2-hydroxyisobutyryl-HIST1H1C (K109) Antibody

Shipped with Ice Packs
In Stock

Description

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.

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Description

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.

Form
Liquid
Lead Time
Typically, we can ship products within 1-3 working days after receiving your orders. Delivery timelines may vary depending on the purchasing method or location. Please consult your local distributors for precise delivery times.
Synonyms
H1 histone family member 2 antibody; H1.a antibody; H12_HUMAN antibody; H1F2 antibody; H1s-1 antibody; HIST1H1C antibody; Histone 1 H1c antibody; Histone cluster 1 H1c antibody; Histone H1.2 antibody; Histone H1c antibody; Histone H1d antibody; Histone H1s-1 antibody; MGC3992 antibody
Target Names
Uniprot No.

Target Background

Function

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.

Gene References Into Functions
  1. Research findings demonstrate a network of E2F target genes influenced by H1.2, where H1.2 enhances the global association of pRb with chromatin, amplifies transcriptional repression by pRb, and facilitates pRb-dependent cell-cycle arrest. PMID: 28614707
  2. BRG1 participates in gene repression by interacting with H1.2, facilitating its deposition and stabilizing nucleosome positioning around the transcription start site. PMID: 27390128
  3. Studies have revealed the presence of histones H1.2 and H1.4 in MDA-MB-231 metastatic breast cancer cells. Phosphorylation at S173 of histone H1.2 and S172, S187, T18, T146, and T154 of H1.4 significantly increases during M phase, suggesting a cell cycle-dependent nature of these events. Additionally, the study reports the observation of the H1.2 SNP variant A18V in MCF-10A cells. PMID: 26209608
  4. Integration with apoptotic intermediates (via C-terminal tail interactions) may represent a more generalized function of linker histone isoforms in apoptotic cascades. PMID: 24525734
  5. Histone H1.2-T165 post translational modifications are dispensable for chromatin binding and cell proliferation, while the H1.4-K26 modifications are essential for proper cell cycle progression. PMID: 24873882
  6. H1.2 interacts with Cul4A and PAF1 to activate developmental regulatory genes. PMID: 24360965
  7. H1.2 exhibits lower abundance compared to other histone H1 variants at the transcription start sites of inactive genes. Promoters enriched in H1.2 differ from those enriched in other histone H1 variants and tend to be repressed. PMID: 24476918
  8. Mutations in linker histone genes HIST1H1 B, C, D, and E; OCT2 (POU2F2); IRF8; and ARID1A underlie the pathogenesis of follicular lymphoma. PMID: 24435047
  9. Evidence suggests that the p53 acetylation-H1.2 phosphorylation cascade serves as a unique mechanism for triggering p53-dependent DNA damage response pathways. PMID: 22249259
  10. Studies have confirmed N-terminal acetylation on all isoforms along with a single internal acetylation site. Phosphorylation sites were identified on peptides containing the cyclin dependent kinase (CDK) consensus motif. PMID: 15595731
  11. The binding of histone H1 to a general amyloid-like motif suggests that histone H1 may play a common role in diseases associated with amyloid-like fibrils. PMID: 16854430
  12. Histone H1.2 was observed to translocate from the nucleus to the mitochondria following treatment with bleomycin and co-localize with Bak in mitochondria. PMID: 17879944
  13. Research indicates that the recruitment of YB1, PURalpha, and H1.2 to the p53 target gene Bax is necessary for repression of p53-induced transcription. PMID: 18258596

Show More

Hide All

Database Links

HGNC: 4716

OMIM: 142710

KEGG: hsa:3006

STRING: 9606.ENSP00000339566

UniGene: Hs.7644

Protein Families
Histone H1/H5 family
Subcellular Location
Nucleus. Chromosome. Note=Mainly localizes in euchromatin. Distribution goes in parallel with DNA concentration.

Q&A

What is 2-hydroxyisobutyryl-HIST1H1C and what is its significance in epigenetic research?

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 .

What applications are validated for 2-hydroxyisobutyryl-HIST1H1C antibodies?

The 2-hydroxyisobutyryl-HIST1H1C antibody has been validated for multiple applications in epigenetic and chromatin research:

  • Enzyme-Linked Immunosorbent Assay (ELISA)

  • Immunocytochemistry (ICC)

  • Immunofluorescence (IF)

  • Chromatin Immunoprecipitation (ChIP)

  • Western blotting for detecting expression levels and knockout validation

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.

How do I determine the specificity of a 2-hydroxyisobutyryl-HIST1H1C antibody?

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.

What is the difference between site-specific antibodies for different lysine residues in HIST1H1C?

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.

How can I design experiments to investigate the functional relationship between 2-hydroxyisobutyryl-HIST1H1C and autophagy in disease models?

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:

    • Use AAV-mediated HIST1H1C overexpression in animal models

    • Implement knockdown of HIST1H1C using siRNA in diabetic mouse retinas

    • Analyze autophagy markers, inflammation indicators, glial activation, and neuron loss

  • Cellular toxicity measurements:

    • Perform MTT assays (cells plated at 5000 cells/well, treated for 48h, followed by 4h MTT incubation) to measure viability changes

This comprehensive approach provides multiple lines of evidence for the functional relationship between 2-hydroxyisobutyryl-HIST1H1C and autophagy in disease contexts.

How do I differentiate between the effects of 2-hydroxyisobutyrylation and other acylation modifications on HIST1H1C function?

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:

    • Identify and manipulate the enzymes responsible for adding or removing each modification

    • Determine if inhibiting deacetylases affects 2-hydroxyisobutyrylation levels and vice versa

    • Analyze how modifying enzymes like SIRT1 and HDAC1 differently affect various acylation types

  • 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.

What are the consequences of histone H1 reduction on replicative stress, and how can I measure these effects experimentally?

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:

    • Decreases in replication fork rate

    • Increases in fork asymmetry

    • Transcription-dependent replicative stress

    • Enhanced non-coding RNA chromatin association

    • Elevated DNA damage signaling

  • DNA fiber analysis methodology:

    • Label nascent DNA strands with nucleotide analogs (e.g., CldU followed by IdU)

    • Stretch DNA fibers on microscope slides

    • Detect labeled fibers using specific antibodies

    • Measure fork rate by calculating the length of labeled tracks

    • Assess fork asymmetry by comparing the lengths of sister forks

  • 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:

    • Measure γH2AX foci formation as a marker of DNA double-strand breaks

    • Monitor activation of DNA damage response proteins (ATM, ATR, Chk1, Chk2)

    • Assess the phosphorylation status of these proteins by western blotting

    • Determine if DNA damage signaling is reduced by transcription inhibition

  • 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.

How can I investigate the cross-talk between 2-hydroxyisobutyrylation and other epigenetic modifications in regulating gene expression?

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.

What is the role of 2-hydroxyisobutyryl-HIST1H1C in disease pathogenesis, particularly in diabetic retinopathy?

The role of 2-hydroxyisobutyryl-HIST1H1C in diabetic retinopathy pathogenesis involves several interconnected mechanisms:

  • Upregulation in diabetic conditions:

    • Both histone HIST1H1C and ATG proteins are upregulated in the retinas of diabetic rodents

    • This upregulation correlates with disease progression and severity

  • Autophagy regulation pathway:

    • HIST1H1C overexpression upregulates SIRT1 and HDAC1

    • These deacetylases maintain the deacetylation status of H4K16

    • Deacetylated H4K16 leads to upregulation of ATG proteins

    • Increased ATG proteins promote autophagy in retinal cells

  • Inflammation and cell toxicity mechanisms:

    • HIST1H1C overexpression promotes inflammation via increased production of pro-inflammatory cytokines

    • Enhanced autophagy contributes to cell toxicity under diabetic conditions

    • Both processes contribute to retinal cell damage and death

  • Experimental evidence from animal models:

    • AAV-mediated HIST1H1C overexpression in rodent retinas leads to:

      • Increased autophagy

      • Inflammation

      • Glial activation

      • Neuron loss

    • These pathological changes mirror those identified in early diabetic retinopathy

  • Therapeutic potential:

    • Knockdown of HIST1H1C by siRNA in diabetic mouse retinas significantly attenuates:

      • Diabetes-induced autophagy

      • Inflammation

      • Glial activation

      • Neuron loss

    • This suggests HIST1H1C as a potential therapeutic target for preventing diabetic retinopathy

These findings highlight the central role of 2-hydroxyisobutyryl-HIST1H1C in the pathogenesis of diabetic retinopathy and suggest new avenues for therapeutic intervention.

What are the optimal conditions for using 2-hydroxyisobutyryl-HIST1H1C antibodies in ChIP experiments?

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:

    • Use antibodies specifically validated for ChIP applications

    • Perform preliminary tests with different antibody concentrations (2-5μg per reaction)

    • Include appropriate controls (IgG negative control, input DNA)

  • 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.

How can I quantitatively assess changes in 2-hydroxyisobutyrylation levels across different experimental conditions?

Quantitative assessment of 2-hydroxyisobutyrylation levels requires multiple complementary approaches:

  • Western blotting with densitometry:

    • Use validated antibodies against 2-hydroxyisobutyryl-HIST1H1C

    • Include loading controls (vinculin or total histone H3)

    • Perform densitometric analysis using software like ImageJ

    • Calculate relative levels normalized to total protein or specific loading controls

  • Mass spectrometry-based quantification:

    • Use stable isotope labeling approaches (SILAC, TMT, iTRAQ)

    • Enrich 2-hydroxyisobutyrylated peptides using antibody-based affinity enrichment

    • Analyze using nano-HPLC/MS/MS

    • Compare relative abundance of modified peptides across conditions

  • 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:

    • Use immunofluorescence staining with specific antibodies

    • Acquire images using consistent exposure settings

    • Measure fluorescence intensity using image analysis software

    • Compare signal intensity across different treatment conditions

  • ELISA-based approaches:

    • Develop or use commercial ELISA kits with 2-hydroxyisobutyryl-specific antibodies

    • Generate standard curves with synthetic peptides

    • Measure modification levels in protein extracts

    • Calculate absolute or relative quantification based on standards

These complementary approaches provide robust quantitative assessment of changes in 2-hydroxyisobutyrylation levels across different experimental conditions.

What are the key considerations when designing 2-hydroxyisobutyryl-HIST1H1C overexpression or knockdown experiments?

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

    • Verify expression by western blot and immunofluorescence

    • Allow sufficient time post-transfection (typically 48h) before analysis

  • Knockdown approach selection:

    • Choose between transient (siRNA) and stable (shRNA) knockdown systems

    • For stable knockdown, select appropriate selection markers (e.g., puromycin at 1μg/ml)

    • Design multiple target sequences to control for off-target effects

    • In vivo knockdown may require specialized delivery systems like AAV vectors

  • Controls and validation:

    • Include empty vector controls for overexpression

    • Use non-targeting siRNA/shRNA controls for knockdown

    • Verify knockdown/overexpression efficiency at both mRNA (qPCR) and protein levels

    • Consider using knockout cell lines as positive controls for antibody 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:

    • Select appropriate assays based on hypothesized function (e.g., autophagy assays, MTT for viability)

    • Include multiple independent measures of each phenotype

    • Consider genome-wide approaches (RNA-seq, ChIP-seq) to identify comprehensive effects

These considerations will help ensure robust and interpretable results from 2-hydroxyisobutyryl-HIST1H1C overexpression or knockdown experiments.

How do I troubleshoot non-specific binding or high background when using 2-hydroxyisobutyryl-HIST1H1C antibodies?

Troubleshooting non-specific binding or high background requires systematic optimization:

  • Antibody validation and quality control:

    • Verify antibody specificity using knockout controls

    • Titrate antibody concentration to determine optimal working dilution

    • Consider different antibody sources if persistent issues occur

    • Use freshly prepared antibody dilutions and avoid repeated freeze-thaw cycles

  • 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.

What are the common pitfalls in interpreting results from 2-hydroxyisobutyryl-HIST1H1C studies, and how can they be avoided?

Common pitfalls in interpreting 2-hydroxyisobutyryl-HIST1H1C studies and strategies to avoid them include:

  • Cross-reactivity misinterpretation:

    • Pitfall: Antibodies may recognize other histone modifications or proteins

    • Solution: Always validate with knockout controls and include peptide competition assays

    • Example: Research shows antibodies may detect additional bands beyond the expected HIST1H1C signal

  • Causation vs. correlation confusion:

    • Pitfall: Assuming changes in 2-hydroxyisobutyrylation directly cause observed phenotypes

    • Solution: Perform rescue experiments and use site-specific mutants

    • Example: In diabetic retinopathy studies, knockdown experiments were essential to establish causality

  • Cell type and context dependency oversights:

    • Pitfall: Generalizing findings from one cell type to others

    • Solution: Validate key findings in multiple cell types and in vivo when possible

    • Example: Different cell types may show varying dependencies on HIST1H1C for autophagy regulation

  • Overlooking modification crosstalk:

    • Pitfall: Studying 2-hydroxyisobutyrylation in isolation

    • Solution: Consider other modifications that might co-occur or compete for the same lysine residues

    • Example: Research shows overlap between 2-hydroxyisobutyrylation, acetylation, and succinylation sites

  • Technical biases in enrichment methods:

    • Pitfall: Antibody-based enrichment may favor certain sequence contexts

    • Solution: Complement with mass spectrometry and use multiple antibodies when possible

    • Example: Studies have shown that 2-hydroxyisobutyrylation sites have sequence preferences that might affect detection

  • Neglecting dynamic regulation:

    • Pitfall: Taking a static snapshot of modifications

    • Solution: Perform time-course experiments and consider enzyme regulation

    • Example: Studies on SIRT1 and HDAC1 show dynamic regulation of histone modifications

By recognizing and addressing these common pitfalls, researchers can ensure more robust and reliable interpretations of 2-hydroxyisobutyryl-HIST1H1C studies.

What are the emerging techniques for studying 2-hydroxyisobutyrylation dynamics in live cells?

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.

How might understanding 2-hydroxyisobutyryl-HIST1H1C function lead to novel therapeutic approaches for diseases like diabetic retinopathy?

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:

    • AAV-delivered siRNA targeting HIST1H1C in affected tissues

    • Research shows knockdown of HIST1H1C by siRNA in diabetic mouse retinas attenuated pathological changes

    • CRISPR-based approaches to modify specific lysine residues to prevent 2-hydroxyisobutyrylation

  • Autophagy modulation:

    • Since HIST1H1C regulates autophagy in diabetic retinopathy , therapies could target:

    • Downstream components of the autophagy pathway (ATG proteins)

    • The SIRT1-HDAC1-H4K16 axis that mediates HIST1H1C effects on autophagy

    • Selective autophagy inducers or inhibitors based on disease stage

  • 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.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.