Acetyl-HIST1H1B (K48) Antibody

Shipped with Ice Packs
In Stock

Description

Applications in Research

The antibody is employed in diverse experimental workflows:

Epigenetic Studies

  • Immunocytochemistry (ICC): Detects acetylated K48 in fixed cells (e.g., HeLa cells treated with sodium butyrate, an HDAC inhibitor) .

  • Chromatin Immunoprecipitation (ChIP): Identifies genomic regions associated with acetylated HIST1H1B, enabling analysis of chromatin accessibility and transcriptional activity .

  • ELISA: Quantifies acetylation levels in lysates or purified chromatin fractions .

Proteomic and Functional Studies

  • Exercise-Induced Acetylation: In a study on high-intensity interval training (HIIT), the antibody was used to identify acetylated HIST1H1B (K48) as part of a broader acetylome remodeling response. This modification correlated with enhanced mitochondrial function and metabolic adaptation .

Role in Chromatin Structure

HIST1H1B binds linker DNA between nucleosomes, stabilizing chromatin compaction. Acetylation at K48 disrupts histone-DNA interactions, promoting chromatin decondensation and transcriptional activation .

Exercise-Driven Acetylation

A proteomic study using this antibody revealed:

ParameterPre-HIITPost-HIITSignificance
Acetylation Sites642Increased mitochondrial and cytoskeletal acetylation
HIST1H1B (K48) AcetylationDetectedEnhancedLinked to improved metabolic plasticity

Source: eLife

This data highlights the antibody’s utility in mapping dynamic acetylation changes in response to physiological stressors.

Comparative Analysis of Acetylation Sites

While the K48-specific antibody targets a distinct site, related antibodies (e.g., K16 acetylation) are also used to study histone H1 modifications. Key differences include:

SiteAntibodyApplicationsRelevance
K16Acetyl-HIST1H1B (K16) Cancer, development studiesImplicated in gene silencing and chromatin relaxation
K48Acetyl-HIST1H1B (K48) Exercise physiology, metabolismAssociated with chromatin remodeling and mitochondrial adaptation

Potential Therapeutic and Diagnostic Implications

  • Disease Models: Aberrant histone acetylation is linked to cancer, autoimmune disorders, and metabolic diseases. The antibody could aid in identifying biomarkers for therapeutic intervention .

  • Mechanistic Insights: By tracking HIST1H1B acetylation, researchers can elucidate how chromatin modifiers (e.g., histone acetyltransferases) regulate cellular responses to stress .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchasing method and location. Please consult your local distributors for specific delivery timeframes.
Synonyms
Histone H1.5 (Histone H1a) (Histone H1b) (Histone H1s-3), HIST1H1B, H1F5
Target Names
HIST1H1B
Uniprot No.

Target Background

Function
Histone H1 protein interacts with the linker DNA segments between nucleosomes, forming the chromatin fiber, a critical macromolecular structure. Histones H1 play a crucial role in the condensation of nucleosome chains into higher-order structural fibers. Additionally, they act as regulators of individual gene transcription through chromatin remodeling, nucleosome spacing, and DNA methylation.
Gene References Into Functions
  1. Research indicates that normal ovarian tissues exhibit strong expression of histone H1.5, whereas ovarian granulosa cell tumors (GCTs) express this protein at low levels. Conversely, PLZF protein expression does not significantly differ between these two groups. PMID: 25023763
  2. Statistically significant variations in staining patterns have been observed for histone H1.5, enabling differentiation between leiomyosarcomas and leiomyomas. PMID: 24784718
  3. Promyelocytic leukemia zinc finger (PLZF) and histone H1.5 demonstrate distinct staining patterns in low- and high-grade pulmonary neuroendocrine tumors. PMID: 23416030
  4. FoxP3 interacts with H1.5, altering its binding to target genes, thereby modulating their expression and influencing Treg cell function. PMID: 21654845
  5. The mode of chromatin fiber compaction varies depending on the salt environment and the presence of linker histone H1. PMID: 16185066
  6. Phosphorylation of human H1 variants occurs non-randomly during both interphase and mitosis. Distinct serine- or threonine-specific kinases are involved in different cell cycle phases. PMID: 16377619
  7. Phosphorylation at threonine 10 is observed in prometaphase and disappears in telophase. This hyperphosphorylated form of H1.5 is primarily chromatin-bound in metaphase when chromatin condensation reaches its maximum. PMID: 19136008

Show More

Hide All

Database Links

HGNC: 4719

OMIM: 142711

KEGG: hsa:3009

STRING: 9606.ENSP00000330074

UniGene: Hs.131956

Protein Families
Histone H1/H5 family
Subcellular Location
Nucleus. Chromosome. Note=According to PubMed:15911621 more commonly found in heterochromatin. According to PubMed:10997781 associates with actively transcribed chromatin and not heterochromatin.
Tissue Specificity
Ubiquitous. Expressed in the majority of the cell lines tested and in testis.

Q&A

What is HIST1H1B and what cellular functions does it perform?

HIST1H1B, also known as Histone H1.5 (alternatively named Histone H1a, Histone H1b, or Histone H1s-3), is a linker histone that binds to DNA between nucleosomes to form the chromatin fiber structure. This protein performs several critical functions:

These functions are essential for proper gene expression control and DNA compaction within the nucleus. As a linker histone, HIST1H1B establishes higher-order chromatin structure that affects all DNA-related processes including transcription, replication, and repair.

What is the significance of K48 acetylation in HIST1H1B function?

Acetylation at lysine 48 (K48) represents a specific post-translational modification of HIST1H1B that affects its interaction with DNA and other nuclear proteins. Research demonstrates that:

This specific modification provides a mechanism for dynamic control of chromatin structure in response to cellular signals and environmental changes.

How does HIST1H1B acetylation differ from other histone modifications?

While histones undergo numerous post-translational modifications, HIST1H1B acetylation has distinct characteristics:

  • Unlike core histone acetylation which often occurs at multiple residues, HIST1H1B acetylation appears more targeted

  • Acetylation of linker histones like HIST1H1B primarily affects higher-order chromatin structure rather than the nucleosome core

  • HIST1H1B K48 acetylation may have specialized functions in gene regulation compared to better-studied modifications like H3K27ac or H3K9ac

  • As a linker histone modification, it potentially affects larger domains of chromatin structure compared to core histone modifications

The functional consequences of HIST1H1B acetylation work in concert with other histone modifications to establish a complex "histone code" that regulates chromatin-dependent processes.

What are the optimal methods for detecting Acetyl-HIST1H1B (K48) in different experimental systems?

Detection of Acetyl-HIST1H1B (K48) requires specific approaches depending on the experimental context:

ApplicationRecommended MethodOptimal ConditionsNotes
Western BlottingUse specific anti-Acetyl-HIST1H1B (K48) antibody1:100-1:1000 dilution in 5% BSAInclude appropriate positive controls (e.g., sodium butyrate-treated cells)
ImmunofluorescenceConfocal microscopy with anti-Acetyl-HIST1H1B (K48)1:50-1:200 dilutionCo-staining with nuclear markers recommended
ChIPAnti-Acetyl-HIST1H1B (K48) antibody with magnetic beadsFollow ChIP-IT protocolsCan be paired with sequencing for genome-wide analysis
ELISAAnti-Acetyl-HIST1H1B (K48) in 96-well format1:2000-1:10000 dilutionUseful for quantitative measurements

For optimal results, always validate antibody specificity using appropriate negative controls (non-acetylated peptides/proteins) and positive controls (cells treated with histone deacetylase inhibitors).

How can researchers distinguish between specific acetylation at K48 versus other lysine residues in HIST1H1B?

Ensuring specificity for K48 acetylation requires careful methodological considerations:

  • Antibody selection and validation:

    • Use antibodies raised against synthetic peptides containing acetylated K48 specifically

    • Confirm specificity through peptide competition assays using acetylated and non-acetylated peptides

    • Perform western blots with recombinant HIST1H1B carrying site-directed mutations at K48

  • Mass spectrometry approaches:

    • Employ targeted LC-MS/MS to distinguish acetylation sites

    • Analyze tryptic digests to identify specific acetylated peptides

    • Quantify modification stoichiometry at different lysine residues

  • Mutational analysis:

    • Generate K48R mutants (prevents acetylation) for functional studies

    • Create K48Q mutants (mimics constitutive acetylation) to compare phenotypes

These approaches can be combined to ensure confident identification and functional assessment of K48-specific acetylation.

What sample preparation techniques yield optimal results for Acetyl-HIST1H1B (K48) detection?

Sample preparation critically affects detection of this modification:

  • Nuclear extraction protocols:

    • Use hypotonic lysis buffers with appropriate detergents

    • Include deacetylase inhibitors (e.g., sodium butyrate, TSA, nicotinamide)

    • Maintain low temperature throughout extraction to prevent enzymatic deacetylation

  • Histone isolation:

    • Acid extraction with 0.2M H₂SO₄ or HCl preserves acetylation marks

    • Salt extraction methods should include HDAC inhibitors

    • For ChIP applications, optimize crosslinking conditions (1% formaldehyde, 10 minutes)

  • Storage considerations:

    • Store samples with 50% glycerol at -80°C

    • Avoid repeated freeze-thaw cycles

    • Use protease and deacetylase inhibitor cocktails

These techniques ensure preservation of the acetylation mark during experimental procedures.

What is the role of HIST1H1B in cancer progression, particularly in breast cancer?

HIST1H1B has been implicated as a significant factor in cancer development and progression:

  • Expression patterns in cancer:

    • HIST1H1B expression is significantly upregulated in breast cancer compared to normal tissues

    • Particularly elevated in basal-like breast cancer (BLBC) subtype compared to other subtypes

    • HIST1H1B overexpression correlates with poor patient prognosis and reduced survival

  • Functional roles in tumorigenesis:

    • Promotes colony formation in soft-agar assays with breast cancer cell lines

    • Knockdown of HIST1H1B reduces tumor growth in xenograft models

    • Associated with larger tumor size and higher tumor grade in clinical samples

    • Significantly higher probability of metastasis in tumors with high HIST1H1B expression

  • Molecular mechanisms:

    • Promotes CSF2 expression through direct binding to its promoter

    • CSF2 upregulation orchestrates breast cancer growth and suppresses immune response

    • May regulate stem cell phenotype in cancer cells

    • Functions as a transcriptional regulator through chromatin remodeling

These findings strongly support HIST1H1B as a potential prognostic biomarker and therapeutic target in breast cancer, particularly the basal-like subtype.

How is acetylation of HIST1H1B at K48 affected by physiological stress conditions?

The acetylation status of HIST1H1B at K48 responds dynamically to various physiological conditions:

  • Exercise-induced changes:

    • High-intensity interval training (HIIT) increases HIST1H1B K48 acetylation despite reduced HIST1H1B protein abundance

    • This suggests selective acetylation regulation independent of protein expression levels

    • May represent an adaptive response to metabolic or oxidative stress

  • Metabolic regulation:

    • Interacts with metabolic enzymatic pathways and mitochondrial proteins

    • Changes in acetylation may reflect cellular energetic status

    • Associated with mitochondrial function through potential regulatory mechanisms

  • Stress response pathways:

    • Shows altered acetylation patterns during cellular stress conditions

    • May participate in the adaptive transcriptional response to environmental challenges

    • Potentially modifies chromatin accessibility during stress adaptation

These findings indicate that K48 acetylation represents a regulated stress-responsive modification with potential implications for cellular adaptation and gene expression control.

What genetic and epigenetic mechanisms regulate HIST1H1B expression and acetylation?

HIST1H1B expression and acetylation are controlled through multiple regulatory mechanisms:

  • Genetic regulation:

    • Copy number amplification of HIST1H1B correlates with increased expression in breast cancer

    • Genomic amplification particularly associated with the basal-like breast cancer subtype

  • Epigenetic regulation:

    • Promoter hypomethylation of HIST1H1B correlates with increased expression

    • Inverse correlation between HIST1H1B promoter methylation and mRNA levels

    • Specific methylation patterns at CpG sites in the HIST1H1B promoter region associated with expression levels

  • Acetylation regulation:

    • Regulated by histone acetyltransferases and deacetylases

    • May be influenced by metabolic intermediates (e.g., acetyl-CoA levels)

    • Dynamic process responsive to cellular signaling pathways

This multilevel regulation highlights the complex control mechanisms governing HIST1H1B expression and post-translational modifications in health and disease.

How can ChIP-Seq be optimized for genome-wide mapping of Acetyl-HIST1H1B (K48) binding patterns?

Optimization of ChIP-Seq for Acetyl-HIST1H1B (K48) requires specialized techniques:

  • Chromatin preparation:

    • Use dual crosslinking with DSG (disuccinimidyl glutarate) followed by formaldehyde

    • Optimize sonication conditions to achieve fragments of 200-300bp

    • Include spike-in controls with known acetylation patterns for normalization

  • Immunoprecipitation optimization:

    • Pre-clear chromatin with protein A/G beads to reduce background

    • Use highly specific antibodies validated for ChIP applications

    • Implement sequential ChIP for studying co-occupancy with other histone marks

  • Data analysis considerations:

    • Apply appropriate peak-calling algorithms (e.g., MACS2) optimized for histone modifications

    • Normalize to input controls and spike-in references

    • Integrate with other histone modification data for comprehensive epigenomic profiling

    • Associate binding patterns with gene expression data for functional interpretation

This workflow enables genome-wide mapping of Acetyl-HIST1H1B (K48) occupancy and correlation with transcriptional activity.

What approaches can be used to study the functional interplay between HIST1H1B acetylation and other histone modifications?

Investigating the cross-talk between HIST1H1B acetylation and other modifications requires multi-modal approaches:

  • Sequential ChIP (Re-ChIP):

    • Perform initial ChIP with anti-Acetyl-HIST1H1B (K48) antibody

    • Re-immunoprecipitate with antibodies against other modifications

    • Identify genomic regions with co-occurrence of multiple marks

  • Mass spectrometry-based combinatorial PTM analysis:

    • Use middle-down or top-down proteomics to analyze intact histone tails

    • Identify co-occurring modifications on the same histone molecule

    • Quantify modification stoichiometry and combinations

  • CRISPR-based epigenome editing:

    • Target specific writers/erasers to genomic loci

    • Analyze consequent changes in multiple histone modifications

    • Establish causal relationships between different marks

  • Proximity ligation assays:

    • Detect physical proximity between differently modified histones

    • Visualize spatial relationships between modification patterns

    • Quantify co-occurrence in different nuclear compartments

These techniques provide complementary insights into the complex interplay between HIST1H1B acetylation and the broader epigenetic landscape.

What are the current challenges in developing therapeutic approaches targeting HIST1H1B and its post-translational modifications?

Developing therapeutics targeting HIST1H1B faces several significant challenges:

  • Specificity issues:

    • Distinguishing HIST1H1B from other H1 variants

    • Selectively targeting specific modifications (e.g., K48 acetylation)

    • Avoiding off-target effects on core histones

  • Delivery challenges:

    • Nuclear delivery of potential inhibitors or modulators

    • Cell-type specific targeting in heterogeneous tissues

    • Achieving sufficient concentration at chromatin sites

  • Functional complexity:

    • Multiple and context-dependent roles of HIST1H1B

    • Integration with other epigenetic marks and chromatin regulators

    • Diverse downstream effects depending on cellular context

  • Biomarker development:

    • Developing reliable assays for HIST1H1B modifications in clinical samples

    • Correlating modification patterns with disease progression

    • Establishing predictive biomarkers for therapeutic response

Addressing these challenges requires interdisciplinary approaches combining structural biology, medicinal chemistry, cancer biology, and clinical research.

How should researchers interpret discrepancies between HIST1H1B protein abundance and K48 acetylation levels?

Interpreting apparent contradictions between protein levels and acetylation requires careful analysis:

  • Methodological considerations:

    • Antibody specificity for total versus modified protein

    • Different detection thresholds for various techniques

    • Potential masking of epitopes in certain cellular contexts

  • Biological explanations:

    • Selective acetylation of a subpopulation of HIST1H1B molecules

    • Compartmentalization of modified versus unmodified protein

    • Differential stability of acetylated versus non-acetylated forms

  • Analytical approaches:

    • Calculate modification stoichiometry (ratio of modified to total protein)

    • Perform time-course analyses to detect dynamic changes

    • Use absolute quantification methods for both total protein and acetylated form

For example, one study observed increased K48 acetylation despite reduced HIST1H1B protein abundance following exercise, suggesting selective regulation of this modification independent of protein expression .

What statistical approaches are most appropriate for analyzing changes in HIST1H1B acetylation across experimental conditions?

Robust statistical analysis of HIST1H1B acetylation data requires:

  • Normalization strategies:

    • For western blot: normalize to total HIST1H1B or other stable histone

    • For ChIP-seq: normalize to input, spike-in controls, or invariant regions

    • For proteomics: use stable isotope labeling or label-free quantification

  • Statistical tests:

    • Paired t-tests for before/after interventions in the same subjects

    • Permutation-based FDR-corrected tests for multiple comparison scenarios

    • Combined significance-fold change metrics (e.g., π-value approach)

  • Sample size considerations:

    • Power analysis based on expected effect size

    • Minimum of 3-4 biological replicates for preliminary studies

    • Larger cohorts (7+ samples) for human studies with greater variability

For example, a study examining exercise effects used permutation-based FDR-corrected paired t-tests and a significance score (π-value) that combines statistical significance with fold change to identify differentially acetylated sites .

How might single-cell technologies advance our understanding of HIST1H1B acetylation heterogeneity in complex tissues?

Single-cell approaches offer new insights into HIST1H1B biology:

  • Single-cell epigenomics approaches:

    • scCUT&Tag for profiling Acetyl-HIST1H1B (K48) in individual cells

    • Single-cell ATAC-seq to correlate with chromatin accessibility

    • Integration with scRNA-seq for linking to transcriptional output

  • Cellular heterogeneity analysis:

    • Identify cell subpopulations with distinct HIST1H1B acetylation patterns

    • Map acetylation changes during cellular differentiation trajectories

    • Characterize rare cell populations within tumors

  • Spatial epigenomics:

    • Combine imaging-based detection with single-cell sequencing

    • Map HIST1H1B modifications in the context of tissue architecture

    • Correlate with spatial transcriptomics data

These technologies would help determine whether HIST1H1B acetylation patterns differ in cancer stem cells versus differentiated tumor cells, potentially explaining heterogeneous tumor responses to therapy .

What is the potential role of HIST1H1B acetylation in the response to cancer immunotherapy?

Emerging evidence suggests HIST1H1B may influence immunotherapy response:

  • Immunomodulatory functions:

    • HIST1H1B regulates CSF2 expression, which affects immune cell function

    • May modulate the tumor microenvironment and immune infiltration

    • Could affect antigen presentation through chromatin remodeling

  • Biomarker potential:

    • HIST1H1B acetylation patterns might predict immunotherapy response

    • Could help stratify patients for appropriate immunotherapy selection

    • May serve as companion diagnostics for novel immunotherapeutic approaches

  • Therapeutic combinations:

    • Targeting HIST1H1B modifications might enhance immunotherapy efficacy

    • Combining epigenetic modulators with immune checkpoint inhibitors

    • Potential for synthetic lethality with immunotherapeutic approaches

This represents a promising frontier in cancer research that merits further investigation.

How does HIST1H1B acetylation integrate with broader metabolic networks in healthy and disease states?

The connection between HIST1H1B acetylation and metabolism represents an exciting research frontier:

  • Metabolic regulation of acetylation:

    • Acetyl-CoA availability influences histone acetylation globally

    • Changes in cellular metabolism alter acetyltransferase activity

    • Mitochondrial function may communicate with nuclear HIST1H1B acetylation

  • HIST1H1B as a metabolic sensor:

    • Acetylation may respond to cellular energetic status

    • Could integrate multiple metabolic signals to regulate gene expression

    • May function differently in metabolically reprogrammed cancer cells

  • Therapeutic implications:

    • Metabolic interventions could modulate HIST1H1B acetylation

    • Targeting metabolic pathways might normalize aberrant HIST1H1B acetylation

    • Exercise-induced metabolic changes appear to regulate HIST1H1B acetylation

This metabolic-epigenetic interface provides opportunities for novel therapeutic approaches targeting the underlying metabolic dysregulation in cancer and other diseases.

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 2024 Thebiotek. All Rights Reserved.