The Mono-methyl-HIST1H1C (K96) Antibody is a polyclonal rabbit-derived antibody specifically designed to detect mono-methylation at lysine 96 (K96) of the histone H1.1C protein, also known as Histone H1.2 (gene symbol: HIST1H1C). Histone H1 is a linker histone critical for chromatin structure and gene regulation, with its post-translational modifications influencing chromatin condensation, transcription, and cellular processes like autophagy .
The antibody targets a peptide sequence encompassing the methylated lysine residue at position 96 of Histone H1.2. Its immunogen is derived from human Histone H1.2, ensuring specificity for the mono-methylated state of K96. The antibody is affinity-purified and validated for use in:
ELISA (enzyme-linked immunosorbent assay)
WB (western blotting)
IF (immunofluorescence)
The antibody is primarily used in epigenetic and nuclear signaling research. Key applications include:
Histone H1.2 (HIST1H1C) has been implicated in autophagy regulation. Overexpression of HIST1H1C promotes autophagy flux by upregulating ATG proteins and deacetylating histone H4K16 (H4K16Ac), a marker linked to autophagy activation . The Mono-methyl-HIST1H1C (K96) Antibody could be instrumental in studying this mechanism, as K96 methylation may modulate chromatin-interacting proteins or transcriptional regulators .
Histone methylation, including K96 mono-methylation, recruits chromatin remodelers and transcription factors to regulate gene expression. This antibody enables precise tracking of these epigenetic modifications, which are critical in development, cancer, and metabolic disorders .
HIST1H1C (also known as H1.2) is one of the somatic linker histone H1 variants that associates with linker DNA between nucleosomes. As part of the histone H1 family, HIST1H1C belongs to the five main somatic subtypes (H1.1-H1.5) whose genes reside within the histone gene cluster 1 on chromosome 6p region . Functionally, HIST1H1C binding to linker DNA leads to more compacted chromatin, thereby decreasing accessibility of regulatory proteins and chromatin modifiers to DNA . Like other H1 variants, HIST1H1C plays important roles in nucleosome positioning, chromatin compaction, and higher-order chromatin structure maintenance, which collectively influence gene expression patterns . Despite having over 60% sequence identity with other H1 subtypes, HIST1H1C possesses variant-specific functions, as evidenced by the different phenotypes observed in H1-variant knockdown models .
Developing antibodies against specific histone H1 methylation sites presents several technical challenges. First, the high sequence similarity (>60%) among mammalian H1 subtypes (H1.1-H1.5) makes it difficult to generate antibodies that can distinguish between similar epitopes across variants . Second, histone H1 has a high lysine content, which creates challenges for both antibody development and subsequent proteomics validation . Specifically for bottom-up proteomics:
Trypsin digestion yields small, hydrophilic peptides that are difficult to detect by mass spectrometry due to poor retention in C18 RP-HPLC columns .
Regions with the highest density of lysine residues (such as the C-terminal domain) tend to have low coverage in proteomics analyses .
Many promiscuous peptides matching the sequence of several subtypes are often found in bottom-up studies, complicating variant-specific assignment .
These challenges have been partially addressed through techniques like propionylation of amine groups before or after tryptic digestion, significantly improving the MS coverage of H1 .
To distinguish mono-methylation at K96 of HIST1H1C from similar modifications on other H1 variants, researchers should employ multiple complementary approaches:
Researchers should validate antibody specificity using recombinant HIST1H1C proteins with and without the K96 methylation, alongside other H1 variants with similar modifications .
For optimal Western blotting results with Mono-methyl-HIST1H1C (K96) Antibody, researchers should follow these evidence-based protocols:
Sample preparation: Use nuclear extracts rather than whole cell lysates for enrichment of histone proteins . The Nuclear Extraction kit (Active Motif) has been successfully used in histone H1 studies .
Protein extraction: For histones, use CelLytic M cell lysis reagent containing complete protease inhibitor cocktail to prevent degradation and preserve post-translational modifications .
Gel electrophoresis considerations:
Transfer and detection:
Controls to include:
Optimizing ChIP protocols for Mono-methyl-HIST1H1C (K96) Antibody requires attention to several key parameters:
Crosslinking optimization: Unlike core histones, H1 has more dynamic binding to chromatin. Optimizing formaldehyde crosslinking time (typically 10-15 minutes) is crucial for capturing the true genomic distribution of HIST1H1C .
Sonication parameters: Since H1 binds to linker DNA, adjust sonication conditions to generate fragments of appropriate size (typically 200-500 bp) without disrupting specific histone-DNA interactions .
Antibody concentration and incubation: Based on studies with other histone H1 antibodies, optimal results are often achieved with longer incubation times (overnight at 4°C) and higher antibody concentrations than those used for core histone modifications .
Washing stringency: Balance between removing non-specific interactions while preserving specific antibody-antigen binding. This may require empirical optimization of salt concentrations in wash buffers .
Elution conditions: Standard elution buffers used for other histone modifications are generally suitable, but may need optimization for specific experimental goals.
Data analysis considerations: Given that H1 shows enhanced enrichment at nucleosome-free regions and depletion from coding sequences of expressed genes, analysis algorithms should be adapted accordingly .
When using antibodies for in vivo experiments, several pharmacokinetic factors must be carefully considered:
Antibody clearance: Research shows that antibody concentrations can drop dramatically after initial administration, potentially becoming undetectable after approximately 10 days due to clearance mechanisms .
Anti-drug antibody (ADA) formation: The immune system may produce ADAs against the therapeutic antibody, significantly increasing clearance rates and reducing efficacy. Studies have shown that ADA formation can render plasma concentrations of the therapeutic antibody undetectable in most animals .
Correlation between plasma concentration and efficacy: Higher antibody concentrations in plasma generally correlate with better therapeutic efficacy, highlighting the importance of maintaining sufficient antibody levels throughout the experiment .
Combination treatments: When antibodies are used in combination with other therapeutic agents, unexpected pharmacokinetic interactions may occur. For instance, anti-PD-1 antibody concentrations were lower at later time points in animals receiving combination treatment compared to those receiving anti-PD-1 alone .
Measurement protocols: It is strongly recommended to measure both plasma concentration and anti-drug antibody formation throughout in vivo studies to correctly interpret pharmacodynamic data .
Histone H1 variants exhibit diverse post-translational modifications that differentially impact gene expression. The pattern and distribution of these modifications vary between variants and influence their regulatory functions:
Research shows that different methylation sites on H1 variants can have distinct functions. For example, WHSC1-mediated mono-methylation of H1.4 at K85 enhances stemness features in squamous cell carcinoma of head and neck (SCCHN) cells, inducing transcriptional activation of OCT4 . By analogy, mono-methylation of HIST1H1C at K96 likely has specific regulatory functions potentially distinct from those of other H1 modifications.
Research on histone H1 and DNA methylation reveals complex interplay between these epigenetic mechanisms:
H1 depletion affects DNA methylation patterns: Studies in Neurospora crassa show that H1 limits DNA methylation, with loss of H1 causing a global increase in DNA methylation . This suggests H1 acts as a nonspecific chromatin binding protein that can limit accessibility of the DNA methylation machinery .
Separation of methylation pathways: H1 enforces the separation of euchromatic and heterochromatic DNA methylation pathways by excluding the small RNA-generating branch of non-CG methylation from heterochromatin . This prevents triggering RNA-directed DNA methylation at heterochromatic transposable elements .
Impact of methylated H1 on DNA methyltransferase recruitment: While not directly addressed in the search results, by analogy with other histone modifications, mono-methylation of HIST1H1C at K96 may create or disrupt binding sites for readers that interact with DNA methylation machinery.
Context-dependent effects: The relationship between H1 and DNA methylation appears complex and context-dependent. For instance, in h1met1 mutants (lacking both H1 and the CG methyltransferase MET1), perturbations in non-CG methylation and H3K9me2 are observed at certain transposable elements .
The application of Mono-methyl-HIST1H1C (K96) Antibody in cancer research builds on emerging evidence linking histone H1 modifications to oncogenesis:
Diagnostic and prognostic potential: Histone H1 variants show altered expression and modification patterns in cancer, with the TCGA revealing genomic aberrations in linker histones in 6-12% of pancreatic, cervical, head and neck, and colorectal cancers . Mono-methyl-HIST1H1C (K96) Antibody could help establish whether this specific modification correlates with particular cancer subtypes or outcomes.
Investigation of cancer-specific histone methyltransferases: Research has identified WHSC1 as a methyltransferase that mono-methylates H1.4 at K85 in SCCHN cells . Similar studies using Mono-methyl-HIST1H1C (K96) Antibody could identify enzymes responsible for this specific modification and evaluate their potential as therapeutic targets.
Stemness and differentiation studies: H1.4 K85 mono-methylation induces transcriptional activation of OCT4 and stemness features in cancer cells . Researchers could investigate whether HIST1H1C K96 mono-methylation plays similar roles in cancer stem cell properties.
Therapeutic development: Targeting histone modifiers in SCCHN has therapeutic potential, with ongoing efforts to develop WHSC1 inhibitors . Understanding the role of HIST1H1C K96 mono-methylation could similarly inform development of targeted therapies.
Experimental design considerations:
Include appropriate controls when evaluating antibody specificity in diverse cancer cell lines
Combine chromatin immunoprecipitation with DNA/RNA sequencing to identify genes regulated by this modification
Consider the impact of tumor heterogeneity on detection of histone modifications
Integrate findings with other epigenetic marks to understand the broader regulatory context
Researchers should be aware of several potential pitfalls when using histone modification-specific antibodies:
Cross-reactivity issues:
High sequence similarity between H1 variants (>60% identity) increases the risk of cross-reactivity
Similar modification sites across different H1 variants may not be distinguished by the antibody
Other lysine modifications (acetylation, di/tri-methylation) at the same position might interfere with antibody recognition
Technical factors affecting detection:
Insufficient protein extraction from nuclear fractions
Degradation of modified histones during sample preparation
Inadequate blocking leading to non-specific binding
Batch-to-batch variability in antibody production
Biological confounders:
Variations in HIST1H1C expression levels across cell types
Dynamic nature of histone modifications during cell cycle progression
Heterogeneity in modification patterns within cell populations
Potential masking of the epitope in certain chromatin contexts
Validation approaches to minimize false results:
Use both positive and negative controls in each experiment
Employ peptide competition assays to confirm specificity
Validate results with orthogonal methods such as mass spectrometry
Consider using genetic models with mutations at the K96 position as definitive controls
When faced with discrepancies between antibody-based and mass spectrometry (MS) results for HIST1H1C modifications, consider these interpretative approaches:
Technical limitations of MS for histone H1 analysis:
Antibody-specific considerations:
Potential cross-reactivity with similar epitopes
Sensitivity differences compared to MS
Batch-to-batch variability
Reconciliation strategies:
Propionylation of amine groups before or after tryptic digestion to improve MS coverage
Separation of individual subtypes before proteolytic digestion using capillary electrophoresis or 2D-electrophoresis
Combining bottom-up and top-down proteomics approaches for more comprehensive analysis
Considering the relative quantitative strengths of each method (MS for abundance, antibodies for spatial distribution)
Biological explanations for discrepancies:
Cell type-specific or context-dependent modifications
Dynamic modifications that may be captured differently by each method
Epitope masking in certain chromatin contexts detected by one method but not the other
While the search results don't specifically address single-cell applications for studying HIST1H1C modifications, we can extrapolate from current methodological advances in histone biology:
Single-cell adaptations of conventional techniques:
Single-cell ChIP-seq protocols can be adapted for Mono-methyl-HIST1H1C (K96) Antibody
CUT&Tag or CUT&RUN methods offer improved sensitivity for limited cell numbers
Single-cell ATAC-seq can be used to correlate chromatin accessibility with H1 modification status
Imaging-based approaches:
Super-resolution microscopy combined with specific antibodies allows visualization of modification distribution in single cells
Mass cytometry (CyTOF) with metal-conjugated antibodies enables multiplexed detection of multiple histone modifications simultaneously
Computational considerations:
Single-cell data requires specialized computational pipelines to address sparsity and technical noise
Integration with other single-cell omics data (RNA-seq, DNA methylation) provides comprehensive epigenetic landscapes
Trajectory analysis methods can reveal dynamics of H1 modifications during cellular processes
Validation strategies:
Correlation with bulk methods in the same cell populations
Use of genetically encoded reporters for live-cell tracking of modification status
Cross-validation with orthogonal single-cell techniques
These advanced methodologies promise to reveal cell-to-cell heterogeneity in HIST1H1C modifications and their functional consequences in complex biological systems.
Several emerging technologies hold promise for improving antibody-based detection of histone H1 modifications:
Recombinant antibody engineering:
Phage display selection against specific modified peptides to generate highly specific antibodies
Nanobodies (single-domain antibodies) that may access epitopes with less steric hindrance in compact chromatin
Bispecific antibodies that recognize both the modification and unique sequence features of HIST1H1C
Proximity-based detection methods:
Proximity ligation assays to detect co-occurrence of multiple modifications or protein interactions
APEX2-based proximity labeling to map the protein environment of modified HIST1H1C
Split-protein complementation systems to visualize modification-dependent interactions
Advanced mass spectrometry integration:
CRISPR-guided proteomics to focus MS analysis on specific genomic regions
Antibody-coupled mass spectrometry for enrichment of low-abundance modifications
Targeted quantitative proteomics with heavy-labeled peptide standards for absolute quantification
Spatial omics approaches:
In situ sequencing combined with antibody detection to map genomic locations of modifications
Spatial proteomics to understand nuclear distribution of modified histones
Correlative light-electron microscopy to link modification status with ultrastructural features
Understanding HIST1H1C mono-methylation has several potential implications for epigenetic therapy development:
Targeting specific methyltransferases:
Exploiting connections to DNA methylation:
Combinatorial epigenetic therapies:
Understanding the interplay between HIST1H1C modifications and other epigenetic marks could inform rational combinations of epigenetic drugs
For example, combining inhibitors of HIST1H1C methylation with those targeting DNA methylation or histone deacetylation might yield synergistic effects
Biomarker development:
Delivery challenges and solutions:
Development of targeted delivery systems for epigenetic drugs to specific cell types or nuclear compartments
Design of engineered proteins or peptides that can recognize and mask specific histone modifications
Several fundamental questions remain to be addressed regarding HIST1H1C K96 mono-methylation:
Enzymatic regulation:
Which methyltransferases catalyze HIST1H1C K96 mono-methylation?
Which demethylases remove this modification?
How is the activity of these enzymes regulated in different cellular contexts?
Reader proteins and signaling:
What proteins specifically recognize mono-methylated K96 of HIST1H1C?
How does this recognition translate into downstream effects on chromatin structure and gene expression?
Does this modification create or disrupt interaction surfaces for chromatin regulators?
Genomic distribution and dynamics:
What is the genomic distribution of HIST1H1C K96 mono-methylation?
How does this distribution change during development, differentiation, or disease progression?
What is the relationship between this modification and chromatin accessibility?
Cross-talk with other epigenetic mechanisms:
How does HIST1H1C K96 mono-methylation interact with other histone modifications?
What is its relationship with DNA methylation and chromatin remodeling?
Does it influence or respond to non-coding RNA regulation?
Evolutionary conservation and specialization:
Is this modification conserved across species?
Does it serve similar functions in different organisms or cell types?
How has the regulation and function of this modification evolved?
Addressing these questions will require integrated approaches combining biochemical, genomic, and functional studies with advanced imaging and computational analysis.