The antibody is optimized for detecting HIST1H1C in diverse experimental contexts.
Purpose: Quantitative analysis of HIST1H1C protein levels in cell lysates.
Validation: Tested in human cell lines (e.g., HeLa, A549) and tissues (e.g., lung, kidney).
Observed Band: ~21 kDa (expected molecular weight).
Controls: Knockout cell lines (e.g., HIST1H1C-KO A549) to confirm specificity .
Purpose: Localization of HIST1H1C in tissue sections or cultured cells.
Protocol:
Fixation with PFA (3.7%, 10 min).
Permeabilization and blocking.
Primary antibody incubation (1:50–1:200, 1 hr at RT).
Secondary antibody (e.g., Alexa Fluor® 488-conjugated anti-rabbit IgG).
Use Case: Mapping HIST1H1C distribution in nuclear periphery or nucleoli .
Purpose: Identification of genomic regions bound by HIST1H1C.
Workflow:
Crosslinking, chromatin shearing, and immunoprecipitation.
Sequencing ChIP-enriched DNA to map binding sites.
Relevance: Studies on HIST1H1C’s role in chromatin architecture and epigenetic regulation .
HIST1H1C is a linker histone that stabilizes nucleosome structure and regulates chromatin compaction. Key roles include:
Immune Response: Modulates interferon-β (IFN-β) production by interacting with IRF3 and antagonizing viral proteins like influenza NS2 .
Cancer Biology: Mutations in H1 variants (e.g., HIST1H1C) drive lymphoma by disrupting 3D chromatin organization, leading to oncogene activation .
Chromatin Architecture: Enriched in low-GC, heterochromatic regions but excluded from nucleoli and high-GC regions .
Post-Translational Modifications:
| Application | Dilution | Cell/Tissue Tested | Outcome |
|---|---|---|---|
| WB | 1:500 | A549, HeLa | Clear 21 kDa band; absent in KO cells |
| IHC | 1:100 | HepG2, Jurkat | Nuclear periphery staining |
| ChIP | 1:200 | HeLa | Enrichment at low-GC genomic regions |
Cross-Reactivity: Potential interference with other H1 variants (e.g., H1.4) due to sequence homology .
Validation Gaps: Limited public data on ChIP-seq resolution; requires optimization for specific cell types.
Epigenetic Studies: Requires pairing with H3K9me2/HP1α markers to confirm heterochromatin association .
HIST1H1C encodes histone H1.2, a linker histone variant that compacts nucleosomes into higher-order chromatin fibers and controls genome organization. Histone H1.2 belongs to the "low-GC" group of H1 variants (along with H1.0, H1.3, and H1.5) that are predominantly enriched at the nuclear periphery and in low-GC genomic regions . This specific nuclear distribution pattern is universally observed across multiple cell lines, indicating a fundamental role in chromatin architecture . Functionally, H1.2 acts as a transcriptional repressor by limiting chromatin accessibility, helping to sequester developmental genes into architecturally inaccessible genomic compartments . Recent studies demonstrate that H1.2 is largely excluded from domains where histone H3K4 is hypermethylated or H4K8 is hyperacetylated, supporting its role as a repressor of histone modifications .
The HIST1H1C (Ab-164) Antibody can be used in multiple experimental applications including immunofluorescence (IF), chromatin immunoprecipitation (ChIP), ChIP-sequencing, and western blotting. For immunofluorescence, the antibody works optimally after fixing cells with 4% paraformaldehyde (20 minutes at room temperature) followed by permeabilization with methanol (10 minutes at room temperature) . For ChIP applications, the antibody can effectively precipitate chromatin fragments associated with H1.2, allowing for the identification of genomic binding sites and subsequent PCR or sequencing analysis . When performing ChIP-Seq with this antibody, researchers typically observe enrichment of H1.2 at low-GC regions and the nuclear periphery, consistent with its known distribution patterns . Western blotting applications require standard protocols with careful consideration of blocking conditions (5% BSA is recommended) to reduce background signal .
The HIST1H1C (Ab-164) Antibody specifically recognizes histone H1.2, distinguishing it from other H1 variants due to its high specificity. This specificity is critical because different H1 variants exhibit distinct nuclear distribution patterns and genomic localization profiles. For instance, while H1.2 (along with H1.3 and H1.5) is enriched at the nuclear periphery, H1.4 and H1X are more homogeneously distributed throughout the nucleus, with H1X showing particular abundance in nucleoli . Additionally, H1.2 belongs to the "low-GC" group of H1 variants enriched in low-GC genomic regions, whereas H1.4 and H1X are more abundant at high-GC regions . These distinct distribution patterns reflect different functional roles of H1 variants in chromatin organization and gene regulation. When designing experiments, it's essential to validate antibody specificity through appropriate controls to ensure accurate detection of H1.2 without cross-reactivity with other H1 variants.
For optimal immunofluorescence results with HIST1H1C (Ab-164) Antibody, cells should be directly grown on high-performance glass coverslips (0.17 mm thickness, 1.5 H) placed in multi-well plates. The recommended fixation protocol involves 4% paraformaldehyde for 20 minutes at room temperature, followed by permeabilization with methanol for 10 minutes at room temperature . After permeabilization, samples should be blocked with 5% bovine serum albumin diluted in PBS containing 0.1% Triton X-100 . Primary antibody incubation should be performed overnight at 4°C, followed by secondary antibody incubation (e.g., goat anti-rabbit IgG H+L conjugated to Alexa-488 or -647) for 1 hour at room temperature in the dark . Nuclear counterstaining can be performed with Hoechst (25 μg/ml) for 1 hour at room temperature in the dark, followed by mounting with Prolong Glass medium . This protocol preserves the nuclear architecture and ensures specific detection of H1.2 while minimizing background signal.
For ChIP-Seq experiments with HIST1H1C (Ab-164) Antibody, researchers should implement a rigorous protocol that begins with crosslinking chromatin using formaldehyde (typically 1% for 10 minutes at room temperature), followed by quenching with glycine. After cell lysis and sonication to generate chromatin fragments of 200-500 bp, immunoprecipitation should be performed with optimized antibody concentration (typically 2-5 μg per reaction) . Including appropriate controls is crucial: an IgG negative control and a positive control such as anti-H3 antibody . For data analysis, H1.2 enrichment should be normalized against input DNA and analyzed in relation to genomic features such as GC content, gene density, and chromatin accessibility . Based on previous studies, expect H1.2 to show strong correlation with low-GC content regions and enrichment at the nuclear periphery . For comparative analyses across cell lines or conditions, G-bands segmentation provides a useful approach to directly compare H1 variant binding profiles . When interpreting results, consider potential overlaps with histone modifications, as H1.2 is typically excluded from regions with active chromatin marks such as H3K4 methylation and H4K8 acetylation .
To investigate interactions between HIST1H1C (H1.2) and ribosomal proteins, a multi-faceted approach combining co-immunoprecipitation (Co-IP), ChIP, and proximity ligation assays (PLA) can be employed using the Ab-164 antibody. For Co-IP, nuclear extracts should be prepared under mild conditions to preserve protein-protein interactions, followed by immunoprecipitation with the HIST1H1C antibody and immunoblotting for specific ribosomal proteins (such as L22 and L7) . Sequential ChIP (re-ChIP) can determine if H1.2 and ribosomal proteins co-occupy the same genomic regions: perform initial ChIP with HIST1H1C antibody, elute the complexes, and then perform a second ChIP with antibodies against ribosomal proteins . To validate the genomic co-localization, PCR analysis of ChIP DNA should target genes known to be enriched for H1 binding, such as CG8066, Act57B, and Klp38B . For functional studies, RNA interference (RNAi) can be used to deplete histone H1.2, followed by ChIP to assess changes in ribosomal protein association with chromatin . Previous research has shown that depletion of histone H1 reduces the association of ribosomal proteins L22 and L7 with chromatin by several-fold in genes like CG8066, Act57B, and Klp38B, indicating that this association is H1-dependent .
To investigate H1.2's role in cellular mechanical behaviors and stress response, researchers can employ HIST1H1C (Ab-164) Antibody in a comprehensive experimental approach. Begin with immunofluorescence to visualize H1.2 distribution under normal and stress conditions, such as cytokine treatment or mechanical stimulation . Combine this with traction force assays, where cells are seeded onto fluorescently labeled BSA beads and the deformation of these beads is measured to quantify forces generated by individual cells before and after H1.2 depletion . For mechanistic studies, perform ChIP-Seq using the Ab-164 antibody to identify genome-wide binding sites of H1.2 under normal and stress conditions, focusing on genes involved in cytoskeletal regulation, contractility, and extracellular matrix (ECM) deposition . Additionally, analyze chromatin modifications (particularly H3K27Ac) in relation to H1.2 binding sites, as H1.2 has been shown to be required for cytokine-induced reprogramming of activating chromatin modifications . To assess functional outcomes, measure contractile force generation, cytoskeletal organization, cell motility, and ECM deposition in control versus H1.2-depleted cells . Based on previous findings, expect H1.2 depletion to significantly impact these mechanical behaviors, as H1.2 controls genome organization and cellular stress response through modulation of HDACs and BRD4 .
For investigating HIST1H1C mutations in lymphoma, a multi-modal approach utilizing the Ab-164 antibody is recommended. Begin by analyzing HIST1H1C expression and localization patterns in lymphoma samples versus normal B-cells using immunohistochemistry or immunofluorescence with the Ab-164 antibody . When analyzing patient samples, consider that H1C (HIST1H1C) mutations occur with a frequency of approximately 24.7% in diffuse large B-cell lymphomas (DLBCL), with enrichment in the MCD-DLBCL subtype . For mechanistic studies, perform ChIP-Seq using the Ab-164 antibody in lymphoma cells with wild-type versus mutant HIST1H1C to identify altered genomic binding patterns . Integration with Hi-C data is crucial, as H1 loss in lymphoma leads to decompaction of 3D chromatin architecture . Furthermore, gene expression analysis should be performed to identify deregulated genes, particularly focusing on developmental genes that are normally sequestered in architecturally inaccessible genomic compartments by H1 proteins . For functional validation, CRISPR-Cas9 can be used to introduce specific HIST1H1C mutations identified in patients, followed by assessment of cellular phenotypes including proliferation, self-renewal, and lymphomagenesis potential . Previous research indicates that H1 loss in germinal center B-cells confers enhanced fitness and self-renewal properties, ultimately leading to aggressive lymphoma with enhanced repopulating potential .
To minimize non-specific binding in ChIP experiments with HIST1H1C (Ab-164) Antibody, several optimization steps are crucial. First, implement a stringent blocking protocol using 5% BSA in PBS with 0.1% Triton X-100 for at least 1 hour at room temperature prior to antibody incubation . Pre-clearing the chromatin with protein A/G beads and non-specific IgG can significantly reduce background signal. Optimize antibody concentration through titration experiments (typically 2-5 μg is recommended, but this should be empirically determined for each experimental system) . Include appropriate negative controls in all experiments, such as IgG from the same species as the primary antibody, and positive controls like anti-H3 antibody . When designing washing steps, use increasingly stringent wash buffers (low salt, high salt, LiCl, and TE buffers) to remove non-specifically bound proteins while preserving specific interactions . For data analysis, compare enrichment patterns with previously published H1.2 distribution profiles, expecting enrichment at low-GC regions and correlations with specific chromatin modifications . If high background persists, consider crosslinking optimization, as excessive crosslinking can lead to non-specific interactions, while insufficient crosslinking may result in loss of transient interactions.
When studying differential distribution of H1.2 versus other H1 variants using the HIST1H1C (Ab-164) Antibody, comprehensive controls are essential for reliable results. Include antibody specificity controls by performing western blots on recombinant H1 variants or using cells with CRISPR-mediated knockout of specific H1 variants . For immunofluorescence experiments, include co-staining with markers of nuclear compartments such as HP1alpha for heterochromatin to correlate H1.2 distribution with chromatin states . When analyzing nucleolar localization, use established nucleolar markers like NPM1 for co-localization studies . For ChIP-Seq experiments, include parallel ChIP with antibodies against other H1 variants for direct comparison, and use G-bands segmentation as a framework for comparing binding profiles across variants . To account for cell cycle effects, synchronize cells using methods like Thymidine-Nocodazole treatment and analyze H1.2 distribution at different cell cycle stages . For validating functional hypotheses about differential roles of H1 variants, perform gene expression analysis after selective depletion of individual H1 variants using shRNA or CRISPR approaches . Additionally, include drug treatments that disrupt nuclear architecture, such as Actinomycin D for nucleolar disruption, to assess the dynamics of H1.2 redistribution compared to other variants .
To detect and distinguish phosphorylated forms of HIST1H1C (H1.2), researchers should employ phospho-specific antibodies in conjunction with the standard Ab-164 antibody. Based on research with other H1 variants, phosphorylation can significantly alter chromatin binding properties and nuclear distribution patterns of H1 histones . For instance, phosphorylated H1.2 at threonine 165 (H1.2-pT165) shows distinct nucleolar localization that is lost upon treatment with RNA transcription inhibitors like Actinomycin D, suggesting a role in nucleolar dynamics . When designing experiments, include appropriate controls to validate phospho-specific antibody specificity, such as lambda phosphatase treatment to remove phosphorylation marks, or site-directed mutagenesis of the phosphorylation sites to alanine residues . For comprehensive analysis, combine immunofluorescence using phospho-specific antibodies with cell cycle synchronization methods, as H1 phosphorylation levels fluctuate throughout the cell cycle . In ChIP-Seq experiments, compare binding profiles of total H1.2 versus phosphorylated H1.2 to identify genomic regions where phosphorylation may regulate H1.2 function . When analyzing results, consider the dynamic nature of H1 phosphorylation in response to cellular stimuli and stress conditions, as these modifications may reflect transient regulatory mechanisms rather than stable distribution patterns .
When investigating H1.2's role in 3D chromatin architecture using HIST1H1C (Ab-164) Antibody, several methodological considerations are critical. Combine ChIP-Seq with chromosome conformation capture techniques (Hi-C, 4C, or Micro-C) to correlate H1.2 binding with 3D genome organization . For optimal results, perform parallel experiments with and without H1.2 depletion to directly assess its impact on chromatin architecture . When depleting H1.2, consider potential compensatory mechanisms from other H1 variants, as complete loss of all H1 proteins is embryonic lethal but partial depletion can be tolerated through compensation . In the analysis phase, focus on topologically associating domains (TADs) and compartments, as research has shown that H1 loss drives lymphoma primarily through 3D chromatin reorganization . Importantly, distinguish between direct effects of H1.2 on chromatin architecture and secondary effects on gene expression, as H1 proteins function as tumor suppressors by sequestering early developmental genes into architecturally inaccessible genomic compartments . For visualization of 3D chromatin structures in intact nuclei, combine immunofluorescence using the Ab-164 antibody with super-resolution microscopy techniques like STORM or PALM . When interpreting results, consider that the impact of H1.2 on chromatin architecture may vary between cell types and developmental stages, with lymphoid cells being particularly sensitive to H1 depletion or mutation .
To investigate H1.2's role in fibrosis and tissue remodeling, HIST1H1C (Ab-164) Antibody can be employed in a comprehensive experimental approach. Begin with immunohistochemistry or immunofluorescence on tissue sections from normal versus fibrotic tissues to assess H1.2 expression and localization patterns . Recent research has demonstrated a positive correlation between histone H1.0 and periostin (a canonical marker of fibroblast activation) in human heart samples, suggesting similar roles may exist for H1.2 in fibrotic responses . For functional studies in fibroblasts, perform H1.2 depletion using siRNA or CRISPR-Cas9, followed by assessment of key fibrotic phenotypes including contractile force generation, cytoskeletal regulation, motility, and ECM deposition . Traction force assays are particularly valuable, allowing measurement of forces generated by individual fibroblasts before and after H1.2 depletion . At the molecular level, perform ChIP-Seq with the Ab-164 antibody to identify H1.2 binding sites in normal versus activated fibroblasts, focusing on genes involved in fibrotic responses . Combine this with RNA-Seq to correlate changes in H1.2 occupancy with gene expression alterations during fibroblast activation . Previous research has shown that histone H1.0 controls genome organization and cellular stress response through modulation of HDACs and BRD4, mechanisms that likely apply to H1.2 as well .
For investigating HIST1H1C alterations in cancers beyond lymphoma, a multi-faceted approach using the Ab-164 antibody is recommended. Begin with immunohistochemistry or immunofluorescence analysis across cancer tissue microarrays to assess H1.2 expression and localization patterns in different tumor types compared to matched normal tissues . For mechanistic studies, perform ChIP-Seq using the Ab-164 antibody in cancer cell lines with different H1.2 expression levels to identify altered genomic binding patterns . This should be integrated with gene expression analysis to correlate H1.2 binding with transcriptional alterations in cancer cells . For functional validation, manipulate H1.2 levels through overexpression or knockdown in cancer cell lines, followed by assessment of hallmark cancer phenotypes including proliferation, migration, invasion, and response to therapy . When analyzing results, consider potential compensatory mechanisms between different H1 variants, as studies have shown that certain cancer cell lines lack H1.3 and H1.5, leading to redistribution of H1.0 and H1.4 throughout the nucleus . Additionally, explore potential connections between H1.2 and the immune response, as H1-depleted cells exhibit a robust interferon response triggered by the expression of repetitive elements . This approach can provide insights into how H1.2 alterations contribute to cancer development and progression beyond the established role in lymphomagenesis.
To investigate the relationship between H1.2 and cellular aging or senescence using the HIST1H1C (Ab-164) Antibody, researchers should implement a comprehensive experimental strategy. Begin with immunofluorescence analysis comparing H1.2 distribution patterns in young versus senescent cells, such as early versus late passage primary fibroblasts . For senescence induction models, use established methods like replicative senescence, oncogene-induced senescence, or DNA damage-induced senescence, followed by assessment of H1.2 expression, localization, and chromatin binding . ChIP-Seq using the Ab-164 antibody can identify genome-wide redistribution of H1.2 during senescence, particularly focusing on senescence-associated heterochromatin foci (SAHF) and regions with altered chromatin accessibility . Since H1 variants show differential nuclear patterns, with H1.2 typically enriched at the nuclear periphery, evaluate whether this distribution changes during senescence . For mechanistic insights, investigate the relationship between H1.2 and known senescence regulators through co-immunoprecipitation and proximity ligation assays . Additionally, examine whether H1.2 affects the senescence-associated secretory phenotype (SASP) by manipulating H1.2 levels and measuring inflammatory cytokine production . Compare findings in different cell types, including both normal cells (fibroblasts, epithelial cells) and cancer cells, as senescence responses can vary significantly between cell types .
To assess H1.2's impact on gene expression during cellular stress response, multiple complementary techniques involving the HIST1H1C (Ab-164) Antibody should be employed. Begin with stress induction experiments using relevant stimuli such as cytokines, mechanical stress, or metabolic stress, followed by ChIP-Seq with the Ab-164 antibody to map genome-wide redistribution of H1.2 under stress conditions . Parallel RNA-Seq analysis can identify stress-responsive genes and correlate their expression changes with alterations in H1.2 occupancy . For mechanistic insights, focus on H1.2's interaction with chromatin modifiers, particularly HDACs and BRD4, as research has shown that histone H1.0 controls cellular stress response through modulation of these factors . Specifically, investigate H1.2's role in reprogramming of the activating chromatin modification histone H3 lysine 27 acetylation (H3K27Ac) in response to stress stimuli . For functional validation, perform H1.2 depletion or overexpression prior to stress induction, followed by assessment of cellular responses including contractile force generation, cytoskeletal regulation, and ECM deposition . Time-course experiments are particularly valuable, as they can reveal dynamic changes in H1.2 distribution and gene regulation during the initiation, adaptation, and resolution phases of stress responses . Previous research suggests that proper histone stoichiometry is necessary for stress response in fibroblasts, and that the window for modulating chromatin architecture to prevent stress response closes once the transcriptional program is activated .
Several emerging technologies promise to enhance the utility of HIST1H1C (Ab-164) Antibody in future chromatin research. Super-resolution microscopy techniques like STORM, PALM, and lattice light-sheet microscopy will enable real-time visualization of H1.2 dynamics in living cells with unprecedented spatial resolution, providing insights into its role in 3D chromatin organization . Single-cell ChIP-Seq and CUT&TAG approaches can reveal cell-to-cell variability in H1.2 distribution patterns, particularly relevant in heterogeneous populations such as tumor samples or developing tissues . CRISPR-based technologies for targeted epigenome editing (using dCas9 fused to chromatin modifiers) can be combined with the Ab-164 antibody to investigate causal relationships between H1.2 binding, chromatin modifications, and gene expression . Proximity labeling methods like BioID or APEX2 fused to H1.2 can identify novel protein interaction partners in different cellular compartments and conditions . Multi-omics approaches integrating ChIP-Seq with Hi-C, ATAC-Seq, and RNA-Seq will provide comprehensive views of how H1.2 coordinates 3D genome organization with chromatin accessibility and gene expression . Mass spectrometry-based approaches can identify post-translational modifications of H1.2 and their functional significance in different cellular contexts . Finally, organ-on-chip and 3D organoid models will allow investigation of H1.2 functions in more physiologically relevant environments that better recapitulate tissue architecture and mechanical properties .