The Glutaryl-HIST1H2BC (K116) Antibody is a polyclonal antibody developed in rabbits, with the following key attributes:
This antibody is purified via antigen-affinity chromatography and lacks conjugation, ensuring specificity for glutaryl-modified HIST1H2BC . Its validation includes Western blot detection of a 14 kDa band in sodium butyrate-treated 293 and HeLa cell lysates, confirming target specificity .
HIST1H2BC Function: A replication-dependent histone H2B subtype encoded by the HIST1H2BC gene (UniProt: P62807). It assembles nucleosomes, compacting DNA into chromatin and regulating transcription, DNA repair, and replication .
Glutarylation at K116: A lysine modification involving the addition of a glutaryl group. This PTM may influence chromatin structure, gene expression, or DNA damage response pathways, though its exact role remains under investigation .
The antibody enables precise detection of glutaryl-HIST1H2BC (K116) in:
Western Blot: Identifies the 14 kDa HIST1H2BC protein in treated cell lysates (e.g., sodium butyrate-induced) .
Immunofluorescence: Localizes glutaryl-HIST1H2BC to nuclei in fixed/permeabilized HeLa cells .
Epigenetic Studies: Facilitates exploration of glutarylation’s role in chromatin remodeling and disease mechanisms (e.g., cancer, metabolic disorders) .
Western Blot: Clear 14 kDa band observed in sodium butyrate-treated 293 cells, absent in unmodified controls .
Immunofluorescence: Nuclear staining in HeLa cells, colocalizing with DAPI, confirms histone-specific targeting .
Specificity: No cross-reactivity with non-glutarylated HIST1H2BC or other histone variants reported .
Glutarylation is an emerging PTM with potential roles in metabolic regulation and chromatin accessibility. This antibody provides a critical tool for:
Mapping glutarylation sites in disease models (e.g., cancer, neurodegeneration).
Investigating crosstalk between metabolic pathways and epigenetic regulation .
Glutaryl-HIST1H2BC (K116) refers to a specific post-translational modification (PTM) where a glutaryl group is attached to the lysine 116 residue of Histone H2B type 1-C/E/F/G/I protein. This modification is significant in epigenetic research because histone proteins play critical roles in packaging genomic DNA into nucleosomes and regulating gene expression . The nucleosome structure contains two subunits, each consisting of histones H2A, H2B, H3, and H4, with histone H1 serving as a junctional histone .
Glutarylation, like other histone PTMs (methylation, acetylation, phosphorylation, ubiquitination), serves as a signal for chromatin opening/compression and recruits factors that promote or antagonize transcription . Specifically, glutarylation at K116 of HIST1H2BC represents one of the newly discovered modifications that contributes to the "histone code," influencing gene regulation and potentially playing roles in various cellular processes and disease states.
The methodological approach to studying this modification typically begins with specific antibodies that recognize only the glutarylated form of this residue, allowing researchers to investigate its presence, distribution, and function in different cellular contexts .
Glutaryl-HIST1H2BC (K116) Antibody is specifically designed to recognize and bind to histone H2B type 1-C/E/F/G/I that has been modified with a glutaryl group at the lysine 116 position . This specificity distinguishes it from antibodies targeting other histone modifications in several key ways:
Epitope specificity: This antibody recognizes only the glutaryl modification at the K116 position, not other positions or modifications like acetylation or methylation .
PTM chemistry: Glutarylation involves the addition of a five-carbon dicarboxylic acid derivative, which creates a structurally distinct modification compared to smaller modifications like acetylation (two-carbon) or larger ones like ubiquitination .
Research applications: While many histone modification antibodies are validated for multiple applications, the Glutaryl-HIST1H2BC (K116) Antibody has been specifically validated for ELISA, Western blot, and immunofluorescence techniques .
Methodologically, when selecting between different modification-specific antibodies, researchers should consider the following approach:
Verify the exact epitope recognition (position and modification type)
Check cross-reactivity data with similar modifications
Review validation data for your specific application
Consider using complementary antibodies to different modifications in parallel experiments to build a comprehensive picture of the histone modification landscape .
For optimal Western blot results with Glutaryl-HIST1H2BC (K116) Antibody, the following methodological approach is recommended:
Sample Preparation:
Extract histones using acid extraction methods (0.2N HCl or 0.4N H₂SO₄) to preserve post-translational modifications
Include deacetylase and protease inhibitors in extraction buffers
Load 10-20 μg of histone extracts or 50-100 μg of whole cell lysates per lane
Western Blot Protocol:
Dilution range: Use antibody at 1:100-1:1000 dilution for Western blot applications
Blocking: 5% non-fat dry milk or BSA in TBST for 1 hour at room temperature
Primary antibody incubation: Overnight at 4°C in blocking buffer
Washing: 3-5 times with TBST, 5 minutes each
Secondary antibody: Anti-rabbit IgG (the antibody is raised in rabbits)
Detection system: ECL or similar chemiluminescence system
Controls:
Positive control: Human cell lines with known glutarylation (HeLa, HEK293)
Negative control: Samples treated with glutarylation-specific eraser enzymes
Loading control: Total H2B or other core histones
When troubleshooting, consider that the high specificity of this antibody means that signal intensity will directly correlate with the abundance of glutarylation at the specific K116 site, which may vary significantly across different cell types and physiological conditions .
Glutarylation is metabolically linked to the TCA cycle and lysine metabolism, making Glutaryl-HIST1H2BC (K116) Antibody a valuable tool for investigating metabolism-epigenetics connections. A comprehensive methodological approach includes:
Experimental Design:
Metabolic perturbation experiments:
Treat cells with glutaryl-CoA precursors or inhibitors
Modify cellular energy status (glucose deprivation, fatty acid supplementation)
Induce mitochondrial dysfunction using specific inhibitors
Multi-omics integration:
Combine ChIP-seq using Glutaryl-HIST1H2BC (K116) Antibody with:
Metabolomics to measure TCA cycle intermediates
RNA-seq to assess transcriptional consequences
Proteomics to identify associated protein complexes
Enzyme manipulation studies:
Overexpress or knockdown glutaryl-lysine transferases
Modulate deglutarylase enzymes (typically sirtuins)
Assess changes in glutarylation patterns using the antibody
Data Analysis Framework:
Track changes in glutarylation levels under different metabolic conditions
Correlate glutarylation with gene expression changes
Map glutarylation sites to chromatin regions with specific functions
This methodological approach allows researchers to establish direct connections between cellular metabolism, histone glutarylation at K116, and transcriptional outcomes, potentially revealing new regulatory mechanisms in metabolic diseases .
While standard ChIP protocols provide a foundation, optimizing ChIP with Glutaryl-HIST1H2BC (K116) Antibody requires specific methodological considerations:
Pre-ChIP Considerations:
Cell number: Start with 1-5×10⁶ cells for targeted ChIP; 10-20×10⁶ for ChIP-seq
Crosslinking: 1% formaldehyde for 10 minutes at room temperature
Include glutarylation preservatives: Add 5-10 mM sodium butyrate and KDAC inhibitors
ChIP Protocol Optimization:
Chromatin shearing:
Target fragments of 200-500 bp for optimal resolution
Sonication parameters: 10-15 cycles (30s ON/30s OFF) at medium power
Verify fragmentation by agarose gel electrophoresis
Immunoprecipitation:
Antibody amount: 2-5 μg per ChIP reaction
Pre-clearing: 1-2 hours with protein A/G beads
Incubation time: Overnight at 4°C with gentle rotation
Washing stringency: Gradually increase salt concentration (150-500 mM NaCl)
Controls:
Input control: 5-10% of chromatin before immunoprecipitation
IgG control: Same amount of rabbit IgG as the primary antibody
Positive control: Antibody against total H2B or H3
Glutarylation-depleted control: Samples treated with deglutarylases
Analysis Recommendations:
For ChIP-qPCR: Design primers for regions with known or suspected glutarylation involvement
For ChIP-seq: Use specialized peak calling algorithms sensitive to histone modification patterns
Compare glutarylation patterns with other histone marks to identify co-occurrence or mutual exclusion
This optimized approach enhances sensitivity and specificity when mapping glutarylation at K116 across the genome, providing insight into its regulatory functions .
Distinguishing between glutarylation at different lysine residues requires a methodological approach combining antibody-based detection with advanced analytical techniques:
Antibody Specificity Verification:
Peptide competition assay:
Pre-incubate the Glutaryl-HIST1H2BC (K116) Antibody with:
Glutarylated K116 peptide (should eliminate signal)
Glutarylated non-K116 peptides (should not affect signal)
Unmodified K116 peptide (should not affect signal)
Proceed with Western blot or immunofluorescence
Loss of signal only with the glutarylated K116 peptide confirms specificity
Cross-reactivity testing:
Orthogonal Validation Techniques:
Mass spectrometry approach:
Digest histones with trypsin or alternative proteases
Enrich for glutarylated peptides using anti-glutaryl-lysine antibodies
Perform LC-MS/MS analysis with high mass accuracy
Identify specific glutarylation sites based on mass shifts
Quantify relative abundance of modifications at different sites
Site-directed mutagenesis:
Generate K116R, K120R, and other lysine-to-arginine mutants
Express in cells and immunoblot with site-specific antibodies
Signal loss with the specific mutation confirms antibody specificity
Data Integration Framework:
Create a glutarylation site map across the HIST1H2BC protein
Compare relative abundances of glutarylation at different sites
Correlate site-specific glutarylation with functional outcomes
This comprehensive approach ensures accurate identification and functional characterization of glutarylation specifically at the K116 position versus other sites .
Researchers working with Glutaryl-HIST1H2BC (K116) Antibody commonly encounter several challenges that require specific methodological solutions:
Causes:
Low abundance of glutarylation at K116
Glutarylation loss during sample preparation
Suboptimal antibody concentration
Methodological Solutions:
Treat cells with glutarylation enhancers (glutarate or glutaryl-CoA precursors)
Add deglutarylase inhibitors during all steps of sample preparation
Optimize antibody concentration (test range from 1:100 to 1:1000 for WB)
Increase protein loading (up to 50-100 μg for whole cell lysates)
Extend primary antibody incubation time (overnight at 4°C)
Use high-sensitivity detection systems (enhanced chemiluminescence)
Causes:
Insufficient blocking
Cross-reactivity with other glutarylated proteins
Secondary antibody issues
Methodological Solutions:
Optimize blocking (test 3-5% BSA vs. non-fat milk)
Increase washing duration and number of washes
Use highly purified antibody preparations
Perform peptide competition controls to verify specificity
Reduce secondary antibody concentration
Include 0.1-0.3% Triton X-100 in wash buffers
Causes:
Variability in glutarylation levels under different conditions
Technical variations in sample preparation
Lot-to-lot antibody variations
Methodological Solutions:
Standardize cell culture conditions (passage number, confluence)
Create standard operating procedures for histone extraction
Include internal controls in each experiment
Test and validate each new antibody lot
Generate standard curves using known glutarylated samples
Causes:
Fixation effects on epitope accessibility
Buffer incompatibility
Antibody format limitations
Methodological Solutions:
For IF: Test different fixation methods (4% PFA vs. methanol)
For ChIP: Optimize crosslinking conditions
For flow cytometry: Test permeabilization protocols
Consider using secondary antibody conjugates optimized for specific applications
This systematic troubleshooting approach helps researchers overcome technical challenges to obtain reliable and reproducible results when studying glutarylation at K116 of HIST1H2BC .
Multiplexed detection of histone modifications provides insight into the complex interplay between glutarylation and other epigenetic marks. A comprehensive methodological approach includes:
Sequential Immunoblotting Strategy:
Strip-and-reprobe technique:
Perform initial Western blot with Glutaryl-HIST1H2BC (K116) Antibody
Document results thoroughly
Strip membrane (mild stripping buffer: 200mM glycine, 0.1% SDS, 1% Tween-20, pH 2.2)
Verify complete stripping with secondary antibody only
Reprobe with antibodies against other modifications
Typical order: start with lowest abundance modification and proceed to higher abundance
Parallel blotting approach:
Run identical samples on multiple gels
Transfer and probe each membrane with different modification-specific antibodies
Align images based on molecular weight markers and loading controls
Multiplex Immunofluorescence Methods:
Sequential immunostaining:
Advanced multiplexing techniques:
Tyramide signal amplification for sequential staining with same-species antibodies
Spectral unmixing for closely overlapping fluorophores
Antibody stripping and restaining protocols
Mass Spectrometry-Based Approach:
Bottom-up histone analysis:
Digest histones with appropriate proteases
Analyze by LC-MS/MS with data-dependent acquisition
Identify peptides carrying multiple modifications
Quantify relative abundances of different modification combinations
Middle-down approach:
Use GluC or AspN to generate larger histone fragments
Analyze by high-resolution MS
Map combinatorial modification patterns
Data Integration and Visualization:
Generate modification co-occurrence matrices
Apply machine learning algorithms to identify modification patterns
Create visual maps of histone modification networks
This comprehensive multiplexing approach allows researchers to determine whether glutarylation at K116 co-occurs with, or is mutually exclusive of, other histone modifications, providing insight into the functional histone code .
The Glutaryl-HIST1H2BC (K116) Antibody is increasingly employed in cutting-edge disease research applications that leverage methodological innovations:
Cancer Research Applications:
Patient-derived xenograft (PDX) models:
Use the antibody to track glutarylation changes during tumor progression
Methodological approach:
Establish PDX models from patient biopsies
Monitor glutarylation at different stages using immunohistochemistry
Correlate with disease progression and treatment response
Create predictive biomarker panels combining glutarylation with other markers
Drug discovery pipelines:
Screen compounds affecting glutarylation enzymes
Methodological approach:
High-throughput screening with automated Western blots
Quantify glutarylation changes using image analysis software
Establish dose-response relationships
Validate hits in cellular and animal models
Neurodegenerative Disease Research:
Brain region-specific epigenetic mapping:
Map glutarylation patterns across brain regions
Methodological approach:
Immunohistochemistry on brain sections
Compare glutarylation in affected vs. unaffected regions
Correlate with neuronal function and pathology
Integrate with other epigenetic marks in three-dimensional chromatin maps
Single-cell epigenomics:
Analyze glutarylation heterogeneity in neuronal populations
Methodological approach:
Single-cell Western blot or CyTOF with the antibody
Identify cell type-specific glutarylation signatures
Correlate with disease susceptibility
Metabolic Disease Innovations:
Circadian rhythm studies:
Track glutarylation changes throughout the day
Methodological approach:
Time-course sampling and Western blot analysis
ChIP-seq at different time points
Correlate with metabolic fluctuations and gene expression
Nutrigenomic applications:
Investigate dietary influences on histone glutarylation
Methodological approach:
Dietary interventions followed by histone analysis
Compare different dietary components' effects on glutarylation
Link to metabolic outcomes and gene expression changes
Emerging Technologies:
Spatial transcriptomics integration:
Combine glutarylation detection with spatial gene expression
Methodological approach:
Perform immunofluorescence with the antibody
Follow with in situ RNA hybridization
Create spatial maps correlating glutarylation with gene expression
CRISPR epigenome editing:
Target glutarylation machinery to specific genomic loci
Methodological approach:
Design dCas9-glutaryltransferase fusions
Verify targeted glutarylation using ChIP with the antibody
Assess functional consequences of site-specific glutarylation
These advanced applications demonstrate how Glutaryl-HIST1H2BC (K116) Antibody enables researchers to explore the role of histone glutarylation in disease pathogenesis and potential therapeutic interventions .
For optimal immunofluorescence results with Glutaryl-HIST1H2BC (K116) Antibody, researchers should follow this detailed methodological protocol:
Sample Preparation:
Cell culture preparation:
Grow cells on coverslips or chamber slides to 70-80% confluence
Consider synchronizing cells if studying cell-cycle dependent changes
Fixation options:
Primary method: 4% paraformaldehyde for 15 minutes at room temperature
Alternative method: Ice-cold methanol for 10 minutes at -20°C
Note: Test both methods as epitope accessibility may vary
Permeabilization:
Use 0.1-0.2% Triton X-100 in PBS for 10 minutes
For nuclear-specific staining, ensure complete permeabilization
Immunostaining Protocol:
Blocking:
Block with 3-5% BSA or normal serum in PBS for 1 hour at room temperature
Include 0.1% Tween-20 to reduce background
Primary antibody incubation:
Washing:
Wash 3 times with PBS containing 0.05% Tween-20, 5 minutes each
Ensure thorough washing to minimize background
Secondary antibody:
Apply anti-rabbit secondary antibody (fluorophore-conjugated)
Incubate for 1-2 hours at room temperature
Protect from light to prevent photobleaching
Counterstaining:
Nuclear counterstain: DAPI (1 μg/ml) for 5 minutes
Optional: Add phalloidin for F-actin visualization
Mounting:
Mount with anti-fade mounting medium
Seal edges with nail polish for long-term storage
Controls and Validation:
Positive control: Human cell lines with known glutarylation
Negative controls:
Primary antibody omission
Peptide competition (pre-incubate antibody with immunizing peptide)
Specificity control: Compare with other glutarylation site antibodies
Image Acquisition and Analysis:
Microscopy settings:
Capture images with confocal or wide-field fluorescence microscopy
Use consistent exposure settings across samples
Z-stack acquisition for 3D analysis of nuclear distribution
Quantification methods:
Measure nuclear intensity using ImageJ or similar software
Perform co-localization analysis with other histone marks
Consider single-cell analysis to detect population heterogeneity
This comprehensive protocol ensures reliable detection and quantification of HIST1H2BC glutarylation at K116 in cellular contexts, allowing for spatial analysis of this epigenetic mark .
Quantitative analysis of Glutaryl-HIST1H2BC (K116) Western blot data requires a rigorous methodological approach to ensure accuracy and reproducibility:
Experimental Design for Quantification:
Sample preparation controls:
Include biological replicates (minimum n=3)
Prepare all samples simultaneously with identical protocols
Include a concentration gradient of positive control samples
Ensure equal protein loading across all wells
Loading and normalization strategy:
Always run total H2B on the same blot or on a parallel blot
Include loading controls (β-actin, GAPDH, or total protein stain)
Consider dual-channel Western blot systems for simultaneous detection
Image Acquisition Protocol:
Optimal image capture:
Use a digital imaging system with linear dynamic range
Avoid oversaturation (check histogram during acquisition)
Capture multiple exposures if signal intensity varies greatly
Include molecular weight markers in all images
File handling:
Save raw, unmodified files in original format
Use lossless compression formats for analysis (TIFF preferred)
Maintain consistent image resolution across experiments
Quantification Methods:
Densitometry analysis workflow:
Define regions of interest (ROIs) of consistent size
Subtract local background for each lane
Measure integrated density or mean gray value
Normalize glutarylation signal to total H2B or loading control
Calculate relative changes compared to control conditions
Advanced quantification approaches:
Time-course analysis: Plot glutarylation changes over time
Dose-response relationship: Plot glutarylation vs. treatment concentration
Signal ratio analysis: Compare glutarylation at different sites
Statistical Analysis Framework:
Basic statistical tests:
Paired or unpaired t-tests for two-group comparisons
ANOVA with appropriate post-hoc tests for multiple groups
Non-parametric tests if normality assumptions are violated
Data presentation guidelines:
Include representative blot images with molecular weight markers
Present quantification data as bar charts with error bars (SEM or SD)
Indicate statistical significance levels (* p<0.05, ** p<0.01, etc.)
Include sample size (n) in figure legends
Example Quantification Table Format:
Treatment Condition | Raw Glutaryl-K116 Signal | Total H2B Signal | Normalized Ratio | Fold Change vs. Control | Statistical Significance |
---|---|---|---|---|---|
Control | 10,245 ± 856 | 25,678 ± 1,243 | 0.399 ± 0.032 | 1.00 | - |
Treatment A | 15,678 ± 1,243 | 24,987 ± 1,532 | 0.627 ± 0.051 | 1.57 | p<0.01 |
Treatment B | 8,756 ± 754 | 26,123 ± 1,347 | 0.335 ± 0.029 | 0.84 | p<0.05 |
This systematic approach to quantitative Western blot analysis ensures reliable measurement of Glutaryl-HIST1H2BC (K116) levels, enabling meaningful comparison across experimental conditions and accurate interpretation of biological significance .
Integrating epigenomic data from Glutaryl-HIST1H2BC (K116) ChIP-seq with other omics datasets requires a sophisticated methodological approach:
Data Generation and Quality Control:
Multi-omics experimental design:
Use matched samples for all omics analyses
Include appropriate replicates (minimum triplicate)
Process all samples in parallel when possible
Incorporate spike-in controls for quantitative comparisons
ChIP-seq specific considerations:
Generate input controls from the same chromatin
Include IgG controls to assess non-specific binding
Consider using spike-in normalization (e.g., Drosophila chromatin)
Sequence to sufficient depth (>20 million uniquely mapped reads)
Computational Analysis Pipeline:
Primary data processing:
ChIP-seq: Alignment, peak calling, signal normalization
RNA-seq: Alignment, quantification, differential expression analysis
ATAC-seq/DNase-seq: Accessibility profiling
Proteomics: Protein identification and quantification
Integration analysis methods:
Correlation analysis:
Calculate correlation between glutarylation signal and gene expression
Generate heatmaps of multi-omics data centered on glutarylation peaks
Perform k-means clustering to identify co-regulated gene sets
Feature overlapping:
Define genomic intervals (promoters, enhancers, gene bodies)
Calculate glutarylation enrichment in these regions
Compare with other histone marks and chromatin features
Advanced statistical integration:
Factor analysis for quantification of latent variables
Bayesian network modeling for causal relationship inference
Machine learning for pattern recognition and prediction
Visualization Strategies:
Genome browser tracks:
Display aligned ChIP-seq data alongside RNA-seq, ATAC-seq
Create custom tracks for different experimental conditions
Generate aggregate plots around genomic features
Integrated visualization tools:
Circos plots for genome-wide interactions
Network diagrams showing relationships between multiple data types
Principal component analysis plots for global data structure
Functional Interpretation Framework:
Pathway enrichment analysis:
Identify biological processes enriched in glutarylated regions
Compare with pathways from differentially expressed genes
Integrate with metabolic pathway analysis if metabolomics data available
Motif analysis:
Identify transcription factor binding motifs enriched near glutarylation sites
Correlate with transcription factor expression data
Test for co-occurrence with other histone modifications
Systems biology approach:
Construct gene regulatory networks incorporating glutarylation data
Identify key nodes and regulatory hubs
Simulate perturbations to predict functional outcomes
Example Multi-omics Integration Workflow:
Perform ChIP-seq with Glutaryl-HIST1H2BC (K116) Antibody
Conduct RNA-seq on matched samples
Optional: Include ATAC-seq, other histone mark ChIP-seq, metabolomics
Process each dataset through appropriate pipelines
Identify glutarylation-enriched regions and associated genes
Correlate glutarylation patterns with gene expression changes
Perform pathway analysis on correlated gene sets
Validate key findings with targeted experiments
This comprehensive integration approach enables researchers to situate glutarylation within the broader epigenetic landscape and understand its functional consequences on gene expression and cellular phenotypes .
Glutaryl-HIST1H2BC (K116) Antibody is poised to play a critical role in several emerging research directions that promise to expand our understanding of epigenetic regulation:
Single-Cell Epigenomics:
Methodological approach:
Adapt ChIP protocols for single-cell applications using Glutaryl-HIST1H2BC (K116) Antibody
Combine with single-cell RNA-seq for correlated epigenetic-transcriptomic profiles
Develop computational frameworks to analyze cell-to-cell variation in glutarylation
Potential to reveal heterogeneity in epigenetic regulation previously masked in bulk analyses
Dynamic Regulation of Glutarylation:
Methodological approach:
Develop real-time imaging systems using fluorescent antibody derivatives
Create glutarylation biosensors for live-cell monitoring
Employ rapid induction systems to track glutarylation kinetics
Apply mathematical modeling to understand dynamic glutarylation regulation
Cross-talk with Other Acylations:
Methodological approach:
Perform multiplexed detection of different acylations (acetylation, butyrylation, glutarylation)
Map modification co-occurrence and exclusivity patterns
Identify shared and specific regulatory enzymes
Develop targeted approaches to manipulate specific acylations independently
Therapeutic Targeting of Glutarylation Pathways:
Methodological approach:
Screen for small molecules affecting glutarylation levels
Develop glutarylation-specific reader, writer and eraser modulators
Use Glutaryl-HIST1H2BC (K116) Antibody to validate target engagement
Apply in disease models to assess efficacy of epigenetic therapies
Evolutionary Conservation of Glutarylation:
Methodological approach:
Test antibody cross-reactivity with model organisms
Compare glutarylation patterns across species
Identify conserved regulatory mechanisms and functional roles
Explore specialized functions in different tissues and organisms
Environmental Influences on Glutarylation:
Methodological approach:
Expose cells/organisms to environmental stressors
Quantify glutarylation changes using the antibody
Correlate with metabolic alterations and transcriptional responses
Investigate transgenerational inheritance of glutarylation patterns
Integration with Structural Biology:
Methodological approach:
Use antibodies to purify glutarylated histones for structural studies
Apply cryo-EM to visualize glutarylated nucleosomes
Perform molecular dynamics simulations to predict structural effects
Develop structure-based approaches to targeting glutarylation
These emerging research directions highlight the potential of Glutaryl-HIST1H2BC (K116) Antibody to contribute to fundamental advances in our understanding of epigenetic regulation and its implications for human health and disease .
The current consensus regarding HIST1H2BC glutarylation is still evolving, but several key insights have emerged from recent research:
Fundamental Biological Roles:
Transcriptional regulation:
Glutarylation at K116 appears to correlate with transcriptional activation in many contexts
The modification likely alters chromatin compaction by neutralizing the positive charge of lysine
Evidence suggests glutarylation may function in recruiting specific transcriptional machinery
Metabolic sensing:
HIST1H2BC glutarylation serves as a link between cellular metabolism and gene expression
Levels fluctuate in response to changes in TCA cycle intermediates and glutaryl-CoA availability
May represent a mechanism for adapting transcriptional programs to metabolic states
Cell cycle regulation:
Emerging evidence suggests dynamic changes in glutarylation during cell cycle progression
May play roles in regulating replication timing and chromosomal segregation
Often works in concert with other histone modifications in a coordinated manner
Disease Associations:
Cancer biology:
Altered glutarylation patterns have been observed in several cancer types
May contribute to oncogenic transcriptional programs and metabolic rewiring
Potential biomarker value for certain cancer subtypes or stages
Metabolic disorders:
Dysregulated glutarylation in conditions with altered energy metabolism
Potential involvement in insulin resistance and diabetes pathophysiology
Links to mitochondrial dysfunction in metabolic syndrome
Neurodegenerative diseases:
Preliminary evidence of altered histone glutarylation in models of neurodegeneration
May influence neuronal gene expression patterns and stress responses
Potential therapeutic target for maintaining neural homeostasis
Regulatory Mechanisms:
Enzymatic control:
"Writers": Several acyltransferases can catalyze glutarylation, though specificity remains unclear
"Erasers": Sirtuin family members (particularly SIRT5) remove glutaryl groups
"Readers": Proteins that specifically recognize glutarylated histones are being identified
Cross-regulation with other modifications:
Glutarylation may compete with acetylation, methylation at shared lysine residues
Evidence for sequential modification patterns during cellular responses
Coordination with non-histone protein glutarylation in signaling networks
Current Research Gaps:
Limited temporal and spatial resolution of glutarylation dynamics in living cells
Incomplete understanding of site-specific functions (K116 vs. other sites)
Need for more disease-specific studies to establish causal relationships
Technical challenges in distinguishing closely related acyl modifications