The HIST1H1C (Ab-186) Antibody is supplied in liquid form, typically in a buffered aqueous glycerol solution . The exact formulation includes preservatives to maintain antibody stability and functionality:
Parameter | Specification |
---|---|
Form | Liquid |
Buffer | 50% Glycerol, 0.01M PBS, pH 7.4 |
Preservative | 0.03% Proclin 300 |
Conjugation | Unconjugated |
Purification Method | Antigen Affinity Purified |
Quantity | Typically 50μL or 100μL |
Isotype | IgG |
The antibody requires specific storage conditions to maintain its activity. For short-term storage (up to 2 weeks), refrigeration at 2-8°C is recommended, while long-term storage should be at -20°C to -80°C in small aliquots to prevent freeze-thaw cycles that could compromise activity .
The target of this antibody, HIST1H1C (also known as Histone H1.2), belongs to the H1 linker histone family, which are key chromatin architectural proteins that facilitate the formation of higher-order chromatin structures . The HIST1H1C gene is located on chromosome 6 in humans and its protein product plays a critical role in chromatin compaction.
HIST1H1C has numerous aliases in the scientific literature, including:
The UniProt ID for human HIST1H1C is P16403, providing a standardized reference for this protein in biological databases .
The HIST1H1C (Ab-186) Antibody has been validated for multiple experimental applications, making it a versatile tool for epigenetic and chromatin research.
Application | Validated | Recommended Dilution |
---|---|---|
Western Blot (WB) | Yes | 1:500-5000 or 1:100-1000 |
Immunoprecipitation (IP) | Yes | 1:200-2000 or 1:200-2000 |
ELISA | Yes | Not specified |
Immunohistochemistry (IHC) | Yes | 1:20-1:50 |
The antibody has demonstrated reactivity specifically with human samples, making it appropriate for studies using human cell lines and tissues .
Published data shows that the HIST1H1C (Ab-186) Antibody has been successfully used in immunoprecipitation analysis of 293 whole cell lysate and Western blot analysis of multiple cell lines including Hela, MCF-7, Jurkat, and HEK293 . These validation experiments confirm the antibody's specificity and utility across different experimental contexts.
Histone H1.2 (HIST1H1C) protein binds to linker DNA between nucleosomes, forming the macromolecular structure known as the chromatin fiber . H1 histones, including H1.2, are necessary for the condensation of nucleosome chains into higher-order structured fibers, playing a crucial role in chromatin compaction and genome organization.
Beyond its structural role, HIST1H1C also functions as a regulator of individual gene transcription through:
Research has shown that H1C, along with H1D and H1E, constitutes more than 90% of the H1 complement in B and T cells, highlighting its particular importance in the hematopoietic system . This predominant expression pattern suggests a specialized role for these H1 variants in immune cell function and development.
Increased resistance to x-ray induced apoptosis in thymocytes and intestine
Otherwise normal development and function
This suggests that while HIST1H1C has a role in apoptotic response to DNA damage, there may be functional redundancy with other H1 variants that compensates for its loss during normal development.
More severe phenotypes emerge when multiple H1 variants are knocked out simultaneously:
H1c−/−H1e−/− double knockout mice show mild growth retardation and abnormal rod photoreceptor packing in the retina
Triple knockout mice lacking H1c, H1e, and H10 exhibit more pronounced growth retardation, lower birth rates, and small thymus
These findings demonstrate that while individual H1 variants may have specific functions, they also work collectively to maintain proper chromatin structure and cellular function.
HIST1H1C (Histone Cluster 1, H1c) is a subtype of the linker histone H1 family that plays crucial roles in chromatin structure regulation and gene expression control. This histone variant is particularly important in epigenetic research because it undergoes various post-translational modifications (PTMs) including acetylation and methylation at specific lysine residues, which directly impact chromatin accessibility . These modifications regulate DNA-histone interactions and influence transcriptional activity by altering the chromatin microenvironment from heterochromatin (condensed, transcriptionally inactive) to euchromatin (less compact, transcriptionally active) states . HIST1H1C antibodies enable researchers to study these modifications and their functional consequences in various biological contexts, making them essential tools for understanding epigenetic regulation mechanisms.
HIST1H1C antibodies are specifically designed to recognize unique epitopes within the HIST1H1C protein, distinguishing it from other histone H1 subtypes. The specificity of these antibodies stems from their recognition of variant NH2-terminal regions that differ between histone H1 subtypes . Early attempts to generate antibodies against intact histone subtypes had limited success, leading researchers to develop antibodies against synthetic peptides or peptide fragments encompassing the variant NH2-terminal regions . HIST1H1C (Ab-186) antibodies typically target specific post-translational modifications at defined amino acid positions, such as acetylation or methylation at particular lysine residues. These antibodies undergo rigorous validation through enzyme-linked immunosorbent assays (ELISA) and protein immunoblot techniques against both purified subtypes and whole cell/nuclear extracts to ensure their specificity .
Multiple post-translational modifications of HIST1H1C can be detected using modification-specific antibodies. These include acetylation at various lysine positions (acLys16, acLys62, acLys84, acLys96), methylation (meLys45, meLys96, meLys186), and dimethylation (2meLys45) . Each modification-specific antibody targets a distinct epitope containing the modified residue, allowing researchers to investigate the presence and dynamics of these modifications in different biological contexts. The specificity of these antibodies is crucial for understanding how different modifications affect chromatin structure and gene expression. Researchers can employ these antibodies in various applications including immunoblotting, immunofluorescence, and chromatin immunoprecipitation to map the distribution and functional significance of each modification across the genome .
The HIST1H1C antibodies are versatile tools with applications across multiple experimental techniques in epigenetic research. Based on validated applications, these antibodies can be effectively employed in:
Chromatin Immunoprecipitation (ChIP): For mapping the genomic distribution of HIST1H1C and its modified forms, particularly useful for identifying target genes regulated by specific histone modifications .
Immunofluorescence (IF): For visualizing the spatial distribution of HIST1H1C variants within the nucleus and tracking changes in localization under different cellular conditions .
Immunocytochemistry (ICC): For detecting HIST1H1C in fixed cells while preserving cellular architecture to study its relationship with other nuclear components .
Western Blotting (WB): For quantitative assessment of HIST1H1C levels and its modifications in different cell types or treatment conditions .
ELISA: For sensitive quantification of specific HIST1H1C variants or modifications in complex biological samples .
The choice of application should be guided by the specific research question, with ChIP being particularly valuable for mechanistic studies linking histone modifications to gene regulation, while imaging techniques provide insights into spatial distribution patterns.
HIST1H1C antibodies enable researchers to investigate the complex relationship between histone modifications and gene expression through several methodological approaches. Chromatin immunoprecipitation followed by sequencing (ChIP-seq) represents a powerful technique where HIST1H1C modification-specific antibodies can be used to precipitate DNA fragments associated with the modified histone, followed by high-throughput sequencing to map genomic locations . This approach reveals the genomic distribution patterns of specific modifications and their correlation with gene expression states.
For integrative analyses, researchers can combine ChIP-seq data with transcriptome profiling (RNA-seq) to establish direct relationships between specific HIST1H1C modifications and transcriptional outcomes. For instance, acetylation of HIST1H1C at lysine residues generally correlates with active transcription by creating a less compact chromatin structure . Mechanistically, these modifications alter the charge quality and microenvironment of local chromatin, allowing entry of transcription factors and other transcriptional coactivators .
Sequential ChIP (re-ChIP) can also be employed to determine whether different modifications co-occur on the same HIST1H1C molecules, providing insights into the histone code hypothesis and combinatorial effects of modifications on gene regulation.
Proper experimental controls are essential for reliable ChIP experiments with HIST1H1C antibodies:
Input Control: A portion of the chromatin sample prior to immunoprecipitation should be set aside as the input control to normalize for differences in chromatin amount and fragmentation efficiency across samples.
Negative Control Antibody: An isotype-matched IgG control antibody should be used in parallel reactions to identify non-specific binding .
Positive Control Antibody: An antibody against a ubiquitous histone mark (e.g., H3K4me3 at active promoters) serves as a positive control for the ChIP procedure.
Peptide Competition Assay: Pre-incubation of the antibody with the synthetic peptide used as the immunogen (e.g., peptide sequence around acetyl-Lys62) should abolish specific signals, confirming antibody specificity .
Known Target Loci: Primers targeting genomic regions known to be enriched or depleted for the specific HIST1H1C modification should be included in qPCR validation.
Cross-reactivity Control: Testing the antibody against other histone H1 subtypes to confirm specificity for HIST1H1C using immunoblot assays against purified subtypes .
These controls collectively ensure signal specificity, procedural success, and reproducibility of ChIP experiments using HIST1H1C antibodies.
Optimal sample preparation for detecting HIST1H1C modifications requires careful consideration of several key factors:
Fixation Protocol: For ChIP and immunofluorescence applications, crosslinking with 1% formaldehyde for 10 minutes at room temperature preserves protein-DNA interactions while maintaining epitope accessibility. Over-fixation can mask epitopes and reduce antibody binding efficiency, particularly for certain modifications like acetylation .
Chromatin Fragmentation: For ChIP applications, sonication should be optimized to generate DNA fragments of 200-500 bp. Enzymatic digestion with micrococcal nuclease provides an alternative approach that may better preserve certain epitopes.
Extraction Buffers: For western blotting and immunoprecipitation, histone extraction buffers containing histone deacetylase inhibitors (e.g., sodium butyrate, trichostatin A) and phosphatase inhibitors are essential to preserve labile modifications. The pH should be carefully controlled as some modifications like acetylation are sensitive to acidic conditions .
Blocking Agents: BSA (1-3%) is preferred over milk for blocking solutions, as milk contains bioactive compounds that can affect histone modification status during lengthy incubations.
Epitope Retrieval: For fixed tissue samples, antigen retrieval methods using citrate buffer (pH 6.0) help expose masked epitopes without disrupting modification-specific antibody recognition sites.
These methodological refinements collectively enhance the signal-to-noise ratio and detection sensitivity for specific HIST1H1C modifications across different experimental platforms.
Verifying antibody specificity is critical for confident interpretation of experimental results. A multi-tiered validation approach is recommended:
Peptide Competition Assays: Pre-incubating the antibody with escalating concentrations of the immunizing peptide (modified versus unmodified) should progressively diminish signal intensity in a modification-specific manner .
Knockout/Knockdown Controls: Using CRISPR/Cas9-mediated knockout or siRNA knockdown of HIST1H1C provides definitive negative controls to confirm specificity.
Immunoblot Panel: Testing against purified recombinant histones with defined modifications confirms the antibody recognizes only the intended modification. Cross-reactivity with other histone variants should be systematically assessed using whole cell and nuclear extracts .
Enzyme Treatment: For acetylation-specific antibodies, treating samples with histone deacetylases should reduce signal intensity; similarly, demethylase treatment can confirm methylation-specific antibodies.
Orthogonal Detection Methods: Confirming findings using alternative detection methods such as mass spectrometry provides independent verification of modification status.
Indirect Immunofluorescence: Evaluating the subcellular distribution pattern of the signal can provide additional confirmation of specificity, as different histone modifications show characteristic nuclear distribution patterns .
This comprehensive validation ensures experimental observations genuinely reflect the target HIST1H1C modification rather than cross-reactivity artifacts.
Optimizing antibody dilutions and incubation conditions is essential for achieving reliable results across different applications:
Application | Recommended Dilution | Incubation Conditions | Special Considerations |
---|---|---|---|
Western Blot | 1:1000 - 1:2000 | Overnight at 4°C | Use PVDF membrane for better protein retention |
Immunofluorescence | 1:100 - 1:500 | 1-2 hours at RT or overnight at 4°C | Permeabilization critical for nuclear access |
ChIP | 2-5 μg per IP reaction | Overnight at 4°C with rotation | Pre-clearing with protein A/G beads reduces background |
ELISA | 1:500 - 1:2000 | 1-2 hours at RT or overnight at 4°C | BSA blocking preferred over milk |
Immunocytochemistry | 1:100 - 1:500 | 2 hours at RT or overnight at 4°C | Triton X-100 (0.1-0.3%) for nuclear permeabilization |
For all applications, initial titration experiments are recommended to determine optimal antibody concentration for specific experimental conditions . Signal-to-noise ratio should guide final dilution selection. For modification-specific antibodies, longer incubation times at lower temperatures (4°C) generally improve specific binding while reducing background. The addition of 0.1% Tween-20 to wash buffers helps minimize non-specific interactions without disrupting specific antibody binding.
Interpreting changes in HIST1H1C modification patterns in disease models requires a systematic analytical approach. Researchers should consider:
Context-Dependent Interpretation: The same modification may have different functional consequences depending on genomic location and cellular context. For example, in diabetic retinopathy, acetylation of histones correlates with increased inflammatory gene expression, but the same modification might have different effects in other tissues .
Temporal Dynamics: Modifications should be analyzed across multiple time points as temporal changes may reveal causative versus consequential relationships. Early changes may represent primary disease drivers while later alterations might reflect compensatory mechanisms.
Correlation with Physiological Parameters: Changes in histone modifications should be correlated with relevant physiological or pathological parameters. In diabetic models, correlations between histone acetylation patterns and metabolic parameters (glucose levels, insulin resistance) provide mechanistic insights .
Modification Networks: Individual modifications should not be interpreted in isolation but considered as part of broader networks. For instance, diabetes downregulates Sirt1 (a histone deacetylase), which subsequently affects H3K9 acetylation patterns with downstream effects on multiple inflammatory pathways .
Functional Validation: Changes in modification patterns should be functionally validated through targeted manipulation of the specific modifying enzymes. For example, inhibitors of histone acetyltransferases can prevent acetylation and induction of inflammatory proteins in diabetic retina models .
This integrated interpretative framework allows researchers to distinguish between causal modifications driving disease pathogenesis and secondary epigenetic changes resulting from altered cellular environments.
Addressing cross-reactivity with related histone variants requires multiple complementary strategies:
Epitope Selection: Prioritize antibodies targeting unique sequence regions that differ between HIST1H1C and other H1 variants. The NH2-terminal region often contains sufficient sequence variation to enable specific recognition .
Validation Against Recombinant Proteins: Test antibody specificity against a panel of purified recombinant histone variants to quantify potential cross-reactivity.
Peptide Competition Assays: Conduct competition assays using both target and potentially cross-reactive peptides to determine relative binding affinities.
Two-Dimensional Immunoblotting: Separate histone variants by isoelectric focusing followed by SDS-PAGE to resolve closely related variants that may co-migrate in one-dimensional separation.
Mass Spectrometry Validation: Use targeted mass spectrometry to independently confirm the identity of immunoprecipitated proteins and distinguish between closely related variants.
Knockout Controls: Utilize genetic models with specific histone variant knockouts to confirm antibody specificity in a biological context.
Sequential Immunoprecipitation: For ChIP applications, perform sequential immunoprecipitation with antibodies against different histone variants to identify uniquely bound genomic regions versus regions bound by multiple variants.
These approaches collectively minimize the risk of misinterpretation due to cross-reactivity with other histone H1 subtypes that share sequence similarity with HIST1H1C.
Distinguishing technical artifacts from biological variation in ChIP-seq experiments with HIST1H1C antibodies requires rigorous quality control:
Technical Replicates: Multiple technical replicates should show high correlation (Pearson r > 0.9) for peak locations and intensities. Peaks appearing inconsistently across replicates likely represent artifacts.
Input Normalization: All ChIP-seq signals should be normalized to input control to account for biases in chromatin accessibility, DNA sequence composition, and sonication efficiency.
Peak Shape Analysis: Genuine histone modification peaks typically show characteristic profiles—sharp peaks for transcription factors versus broader domains for histone modifications. Irregular peak shapes may indicate technical issues.
Spike-in Controls: Adding exogenous chromatin (e.g., from another species) provides an internal control for immunoprecipitation efficiency and normalization across samples.
Batch Effect Correction: Computational methods should be employed to identify and correct for batch effects when comparing samples processed at different times.
Biological Validation: Key findings should be validated using orthogonal techniques such as ChIP-qPCR, CUT&RUN, or CUT&Tag on independent biological samples.
Control for Antibody Lot Variation: Different antibody lots may show variation in specificity and sensitivity. Critical experiments should be repeated with multiple antibody lots or validated with peptide competition assays .
By implementing these quality control measures, researchers can confidently distinguish genuine biological variation in HIST1H1C modification patterns from technical artifacts, enhancing the reproducibility and reliability of epigenomic analyses.
Integrating HIST1H1C modification data with other epigenetic datasets provides a comprehensive understanding of chromatin regulation:
Multi-omics Integration: Combine HIST1H1C ChIP-seq data with other histone modifications (H3K4me3, H3K27ac, H3K9me3), DNA methylation profiles, chromatin accessibility data (ATAC-seq), and transcriptome data (RNA-seq) to build comprehensive regulatory networks .
Spatial Organization Analysis: Integrate HIST1H1C modification patterns with chromosome conformation capture data (Hi-C, 4C) to understand how these modifications influence three-dimensional chromatin architecture and gene expression.
Temporal Dynamics: Analyze HIST1H1C modifications across developmental stages or disease progression to identify critical transition points where epigenetic reprogramming occurs.
Computational Modeling: Employ machine learning approaches to identify combinatorial patterns of modifications that predict specific gene expression outcomes or chromatin states.
Enzyme-Modification Relationships: Correlate the activity and expression of modifying enzymes (HATs, HDACs, HMTs) with HIST1H1C modification patterns to establish regulatory mechanisms . For example, Sirt1 downregulation in diabetic retinopathy models correlates with increased H3K9 acetylation at specific promoters .
This integrative approach reveals how HIST1H1C modifications cooperate with other epigenetic mechanisms to orchestrate gene expression programs in normal development and disease states.
Several cutting-edge technologies are revolutionizing the study of HIST1H1C modifications at single-cell resolution:
Single-Cell CUT&Tag: This technique combines the sensitivity of CUT&Tag with single-cell isolation to profile HIST1H1C modifications across individual cells, revealing heterogeneity within seemingly homogeneous populations.
scChIC-seq (Single-cell Chromatin Immunocleavage sequencing): Uses antibody-directed cleavage to map histone modifications in single cells with improved sensitivity compared to traditional ChIP-based approaches.
Mass Cytometry (CyTOF) with Histone Modification Antibodies: Enables simultaneous quantification of multiple HIST1H1C modifications alongside cellular proteins in single cells, providing insights into modification co-occurrence patterns.
CRISPR-Display: Allows visualization of specific HIST1H1C modifications at defined genomic loci in living cells, enabling real-time tracking of modification dynamics during cellular processes.
Super-Resolution Microscopy: Techniques like STORM and PALM combined with modification-specific antibodies permit visualization of HIST1H1C modification distributions within the nucleus at nanometer resolution, revealing previously undetectable spatial organization patterns.
Combinatorial Indexing: Enables profiling of multiple epigenetic features (including HIST1H1C modifications) across thousands of single cells simultaneously, allowing identification of rare cell states and transition trajectories.
These emerging technologies are transforming our understanding of how HIST1H1C modifications contribute to cellular heterogeneity and epigenetic plasticity in development and disease contexts.