The 2-hydroxyisobutyryl-HIST1H1C (K109) Antibody was raised in a rabbit using a peptide around the site of 2-hydroxyisobutyryl-Lys (109) derived from Human Histone H1.2 as the immunogen. It’s a polyclonal, non-conjugated, IgG purified by antigen affinity. It finds uses in ELISA, Western blot (WB), Immunocytochemistry (ICC), Immunofluorescence (IF), and Chromatin Immunoprecipitation (ChIP). This antibody reacts against the human histone HIST1H1C. It can detect the endogenous levels of HIST1H1C of human-origin. At the moment, there’s no function assigned to the K109 2-hydroxyisobutyryl modified lysine, and it’s still in need to be dilucidated. The HIST1H1C protein interacts with the linker DNA between nucleosomes, facilitating the chromatin condensation to higher-order fibers. And this histone also can affect the nucleosome spacing, chromatin remodeling, and DNA methylation, consequently modulating the gene expression. Therefore, it is vital for correct chromatin high structure formation, regulation, and maintenance.
The 2-hydroxyisobutyryl-HIST1H1C (K109) Antibody was generated in a rabbit using a peptide encompassing the site of 2-hydroxyisobutyryl-Lys (109) derived from Human Histone H1.2 as the immunogen. This antibody is a polyclonal, non-conjugated, IgG purified by antigen affinity. It exhibits utility in ELISA, Western blot (WB), Immunocytochemistry (ICC), Immunofluorescence (IF), and Chromatin Immunoprecipitation (ChIP). This antibody specifically recognizes the human histone HIST1H1C, enabling the detection of endogenous levels of HIST1H1C of human origin. Currently, the functional significance of the K109 2-hydroxyisobutyryl modified lysine remains undeciphered and warrants further investigation. The HIST1H1C protein interacts with the linker DNA between nucleosomes, playing a crucial role in chromatin condensation to higher-order fibers. Additionally, this histone influences nucleosome spacing, chromatin remodeling, and DNA methylation, ultimately modulating gene expression. Consequently, it is essential for the proper formation, regulation, and maintenance of chromatin's intricate higher-order structure.
Histone H1 protein binds to linker DNA between nucleosomes, contributing to the formation of the macromolecular structure known as the chromatin fiber. Histones H1 are indispensable for the condensation of nucleosome chains into higher-order structured fibers. They also act as regulators of individual gene transcription through chromatin remodeling, nucleosome spacing, and DNA methylation.
2-hydroxyisobutyryl-HIST1H1C (also known as Histone H1.2) represents a specific post-translational modification of the linker histone H1 variant found in mammals. Lysine 2-hydroxyisobutyrylation is a relatively recently identified protein post-translational modification initially discovered on histones that affects the association between histone and DNA . While histone acetylation and methylation have been extensively studied, 2-hydroxyisobutyrylation represents a novel epigenetic mark that may regulate distinct biological processes. The significance lies in its potential role in regulating transcriptional activity through altering chromatin structure and accessibility . Research shows that 2-hydroxyisobutyrylated histone sites are conserved across plants, humans, and mice, suggesting evolutionary importance .
The 2-hydroxyisobutyryl-HIST1H1C antibody has been validated for multiple applications in epigenetic and chromatin research:
These applications allow researchers to investigate the presence, localization, and function of 2-hydroxyisobutyrylated HIST1H1C in various experimental contexts. When selecting an antibody, researchers should verify that it has been validated specifically for their intended application and species of interest.
Determining antibody specificity requires multiple validation approaches:
Knockout/knockdown validation: Use HIST1H1C knockout cell lines (such as the commercially available knockout A549 cell line) as negative controls in western blots to confirm the absence of signal at the expected molecular weight .
Peptide competition assay: Pre-incubate the antibody with synthetic 2-hydroxyisobutyrylated peptides corresponding to the target epitope (peptide sequence around 2-hydroxyisobutyryl-Lys site) before application to samples. Loss of signal indicates specificity for the modified epitope .
Cross-reactivity testing: Test the antibody against other histone variants and other lysine modifications (acetylation, methylation, etc.) to ensure it specifically recognizes 2-hydroxyisobutyrylation.
Positive controls: Include samples known to contain the 2-hydroxyisobutyrylated histone, such as human cell lines shown to express this modification .
Multiple detection methods: Confirm findings using complementary techniques (e.g., mass spectrometry and immunoblotting) to validate specificity.
Site-specific antibodies for different lysine residues in HIST1H1C (such as K109 versus K116) recognize distinct epitopes surrounding these specific lysine residues. The differences include:
Epitope sequence: Each antibody is raised against a unique peptide sequence surrounding the specific modified lysine. For example, the K116-specific antibody recognizes the peptide sequence around the 2-hydroxyisobutyrylated Lys-116 site derived from Human Histone H1.2 .
Biological significance: Different lysine residues may have distinct functional roles. The modification at K116 may affect chromatin structure differently than modification at K109 or other sites.
Tissue/condition specificity: The patterns of modification at different lysine residues may vary across tissues, developmental stages, or disease states. Research in diabetic retinopathy, for instance, shows specific changes in HIST1H1C modification patterns .
Application performance: Antibodies targeting different sites may perform differently in various applications, potentially due to epitope accessibility or surrounding amino acid composition.
When selecting between site-specific antibodies, researchers should consider which specific lysine residue is most relevant to their research question and validate each antibody independently.
Designing experiments to investigate the functional relationship between 2-hydroxyisobutyryl-HIST1H1C and autophagy requires a multi-faceted approach:
Overexpression and knockdown studies:
Transfect cells with plasmids expressing human HIST1H1C (such as pH1.2) to investigate the effects of overexpression
Establish stable HIST1H1C knockdown cell lines using shRNA targeting HIST1H1C
Measure autophagy markers (ATG12–ATG5 complex, ATG7, ATG3, and LC3B conversion) via western blot following manipulation
Autophagy flux assessment:
Co-transfect cells with GFP-LC3 and HIST1H1C expression plasmids and count cells with >10 cytoplasmic GFP dots as autophagic cells (minimum 200 cells per treatment)
Treat cells with autophagy inhibitors (chloroquine at 50μM or bafilomycin A1 at 100nM for 12h) to assess flux by measuring SQSTM1/p62 degradation and LC3B-I to LC3B-II conversion
Mechanistic studies:
Investigate whether HIST1H1C regulates autophagy through SIRT1 and HDAC1 by examining H4K16 acetylation status
Use SIRT1 or HDAC1 inhibitors to determine if they can reverse the effects of HIST1H1C overexpression
In vivo models:
Cellular toxicity measurements:
This comprehensive approach provides multiple lines of evidence for the functional relationship between 2-hydroxyisobutyryl-HIST1H1C and autophagy in disease contexts.
Differentiating between the effects of 2-hydroxyisobutyrylation and other acylation modifications requires specialized approaches:
Comparative proteomics analysis:
Perform proteome-wide mapping of multiple acylation types (acetylation, succinylation, 2-hydroxyisobutyrylation) using specific antibodies for each modification
Compare modification patterns to identify sites unique to 2-hydroxyisobutyrylation versus sites that can be modified by multiple acylations
Research in rice showed that while 99 2-hydroxyisobutyrylated sites could be modified by multiple PTMs, 8,924 sites were unique to 2-hydroxyisobutyrylation
Site-directed mutagenesis:
Generate lysine-to-arginine mutants at specific sites to prevent modification
Create lysine-to-glutamine mutants to mimic acetylation
Develop specific mimetics for 2-hydroxyisobutyrylation if available
Compare phenotypes of different mutants to attribute functions to specific modifications
Enzyme manipulation:
Structural analysis:
Use techniques like circular dichroism or nuclear magnetic resonance to determine how each modification affects protein structure
Analyze surface accessibility of modified lysines (research indicates 2-hydroxyisobutyryllysine is less surface accessible than unmodified lysine)
Examine the propensity of modifications to occur in regions of intrinsic disorder and coils
Functional readouts:
Compare the effects of different modifications on downstream processes like transcription, chromatin accessibility, or protein-protein interactions
Use ChIP-seq to map genomic locations bound by differentially modified HIST1H1C
This systematic approach enables attribution of specific functions to different acylation modifications on HIST1H1C.
Histone H1 reduction leads to significant consequences for genome stability and replicative stress. These effects and their experimental measurement include:
Effects of H1 reduction on replication:
DNA fiber analysis methodology:
Transcription inhibition experiments:
Treat cells with transcription inhibitors like 5,6-dichlorobenzimidazole1-β-D-ribofuranoside (DRB)
Compare fork rate and fork asymmetry before and after inhibition
Research shows that both decreased fork rate and increased fork asymmetry caused by H1 reduction are reversed by transcription inhibition
DNA damage assessment:
Non-coding RNA analysis:
Perform RNA-seq to identify and quantify non-coding RNAs
Use RNA immunoprecipitation to detect RNA-chromatin associations
Assess the impact of histone H1 depletion on non-coding RNA levels and localization
These experimental approaches can comprehensively measure the consequences of histone H1 reduction on replicative stress and genome stability.
Investigating the cross-talk between 2-hydroxyisobutyrylation and other epigenetic modifications requires integrated approaches:
Sequential ChIP (Re-ChIP) experiments:
Perform initial ChIP with 2-hydroxyisobutyryl-HIST1H1C antibody
Re-immunoprecipitate the eluted material with antibodies against other modifications
Analyze the co-occurrence of 2-hydroxyisobutyrylation with other modifications at specific genomic loci
Map the genome-wide distribution patterns of co-occurring modifications
Mass spectrometry-based proteomics:
Use affinity enrichment with 2-hydroxyisobutyryl-specific antibodies followed by nano-HPLC/MS/MS
Identify peptides with multiple modifications
Quantify the abundance of different modification combinations
Previous work has identified 9,916 2-hydroxyisobutyryl lysine sites on 2,512 proteins in plants
Functional genomics approaches:
Conduct RNA-seq following manipulation of 2-hydroxyisobutyrylation levels
Perform ChIP-seq for 2-hydroxyisobutyrylation and other modifications
Integrate data to identify genes regulated by specific modification patterns
Use ATAC-seq to measure chromatin accessibility changes
Biochemical analysis of writer/eraser/reader enzymes:
Identify enzymes that regulate 2-hydroxyisobutyrylation (SIRT1, HDAC1)
Determine if these enzymes also regulate other modifications
Investigate whether readers of 2-hydroxyisobutyrylation also recognize other modifications
Analyze how inhibitors of specific enzymes affect multiple modification types
Computational analysis of modification motifs:
Analyze amino acid sequences surrounding modification sites
Identify motifs that favor single versus multiple modifications
Compare conservation of modification sites across species
Research shows that 2-hydroxyisobutyrylation sites have distinct sequence specificities with negative charged amino acids (D and E) strongly preferred around the modified sites
This integrated approach can reveal how 2-hydroxyisobutyrylation interacts with other epigenetic modifications to regulate gene expression.
The role of 2-hydroxyisobutyryl-HIST1H1C in diabetic retinopathy pathogenesis involves several interconnected mechanisms:
Upregulation in diabetic conditions:
Autophagy regulation pathway:
Inflammation and cell toxicity mechanisms:
Experimental evidence from animal models:
Therapeutic potential:
These findings highlight the central role of 2-hydroxyisobutyryl-HIST1H1C in the pathogenesis of diabetic retinopathy and suggest new avenues for therapeutic intervention.
Optimizing ChIP experiments with 2-hydroxyisobutyryl-HIST1H1C antibodies requires attention to several critical parameters:
Crosslinking conditions:
Use freshly prepared formaldehyde (1% final concentration) for 10-15 minutes at room temperature
Quench with glycine (125mM final concentration)
For histone modifications, shorter crosslinking times may be preferable to avoid epitope masking
Chromatin sonication parameters:
Sonicate to achieve DNA fragments between 200-500bp
Verify fragmentation efficiency by agarose gel electrophoresis
Optimize sonication time and amplitude specifically for your cell type
Antibody selection and validation:
Immunoprecipitation conditions:
Pre-clear chromatin with protein A/G beads before adding antibody
Incubate with antibody overnight at 4°C with gentle rotation
Wash stringently to reduce background (typically 3-5 washes with increasing stringency)
Quality control measures:
Quantify enrichment at known target loci by qPCR before proceeding to sequencing
Include both positive regions (known targets) and negative regions
Calculate percent input or fold enrichment over IgG control
Sequential ChIP considerations:
For studying co-occurrence with other modifications, optimize elution conditions
Ensure complete elution of primary antibody before second immunoprecipitation
Verify that epitopes remain intact after first elution
Following these optimized conditions will maximize the specificity and sensitivity of ChIP experiments using 2-hydroxyisobutyryl-HIST1H1C antibodies.
Quantitative assessment of 2-hydroxyisobutyrylation levels requires multiple complementary approaches:
Western blotting with densitometry:
Mass spectrometry-based quantification:
ChIP-qPCR and ChIP-seq analysis:
Perform ChIP using 2-hydroxyisobutyryl-HIST1H1C antibodies
Quantify enrichment at specific loci by qPCR
For genome-wide analysis, perform ChIP-seq
Calculate differential enrichment between conditions using specialized software
Immunofluorescence quantification:
ELISA-based approaches:
These complementary approaches provide robust quantitative assessment of changes in 2-hydroxyisobutyrylation levels across different experimental conditions.
Designing effective 2-hydroxyisobutyryl-HIST1H1C overexpression or knockdown experiments requires careful consideration of several factors:
Overexpression strategy:
Select appropriate expression vectors (e.g., pIRES-Neo vector containing Flag and HA tags)
Consider inducible expression systems to control timing and level of expression
Include proper tags for detection (HA, Flag) without interfering with function
Allow sufficient time post-transfection (typically 48h) before analysis
Knockdown approach selection:
Controls and validation:
Experimental design considerations:
Determine appropriate timepoints for analysis based on protein turnover rates
Include rescue experiments to confirm specificity of observed phenotypes
Assess both direct effects on HIST1H1C and downstream consequences (e.g., autophagy)
Consider cell type-specific effects when selecting experimental systems
Functional readouts:
These considerations will help ensure robust and interpretable results from 2-hydroxyisobutyryl-HIST1H1C overexpression or knockdown experiments.
Troubleshooting non-specific binding or high background requires systematic optimization:
Antibody validation and quality control:
Blocking optimization:
Test different blocking agents (BSA, milk, normal serum)
Increase blocking time (1-2 hours at room temperature or overnight at 4°C)
Add blocking agents to antibody dilution buffer
Consider adding 0.1-0.3% Triton X-100 for immunofluorescence applications
Washing procedure refinement:
Increase number and duration of washes
Use appropriate detergent concentration in wash buffers
Ensure complete buffer removal between washes
Consider more stringent wash buffers for ChIP applications
Sample preparation improvements:
Optimize fixation conditions (time, temperature, fixative concentration)
Ensure adequate permeabilization for immunofluorescence
For western blotting, optimize protein extraction methods to maintain modifications
For ChIP, optimize chromatin fragmentation and pre-clearing steps
Advanced solutions for persistent problems:
Pre-absorb antibody with acetone powder from relevant tissues
Perform negative control subtractions in imaging applications
Use alternative detection systems (e.g., switch from HRP to fluorescence)
Consider using monoclonal antibodies if available for increased specificity
Systematic application of these troubleshooting strategies can significantly reduce non-specific binding and background issues when using 2-hydroxyisobutyryl-HIST1H1C antibodies.
Common pitfalls in interpreting 2-hydroxyisobutyryl-HIST1H1C studies and strategies to avoid them include:
Cross-reactivity misinterpretation:
Causation vs. correlation confusion:
Cell type and context dependency oversights:
Overlooking modification crosstalk:
Technical biases in enrichment methods:
Neglecting dynamic regulation:
By recognizing and addressing these common pitfalls, researchers can ensure more robust and reliable interpretations of 2-hydroxyisobutyryl-HIST1H1C studies.
Emerging techniques for studying 2-hydroxyisobutyrylation dynamics in live cells represent cutting-edge approaches in epigenetic research:
Genetically encoded biosensors:
Development of FRET-based sensors specific for 2-hydroxyisobutyrylation
Creation of modified reader domain constructs that specifically recognize 2-hydroxyisobutyrylated histones
These tools would allow real-time visualization of modification dynamics
CRISPR-based epigenome editing:
Fusion of catalytic domains from 2-hydroxyisobutyrylation writers/erasers to dCas9
Site-specific manipulation of 2-hydroxyisobutyrylation at endogenous loci
Monitoring subsequent effects on gene expression and chromatin structure
Single-molecule tracking in live cells:
Tag 2-hydroxyisobutyrylation readers with photoactivatable fluorescent proteins
Monitor their dynamics and residence time on chromatin
Compare with other epigenetic modification readers to understand relative stability
Click chemistry approaches:
Metabolic labeling with chemical analogs of 2-hydroxyisobutyrate
Bioorthogonal chemistry for visualization and enrichment
Pulse-chase experiments to measure turnover rates
Integration with advanced microscopy:
Super-resolution microscopy to visualize 2-hydroxyisobutyrylation spatial distribution
Lattice light-sheet microscopy for long-term imaging with reduced phototoxicity
Correlative light and electron microscopy to link modification states with ultrastructural features
Microfluidics and single-cell analysis:
Microfluidic platforms for temporally controlled perturbations
Single-cell sequencing approaches to capture heterogeneity in modification patterns
Integration with single-cell proteomics for comprehensive epigenetic profiling
These emerging techniques promise to revolutionize our understanding of 2-hydroxyisobutyrylation dynamics in living systems, moving beyond static snapshots to capture the true temporal nature of this epigenetic modification.
Understanding 2-hydroxyisobutyryl-HIST1H1C function could translate into novel therapeutic approaches through several promising avenues:
Targeted inhibition strategies:
Development of small molecule inhibitors targeting enzymes that catalyze HIST1H1C 2-hydroxyisobutyrylation
Design of peptide-based inhibitors that compete with HIST1H1C for modification enzymes
Creation of degraders (PROTACs) specific for modified HIST1H1C
Gene therapy approaches:
Autophagy modulation:
Anti-inflammatory strategies:
Development of compounds that block the inflammatory pathways activated by HIST1H1C
Targeted delivery of anti-inflammatory agents to affected tissues
Combinatorial approaches addressing both autophagy and inflammation
Biomarker development:
Use of 2-hydroxyisobutyrylated HIST1H1C as a biomarker for disease progression
Development of less invasive diagnostic tools based on modification patterns
Stratification of patients for personalized therapeutic approaches
Metabolic interventions:
Since 2-hydroxyisobutyrylation requires 2-hydroxyisobutyrate derived from metabolism
Dietary or pharmacological approaches to modify precursor availability
Targeting metabolic pathways that generate 2-hydroxyisobutyrate
These therapeutic avenues could potentially address not only diabetic retinopathy but also other conditions where dysregulation of 2-hydroxyisobutyryl-HIST1H1C contributes to pathogenesis.