The HIST1H1C (Ab-45) antibody is validated for multiple techniques, with dilution recommendations varying by application:
Overexpression of histone H1.2 (HIST1H1C) in retinal cells (rMC-1) upregulates autophagy markers (e.g., ATG12–ATG5, LC3B-II) and promotes inflammation (Ccl2, Il6) while reducing cell viability . Conversely, knockdown of HIST1H1C reduces basal and stress-induced autophagy, mitigates high glucose-induced inflammation, and improves cell survival . These findings suggest HIST1H1C is a therapeutic target for diabetic retinopathy.
Autophagy Regulation: HIST1H1C upregulates SIRT1/HDAC1, deacetylating H4K16 and activating ATG proteins .
Inflammation: Overexpression correlates with glial activation (elevated GFAP) and proinflammatory cytokine release .
HIST1H1C (H1.2) is upregulated in hepatocellular carcinoma (HCC) and promotes tumor progression via STAT3 activation. Overexpression in HCC cell lines accelerates proliferation, while knockdown inhibits growth . Mechanistic studies reveal H1.2 enhances STAT3 binding to its promoter, driving oncogenic pathways .
Prognostic Biomarker: Elevated H1.2 levels may indicate aggressive HCC subtypes.
Therapeutic Target: Inhibiting H1.2-STAT3 interactions could suppress tumor growth.
The Ab-45 variant is distinct from other HIST1H1C antibodies due to its K45-specific epitope. Below is a comparison with other HIST1H1C-targeting antibodies:
HIST1H1C (also known as Histone H1.2) is a member of the linker histone H1 family that binds to linker DNA between nucleosomes, forming the macromolecular structure known as the chromatin fiber. Histones H1 are necessary for the condensation of nucleosome chains into higher-order structured fibers and act as regulators of gene transcription through chromatin remodeling, nucleosome spacing, and DNA methylation .
The Ab-45 antibody specifically recognizes an epitope around the lysine 45 (Lys45) residue of human Histone H1.2. The immunogen used to generate this antibody is a synthetic peptide directed towards the middle region of human HIST1H1C with the sequence "ASGSFKLNKK AASGEAKPKV KKAGGTKPKK PVGAAKKPKK AAGGATPKKS" . This antibody enables specific detection of HIST1H1C, distinguishing it from other H1 variants.
The HIST1H1C (Ab-45) antibody has been validated for multiple experimental techniques:
Western Blotting (WB) with recommended dilutions of 1:100-1:1000
Chromatin Immunoprecipitation (ChIP)
Immunohistochemistry (IHC) with recommended dilutions of 1:10-1:100
This versatility makes it a valuable tool for researchers investigating histone H1.2 expression, localization, and function in various experimental contexts. When using this antibody for ChIP applications, it has been successfully employed to identify genomic regions enriched for H1.2 binding, particularly in studies examining gene expression regulation .
The HIST1H1C (Ab-45) antibody demonstrates cross-reactivity with multiple species, making it suitable for comparative studies across different model organisms. Based on sequence homology and experimental validation, the antibody reactivity includes:
Human (100% reactivity)
Mouse (86% predicted reactivity)
Rat (86% predicted reactivity)
Cow (86% predicted reactivity)
Dog (86% predicted reactivity)
Guinea Pig (93% predicted reactivity)
Horse (86% predicted reactivity)
This broad reactivity profile is particularly valuable for researchers conducting evolutionary or comparative studies of histone function across different species.
For optimal performance and longevity of the HIST1H1C (Ab-45) antibody, researchers should:
Store the antibody at -20°C or -80°C immediately upon receipt
Avoid repeated freeze-thaw cycles as these can compromise antibody activity
The antibody is supplied in liquid form containing 50% Glycerol, 0.01M PBS (pH 7.4), and 0.03% Proclin 300 as a preservative
When working with the antibody, aliquot into smaller volumes for single use to avoid repeated freeze-thaw cycles
Proper storage and handling are crucial for maintaining antibody specificity and sensitivity, particularly for applications like ChIP where antibody quality directly impacts experimental outcomes.
Optimizing ChIP protocols for HIST1H1C (Ab-45) antibody requires several key considerations:
When analyzing ChIP-seq data, researchers should note that unlike core histones, H1 variants occur across the genome but with specific enrichment patterns that may correlate with transcriptional activity or chromatin states .
HIST1H1C (H1.2) has several distinctive functional characteristics compared to other H1 variants:
Expression pattern: H1.2 is ubiquitously expressed and produced in a replication-dependent manner, unlike H1.0 and H1X which are replication-independent. This suggests specific roles during cell proliferation .
Genomic distribution: Genome-wide studies have revealed specific features for H1.2 at promoters and across the genome. Unlike H1.5, which shows enrichment in genic and intergenic regions of differentiated human cells but not in embryonic stem cells, H1.2 has distinct binding patterns .
Transcriptional effects: Different H1 variants control the expression of different subsets of genes. While H1.0 has been characterized as a repressor in multiple cell types including mouse fibroblasts, human ESCs, and cancer cells, other variants like H1x appear to function as activators in breast cancer cells . The specific transcriptional impact of H1.2 depends on genomic context and cell type.
Post-translational modifications: H1 variants, including H1.2, have distinct patterns of post-translational modifications that modulate their interactions with different partners, potentially explaining some of their specific functions .
This functional diversity among H1 variants highlights their importance as underappreciated facets of chromatin dynamics that operate independently in various chromatin-based processes .
The relationship between histone H1.2 and ribosomal proteins represents an intriguing area of chromatin biology. To investigate this relationship using the HIST1H1C (Ab-45) antibody, researchers can employ several strategic approaches:
Co-immunoprecipitation (Co-IP): Use the HIST1H1C (Ab-45) antibody to pull down H1.2 and associated proteins, followed by western blotting to detect specific ribosomal proteins (e.g., L22, L7). Similar approaches have previously demonstrated that nuclear ribosomal proteins co-purify with histone H1 .
Sequential ChIP (Re-ChIP): Perform ChIP with HIST1H1C (Ab-45) antibody followed by a second immunoprecipitation with antibodies against ribosomal proteins to identify genomic regions where both proteins co-localize.
Proximity ligation assay (PLA): This technique can detect protein-protein interactions in situ between H1.2 and ribosomal proteins, providing spatial information about their interaction within the nucleus.
Depletion studies: Following H1.2 knockdown or knockout, examine changes in ribosomal protein association with chromatin. Previous research has shown that upon depletion of H1, the association of ribosomal proteins L22 and L7 with chromatin was lost .
Subcellular fractionation: Isolate nuclear fractions and examine co-sedimentation of H1.2 and ribosomal proteins. Previous studies have shown that nuclear fractions containing histone H1 also contained tagged ribosomal protein L22 under various salt conditions, supporting their specific interaction .
It's important to note that the interaction between H1 and ribosomal proteins appears to be specific to the nucleus, as native H1 isolated from nuclei failed to interact with cytoplasmic 40S, 60S, or 80S ribosomes .
To investigate how HIST1H1C impacts transcriptional regulation, researchers can implement multiple complementary approaches:
ChIP-seq with HIST1H1C (Ab-45) antibody: This approach can map genome-wide distribution of H1.2, identifying regions of enrichment or depletion and correlating these patterns with gene expression data. Previous mapping of H1 variants has revealed their distribution relative to genomic features and chromatin states .
RNA-seq following H1.2 manipulation: Compare transcriptomes after H1.2 depletion (via RNAi or CRISPR) or overexpression to identify H1.2-regulated genes. Previous studies have shown that overexpression of ribosomal protein L22, which interacts with H1, caused transcriptional repression of two-thirds of the genes suppressed by histone H1 .
ChIP-qPCR at specific loci: Using the HIST1H1C (Ab-45) antibody, assess H1.2 occupancy at promoters, enhancers or gene bodies of interest. This approach has been used to detect the physical association of H1 and ribosomal proteins with chromatin .
Integration with epigenomic data: Correlate H1.2 binding with histone modifications, DNA methylation, and chromatin accessibility. Studies have shown positive and negative correlations between H1 variants and specific histone modifications like H3K9me3 and H3K4me3 .
Cell type-specific analyses: Compare H1.2 distribution and function across different cell types or differentiation states. For example, H1.5 shows enrichment patterns in differentiated human cells that are not observed in embryonic stem cells .
These approaches can reveal both direct and indirect effects of H1.2 on gene expression, helping to distinguish its specific functions from those of other H1 variants.
When conducting experiments with HIST1H1C (Ab-45) antibody, the following controls and validation steps are critical:
Antibody specificity validation:
Western blot analysis to confirm the antibody detects a single band of the expected molecular weight (approximately 21.4 kDa)
Peptide competition assay using the immunizing peptide (around Lys45) to confirm specific binding
Comparison with other validated HIST1H1C antibodies targeting different epitopes
Testing in H1.2 knockout/knockdown samples to confirm absence of signal
ChIP controls:
IgG negative control from the same species (rabbit)
Input chromatin (non-immunoprecipitated) control
Positive control loci known to be enriched for H1.2
Negative control loci known to lack H1.2 binding
Cross-reactivity assessment:
Testing in multiple species to confirm predicted reactivity
Testing potential cross-reactivity with other H1 variants, particularly those with sequence similarity around the Lys45 region
Technical replicates:
Minimum of three biological replicates to ensure reproducibility
Technical duplicates within each experiment
Batch controls:
When comparing different experimental conditions, ensure the same antibody lot is used
Include common reference samples across batches if different lots must be used
These validation steps are particularly important given that histone variants can have high sequence similarity, and ensuring antibody specificity is crucial for accurate interpretation of experimental results.
Differentiating between the specific functions of H1.2 (HIST1H1C) and other H1 variants requires careful experimental design:
Variant-specific knockdown/knockout: Use siRNA, shRNA, or CRISPR-Cas9 approaches targeting unique regions of H1.2 mRNA. Monitor potential compensatory upregulation of other H1 variants, which may confound results.
Rescue experiments: After H1.2 depletion, reintroduce either wild-type H1.2 or other H1 variants to determine which functions are specific to H1.2 versus those common to the H1 family.
Variant-specific ChIP-seq: Use the HIST1H1C (Ab-45) antibody alongside antibodies specific to other H1 variants to compare genomic distributions. Previous genome-wide distribution analyses have revealed specific features for H1.2 at promoters and genome-wide that distinguish it from other variants .
Domain swapping experiments: Create chimeric H1 proteins containing domains from different variants to identify which regions confer variant-specific functions.
Post-translational modification analysis: Different H1 variants have distinct modification patterns. Using modification-specific antibodies alongside HIST1H1C (Ab-45) antibody can reveal variant-specific regulatory mechanisms .
Cell type-specific analyses: Compare the functions of H1.2 across different cell types, as studies show H1 variants control the expression of different subsets of genes depending on cellular context .
This multi-faceted approach can help distinguish the unique functions of H1.2 from the shared properties of the H1 family.
Investigating the dynamics of HIST1H1C during cellular transitions involves several methodological approaches:
Temporal ChIP-seq analysis: Using the HIST1H1C (Ab-45) antibody, perform ChIP-seq at multiple timepoints during differentiation or stress response to track changes in H1.2 genomic distribution. This approach has revealed that some H1 variants, like H1.5, show different enrichment patterns between differentiated cells and embryonic stem cells .
Correlation with chromatin state changes: Integrate H1.2 ChIP-seq data with maps of chromatin accessibility (ATAC-seq, DNase-seq) and histone modifications to understand how H1.2 redistribution correlates with changing chromatin environments.
Expression analysis: Monitor changes in H1.2 protein and mRNA levels during differentiation or stress using the antibody for western blotting and RT-qPCR, respectively.
Live-cell imaging: If possible, use fluorescently tagged H1.2 to track its dynamics in real-time during cellular transitions.
Functional perturbation: Deplete H1.2 at different stages of differentiation or stress response to determine when its function is most critical.
Notably, while H1.2 is replication-dependent, other variants like H1.0 accumulate in terminally differentiated cells, suggesting transition-specific roles . During human ESC differentiation, for example, H1.0 (another H1 variant) is recruited to pluripotency genes, demonstrating how H1 variants can have stage-specific functions in chromatin regulation .
The HIST1H1C (Ab-45) antibody recognizes an epitope around lysine 45, which can be a site for post-translational modifications (PTMs). Researchers can leverage this antibody along with modification-specific antibodies to study H1.2 regulation:
Sequential ChIP (Re-ChIP): First immunoprecipitate with HIST1H1C (Ab-45) antibody, then with antibodies against specific PTMs (e.g., methylation, acetylation) to identify genomic regions containing modified H1.2.
Complementary antibody approach: Compare ChIP-seq results using HIST1H1C (Ab-45) antibody versus specific antibodies that recognize modified forms, such as methylated Lys45 (2meLys45) or acetylated forms of H1.2. Several modification-specific antibodies are available that target modifications at sites including Lys45, Lys84, Lys96, and Lys186 .
Mass spectrometry analysis: After immunoprecipitation with the HIST1H1C (Ab-45) antibody, perform mass spectrometry to catalog the full range of PTMs present on the precipitated H1.2.
Functional studies: Investigate how specific PTMs affect H1.2 function by introducing point mutations that either prevent modification (e.g., K45R) or mimic constitutive modification (e.g., K45Q for acetylation).
Enzyme inhibitor studies: Treat cells with inhibitors of histone-modifying enzymes (HDACs, HMTs, etc.) and assess changes in H1.2 modification patterns and genomic distribution.
These approaches can reveal how PTMs regulate H1.2 function, as different modifications may modulate its interaction with different partners and explain specific functions observed for this variant .
H1.2 (HIST1H1C) is crucial for chromatin condensation into higher-order structures. To investigate its role in 3D genome organization:
Integration with chromosome conformation data: Correlate H1.2 ChIP-seq data (using the HIST1H1C (Ab-45) antibody) with Hi-C, Micro-C, or 4C data to understand how H1.2 binding relates to topologically associating domains (TADs), compartments, and specific chromatin loops.
Electron microscopy studies: Immunogold labeling with the HIST1H1C (Ab-45) antibody can reveal the spatial distribution of H1.2 within condensed chromatin structures at the ultrastructural level.
Super-resolution microscopy: Techniques like STORM or PALM, using fluorescently labeled HIST1H1C (Ab-45) antibody, can visualize H1.2 distribution within the nucleus at near-molecular resolution.
H1.2 depletion followed by Hi-C: Examine how selective depletion of H1.2 affects 3D chromatin organization compared to depletion of other H1 variants. This approach can reveal H1.2-specific roles in maintaining certain chromatin structures.
Targeted recruitment experiments: Using systems like dCas9-H1.2 fusions, artificially recruit H1.2 to specific genomic loci and observe changes in local and distal chromatin organization.
H1 histones are necessary for the condensation of nucleosome chains into higher-order structured fibers , and H1.2 may have specific effects on chromatin compaction that differ from other variants. Understanding these specific roles is crucial for comprehending the mechanisms of gene regulation and chromatin dynamics.
Researchers may encounter several challenges when working with the HIST1H1C (Ab-45) antibody:
Cross-reactivity with other H1 variants:
Problem: The antibody may detect other H1 variants due to sequence similarity.
Solution: Validate specificity using knockout controls; perform western blots with recombinant H1 variants to assess cross-reactivity; consider using complementary approaches with antibodies targeting different epitopes.
Epitope masking due to protein interactions or modifications:
Problem: The epitope around Lys45 may be obscured by protein-protein interactions or modified in certain contexts.
Solution: Test different extraction and fixation conditions; compare results with antibodies targeting different regions of H1.2; be aware that negative results may reflect epitope masking rather than absence of the protein.
ChIP efficiency issues:
Problem: Lower than expected enrichment in ChIP experiments.
Solution: Optimize crosslinking conditions (1% formaldehyde for 10 minutes is standard, but may need adjustment); increase antibody concentration (starting at 5μg per 25μg chromatin); ensure chromatin is adequately fragmented (200-500bp); include positive controls for ChIP efficiency.
Background in immunofluorescence:
Problem: High background signal in immunofluorescence applications.
Solution: Increase blocking time; use alternative blocking agents; optimize antibody concentration (starting with 1:100 dilution); include appropriate negative controls.
Batch-to-batch variability:
Problem: Different lots of polyclonal antibody may show variable specificity or sensitivity.
Solution: Purchase larger quantities of a single lot for long-term projects; validate each new lot against previous lots; maintain consistent experimental conditions across batches.
Addressing these challenges requires careful optimization and validation, particularly for applications like ChIP where antibody performance directly impacts data quality and interpretability.