactivity with acetylation at similar motifs in other H1 variants
Non-specific binding to highly abundant proteins
Recognition of acetylated lysines in non-histone proteins with similar surrounding sequences
Insufficient blocking leading to background signal
Secondary antibody cross-reactivity with endogenous immunoglobulins
Sources of False Negatives:
Epitope masking by adjacent modifications or protein-protein interactions
Over-fixation in IHC/IF applications obscuring the epitope
Inefficient extraction of chromatin-bound histones
Deacetylation during sample preparation (absence of HDAC inhibitors)
Antibody lot variability affecting recognition efficiency
Technical Considerations:
Validation Approaches:
Include known positive and negative controls in each experiment
Verify key findings with orthogonal techniques (e.g., mass spectrometry)
Test antibody specificity using peptide competition assays
Compare results from multiple antibodies targeting the same modification
The limited availability and specificity of histone H1 antibodies highlighted in the literature emphasizes the importance of thorough validation to avoid misinterpretation of experimental results.
When faced with discrepancies between different detection methods, systematic troubleshooting and interpretation are required:
Method-Specific Considerations:
Western Blot vs. Immunofluorescence: Different sample preparation methods may affect epitope accessibility
ChIP-seq vs. Mass Spectrometry: ChIP measures genomic distribution while MS quantifies global levels
Cell vs. Tissue Results: Cellular heterogeneity in tissues may mask cell-type specific patterns
Resolution Strategies:
Perform side-by-side comparisons using standardized samples
Validate antibodies in the context of each specific application
Consider the sensitivity limitations of each technique
Evaluate whether discrepancies reflect biological variance or technical artifacts
Integration Framework:
Create a matrix comparing results across techniques and conditions
Identify consistent patterns despite methodological differences
Weight evidence based on technical robustness of each approach
Formulate hypotheses that reconcile apparent contradictions
Case Study Approach:
If Western blot shows increased K84 acetylation but ChIP-seq shows decreased genomic binding:
Consider that acetylation might reduce chromatin association
Examine whether acetylation alters HIST1H1C stability or nuclear localization
Investigate potential redistribution rather than absolute change in modification levels
External Validation:
Compare with published literature on similar histone modifications
Consult with specialists in each methodology
Consider biological context and known regulatory mechanisms
This structured approach to resolving conflicting data can transform discrepancies from obstacles into opportunities for deeper mechanistic insights.
Proper statistical analysis is essential for extracting meaningful biological insights from ChIP-seq data:
Quality Control Metrics:
Fragment size distribution (optimal range: 200-500 bp)
Library complexity (PCR duplicate rate <20%)
Mapping quality (>80% uniquely mapped reads)
Signal-to-noise ratio (enrichment over background)
Peak number consistency between replicates
Peak Calling Approaches:
For sharp peaks: MACS2 with appropriate p-value threshold (typically 1e-5)
For broad domains: SICER or RSEG algorithms
Include input controls or IgG controls for background correction
Consider biological replicates during peak calling (IDR method)
Differential Binding Analysis:
Use specialized tools: DiffBind or MAnorm
Apply appropriate normalization methods:
Sequencing depth normalization
Spike-in normalization for global changes
Quantile normalization for technical variation
Control false discovery rate using Benjamini-Hochberg method
Integration with Other Data Types:
Correlation with gene expression (RNA-seq)
Overlap with other histone modifications
Association with chromatin accessibility (ATAC-seq/DNase-seq)
Motif enrichment analysis for transcription factor binding sites
Visualization and Reporting:
Generate average profile plots and heatmaps around features of interest
Use genome browsers for locus-specific visualization
Report effect sizes alongside p-values
Include biological replicates in visualizations
Successful ChIP-seq analysis with histone modification antibodies has been demonstrated in previous studies , and these approaches can be adapted for Acetyl-HIST1H1C (K84) ChIP-seq data analysis.
Several cutting-edge technologies show promise for advancing research on HIST1H1C K84 acetylation:
Single-Cell Epigenomic Approaches:
CUT&Tag and CUT&RUN methods adapted for single-cell analysis
Single-cell mass cytometry (CyTOF) with Acetyl-HIST1H1C (K84) antibodies
Spatial epigenomics to map acetylation patterns in tissue contexts
These approaches will reveal cell-to-cell variability and rare subpopulations
Targeted Manipulation of K84 Acetylation:
CRISPR-based epigenome editing (dCas9 fused to histone acetyltransferases)
Optogenetic control of acetylation/deacetylation enzymes
Chemical biology approaches with targeted degraders or activators
These tools enable causal studies of K84 acetylation function
Real-Time Acetylation Monitoring:
Development of genetically encoded biosensors for K84 acetylation
Live-cell imaging of acetylation dynamics during cellular processes
FRET-based approaches to study protein interactions dependent on K84 acetylation
These methods will reveal temporal dynamics previously inaccessible
Structural Biology Applications:
Cryo-EM studies of chromatin containing acetylated HIST1H1C
Hydrogen-deuterium exchange mass spectrometry to probe structural changes
NMR studies of acetylation-dependent interactions
These approaches will reveal mechanistic details at atomic resolution
Multi-Modal Omics Integration:
Simultaneous profiling of multiple histone marks, transcription, and chromatin accessibility
Machine learning approaches to identify acetylation-dependent regulatory networks
Systems biology modeling of acetylation/deacetylation dynamics
These integrative approaches will place K84 acetylation in broader biological contexts
These emerging technologies will address current limitations in studying histone H1 modifications noted in the literature and accelerate discovery in this important but challenging field.
Several promising research directions could significantly advance our understanding of HIST1H1C K84 acetylation:
Cell Fate Transitions and Development:
Investigation of K84 acetylation dynamics during embryonic development
Role in cellular reprogramming and induced pluripotency
Contribution to lineage specification and terminal differentiation
Potential as a biomarker for developmental stages or cell identity
Disease Relevance and Therapeutic Targeting:
Alterations in cancer and potential diagnostic applications
Role in neurodegenerative disorders and chromatin dysregulation
Involvement in inflammatory responses and immune cell function
Development of small molecules targeting enzymes that regulate K84 acetylation
Mechanistic Understanding of Chromatin Regulation:
Effect on HIST1H1C binding dynamics and residence time on chromatin
Impact on higher-order chromatin structure and phase separation
Interplay with core histone modifications and chromatin remodeling
Relationship to 3D genome organization and topologically associating domains
Evolutionary Conservation and Divergence:
Comparative studies across species to identify conserved functions
Analysis of paralogs and tissue-specific H1 variants
Reconstruction of evolutionary history of K84 acetylation regulation
Identification of conserved reader proteins and regulatory pathways
Environmental and Metabolic Regulation:
Response to environmental stressors and cellular metabolism
Connection to acyl-CoA levels and metabolic state
Role in aging and cellular senescence
Transgenerational inheritance and epigenetic memory
These research directions would address the current knowledge gaps in histone H1 biology highlighted in the literature and potentially reveal novel functions and regulatory mechanisms of HIST1H1C K84 acetylation.