Propionyl-HIST1H4A (K8) Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Biological Context

Propionyl-HIST1H4A (K8) Antibody specifically recognizes histone H4 molecules modified by propionylation at lysine 8. Histone H4 is a core component of nucleosomes, and its N-terminal tail undergoes diverse PTMs—such as acetylation, methylation, and propionylation—to modulate DNA accessibility for transcription, replication, and repair . Propionylation, a less-studied modification, involves the addition of a propionyl group (-COCH2CH3) to lysine residues, potentially influencing chromatin structure and gene expression .

Antibody Development and Validation

The antibody was generated using a synthetic peptide corresponding to the propionylated K8 region of human histone H4. Key validation data include:

Specificity and Cross-Reactivity

  • Specificity: Demonstrated through competitive ELISA and immunoblotting, where pre-incubation with the immunogen peptide abolished signal .

  • Cross-Reactivity: No cross-reactivity with acetylated or butyrylated H4K8, as confirmed by peptide array assays .

Species Reactivity

Validated for human samples (Homo sapiens), with no reported cross-reactivity in mouse or rat models .

Applications

Propionyl-HIST1H4A (K8) Antibody has been optimized for multiple techniques:

ApplicationProtocol Details
Western Blot (WB)Detects ~12 kDa band in human cell lines (HeLa, A549, 293, K562) under propionate treatment .
Immunofluorescence (IF)Localizes nuclear signals in formaldehyde-fixed cells, with enhanced staining in metabolic assays .
Chromatin Immunoprecipitation (ChIP)Effective for mapping genomic regions enriched with H4K8prop, using 2–5 µg per reaction .
Immunoprecipitation (IP)Compatible with high-salt nuclear extracts for studying chromatin-bound histone complexes .

Research Findings

  • Metabolic Modulation: Treatment with sodium propionate (10 mM, 4 hours) enhances H4K8prop levels in human cell lines, suggesting a link between cellular metabolism and histone propionylation .

  • Chromatin Localization: ChIP-seq data from related H4 acetyl-K8 antibodies (e.g., ab15823, ab45166) show enrichment at transcription start sites (TSS), implying a role in transcriptional activation .

  • Disease Relevance: Aberrant H4K8 modifications are implicated in cancer and metabolic disorders, though propionylation-specific studies remain ongoing .

Comparison with Other H4K8 Antibodies

AntibodyModificationClonalityApplications
Propionyl-HIST1H4A (K8) [CAC15461]PropionylPolyclonalWB, IF, ChIP, IP
Anti-H4K8ac [ab15823]AcetylPolyclonalWB, IHC, ChIP
Anti-H4K8ac [EP1002Y] (ab45166)AcetylMonoclonalChIP-seq, Flow Cytometry

Key Advantages and Limitations

  • Advantages:

    • High specificity for propionylation over similar PTMs (e.g., acetylation) .

    • Validated in ChIP, enabling genome-wide mapping .

  • Limitations:

    • Requires antigen retrieval optimization for IHC .

    • Limited data on cross-species reactivity beyond humans .

Future Directions

Further studies are needed to elucidate the functional consequences of H4K8prop in chromatin remodeling and its interplay with other PTMs. This antibody will be critical for probing metabolic-epigenetic crosstalk in diseases like cancer and diabetes.

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your orders. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery time estimates.
Synonyms
dJ160A22.1 antibody; dJ160A22.2 antibody; dJ221C16.1 antibody; dJ221C16.9 antibody; FO108 antibody; H4 antibody; H4.k antibody; H4/a antibody; H4/b antibody; H4/c antibody; H4/d antibody; H4/e antibody; H4/g antibody; H4/h antibody; H4/I antibody; H4/j antibody; H4/k antibody; H4/m antibody; H4/n antibody; H4/p antibody; H4_HUMAN antibody; H4F2 antibody; H4F2iii antibody; H4F2iv antibody; H4FA antibody; H4FB antibody; H4FC antibody; H4FD antibody; H4FE antibody; H4FG antibody; H4FH antibody; H4FI antibody; H4FJ antibody; H4FK antibody; H4FM antibody; H4FN antibody; H4M antibody; HIST1H4A antibody; HIST1H4B antibody; HIST1H4C antibody; HIST1H4D antibody; HIST1H4E antibody; HIST1H4F antibody; HIST1H4H antibody; HIST1H4I antibody; HIST1H4J antibody; HIST1H4K antibody; HIST1H4L antibody; HIST2H4 antibody; HIST2H4A antibody; Hist4h4 antibody; Histone 1 H4a antibody; Histone 1 H4b antibody; Histone 1 H4c antibody; Histone 1 H4d antibody; Histone 1 H4e antibody; Histone 1 H4f antibody; Histone 1 H4h antibody; Histone 1 H4i antibody; Histone 1 H4j antibody; Histone 1 H4k antibody; Histone 1 H4l antibody; Histone 2 H4a antibody; histone 4 H4 antibody; Histone H4 antibody; MGC24116 antibody
Target Names
HIST1H4A
Uniprot No.

Target Background

Function
Histone H4 is a core component of the nucleosome. Nucleosomes are responsible for wrapping and compacting DNA into chromatin, thereby limiting DNA accessibility to cellular machinery requiring DNA as a template. Histones play a crucial role in regulating transcription, DNA repair, DNA replication, and maintaining chromosomal stability. The accessibility of DNA is regulated by a complex network of post-translational modifications of histones, collectively known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Studies show that PP32 and SET/TAF-Ibeta proteins block HAT1-mediated H4 acetylation. PMID: 28977641
  2. Research suggests that post-translational modifications of histones, specifically trimethylation of lysine 36 in H3 (H3K36me3) and acetylation of lysine 16 in H4 (H4K16ac), play a role in DNA damage repair. H3K36me3 stimulates H4K16ac upon DNA double-strand break. SETD2, LEDGF, and KAT5 are required for these epigenetic changes. (SETD2 = SET domain containing 2; LEDGF = lens epithelium-derived growth factor; KAT5 = lysine acetyltransferase 5) PMID: 28546430
  3. Data indicate that Omomyc protein co-localizes with proto-oncogene protein c-myc (c-Myc), protein arginine methyltransferase 5 (PRMT5), and histone H4 H4R3me2s-enriched chromatin domains. PMID: 26563484
  4. H4K12ac is regulated by estrogen receptor-alpha and is associated with BRD4 function and inducible transcription. PMID: 25788266
  5. Systemic lupus erythematosus appears to be associated with an imbalance in histone acetyltransferases and histone deacetylase enzymes, favoring pathologic H4 acetylation. PMID: 25611806
  6. Sumoylated human histone H4 prevents chromatin compaction by inhibiting long-range internucleosomal interactions. PMID: 25294883
  7. Acetylation at lysine 5 of histone H4 is associated with lytic gene promoters during reactivation of Kaposi's sarcoma-associated herpesvirus. PMID: 25283865
  8. An increase in histone H4 acetylation caused by hypoxia in human neuroblastoma cell lines corresponds to increased levels of N-myc transcription factor in these cells. PMID: 24481548
  9. Data indicate that G1-phase histone assembly is restricted to CENP-A and H4. PMID: 23363600
  10. This study focused on the distribution of a specific histone modification, namely H4K12ac, in human sperm and characterized its specific enrichment sites in promoters throughout the whole human genome. PMID: 22894908
  11. SRP68/72 heterodimers are major nuclear proteins whose binding of histone H4 tail is inhibited by H4R3 methylation. PMID: 23048028
  12. TNF-alpha inhibition of AQP5 expression in human salivary gland acinar cells is due to the epigenetic mechanism by suppression of acetylation of histone H4. PMID: 21973049
  13. Our data suggest that global histone H3 and H4 modification patterns are potential markers of tumor recurrence and disease-free survival in non-small cell lung cancer. PMID: 22360506
  14. HAT1 differentially impacts nucleosome assembly of H3.1-H4 and H3.3-H4. PMID: 22228774
  15. Phosphorylation of histone H4 Ser 47 catalyzed by the PAK2 kinase promotes nucleosome assembly of H3.3-H4 and inhibits nucleosome assembly of H3.1-H4 by increasing the binding affinity of HIRA to H3.3-H4 and reducing association of CAF-1 with H3.1-H4. PMID: 21724829
  16. The imatinib-induced hemoglobinization and erythroid differentiation in K562 cells are associated with global histone H4 acetylation. PMID: 20949922
  17. Our findings reveal the molecular mechanisms whereby the DNA sequences within specific gene bodies are sufficient to nucleate the monomethylation of histone H4 lysine 200, which in turn reduces gene expression by half. PMID: 20512922
  18. Histone H4 expression is downregulated by zinc and upregulated by docosahexaenoate in a neuroblastoma cell line. PMID: 19747413
  19. Low levels of histone acetylation are associated with the development and progression of gastric carcinomas, possibly through alteration of gene expression. PMID: 12385581
  20. Overexpression of MTA1 protein and acetylation level of histone H4 protein are closely related. PMID: 15095300
  21. Peptidylarginine deiminase 4 regulates histone Arg methylation by converting methyl-Arg to citrulline and releasing methylamine. Data suggest that PAD4 mediates gene expression by regulating Arg methylation and citrullination in histones. PMID: 15345777
  22. The lack of biotinylation of K12 in histone H4 is an early signaling event in response to double-strand breaks. PMID: 16177192
  23. Incorporation of acetylated histone H4-K16 into nucleosomal arrays inhibits the formation of compact 30-nanometer-like fibers and impedes the ability of chromatin to form cross-fiber interactions. PMID: 16469925
  24. Apoptosis is associated with global DNA hypomethylation and histone deacetylation events in leukemia cells. PMID: 16531610
  25. BTG2 contributes to retinoic acid activity by favoring differentiation through a gene-specific modification of histone H4 arginine methylation and acetylation levels. PMID: 16782888
  26. There is a relationship between histone H4 modification, epigenetic regulation of BDNF gene expression, and long-term memory for extinction of conditioned fear. PMID: 17522015
  27. The H4 tail and its acetylation have novel roles in mediating recruitment of multiple regulatory factors that can change chromatin states for transcription regulation. PMID: 17548343
  28. Brd2 bromodomain 2 is monomeric in solution and dynamically interacts with H4-AcK12. Additional secondary elements in the long ZA loop may be a common characteristic of BET bromodomains. PMID: 17848202
  29. Spermatids Hypac-H4 impairment in mixed atrophy did not deteriorate further by AZFc region deletion. PMID: 18001726
  30. The SET8 and PCNA interaction couples H4-K20 methylation with DNA replication. PMID: 18319261
  31. H4K20 monomethylation and PR-SET7 are important for L3MBTL1 function. PMID: 18408754
  32. High expression of acetylated H4 is more common in aggressive than indolent cutaneous T-cell lymphoma. PMID: 18671804
  33. Our findings indicate an important role of histone H4 modifications in bronchial carcinogenesis. PMID: 18974389
  34. Results indicate, by acetylation of histone H4 K16 during S-phase, early replicating chromatin domains acquire the H4K16ac-K20me2 epigenetic label that persists on the chromatin throughout mitosis and is deacetylated in early G1-phase of the next cell cycle. PMID: 19348949
  35. Acetylated H4 is overexpressed in diffuse large B-cell lymphoma and peripheral T-cell lymphoma relative to normal lymphoid tissue. PMID: 19438744
  36. The release of histone H4 by holocrine secretion from the sebaceous gland may play an important role in innate immunity. PMID: 19536143
  37. Histone modification including PRC2-mediated repressive histone marker H3K27me3 and active histone marker acH4 may be involved in CD11b transcription during HL-60 leukemia cells reprogramming to terminal differentiation. PMID: 19578722
  38. A role of Cdk7 in regulating elongation is further suggested by enhanced histone H4 acetylation and diminished histone H4 trimethylation on lysine 36 - two marks of elongation - within genes when the kinase was inhibited. PMID: 19667075
  39. Data showed the dynamic fluctuation of histone H4 acetylation levels during mitosis, as well as acetylation changes in response to structurally distinct histone deacetylase inhibitors. PMID: 19805290
  40. Data directly implicate BBAP in the monoubiquitylation and additional posttranslational modification of histone H4 and an associated DNA damage response. PMID: 19818714

Show More

Hide All

Database Links

HGNC: 4781

OMIM: 142750

KEGG: hsa:121504

STRING: 9606.ENSP00000367034

UniGene: Hs.143080

Involvement In Disease
Chromosomal aberrations involving HISTONE H4 is a cause of B-cell non-Hodgkin lymphomas (B-cell NHL). Translocation t(3;6)(q27;p21), with BCL6.
Protein Families
Histone H4 family
Subcellular Location
Nucleus. Chromosome.

Q&A

What is Propionyl-HIST1H4A (K8) Antibody and what specific modification does it recognize?

Propionyl-HIST1H4A (K8) Antibody is a primary antibody that specifically recognizes histone H4 protein that has been propionylated at lysine 8 (K8). This post-translational modification is part of the histone code that regulates chromatin structure and gene expression. The antibody targets the peptide sequence surrounding the propionylated lysine 8 position in human Histone H4 and enables researchers to study this specific epigenetic modification .

Unlike general Histone H4 antibodies, this specific antibody allows researchers to distinguish propionylation at K8 from other modifications like acetylation or methylation at the same position. The antibody is typically generated in rabbits (for polyclonal versions) and can be used across multiple experimental applications including Western blotting, immunofluorescence, chromatin immunoprecipitation (ChIP), and ELISA .

How does Propionyl-HIST1H4A (K8) modification differ from other histone H4 modifications?

Histone H4 can undergo various post-translational modifications at different residues, with propionylation at K8 being one specific type. To understand its uniqueness, we can compare it with other common modifications:

Modification TypePositionFunctionDetection Method
PropionylationK8Gene regulation, metabolic sensingPropionyl-HIST1H4A (K8) Antibody
AcetylationK5, K8, K12, K16Transcription activation, DNA repairAcetyl-specific antibodies
MethylationK20Transcription repression, DNA damage responseMethyl-specific antibodies
PhosphorylationS1Mitosis, DNA damagePhospho-specific antibodies

Propionylation at K8 is considered distinct from acetylation, though both are acylation modifications. While acetylation at H4K8 is well-characterized and associated with active transcription , propionylation may represent a unique regulatory mechanism potentially linked to cellular metabolism. The chemical difference between these modifications (propionyl group is larger than acetyl group) may result in distinct functional outcomes and recognition by different reader proteins.

What experimental applications is Propionyl-HIST1H4A (K8) Antibody validated for?

The Propionyl-HIST1H4A (K8) Antibody has been validated for multiple research applications:

  • Enzyme-Linked Immunosorbent Assay (ELISA) - For quantitative detection of propionylated H4K8 in purified samples

  • Western Blotting (WB) - For detecting the presence and relative abundance of propionylated H4K8 in cell or tissue lysates

  • Immunofluorescence (IF) - For visualizing the nuclear localization and distribution patterns of propionylated H4K8

  • Immunoprecipitation (IP) - For isolating propionylated H4K8-containing complexes

  • Chromatin Immunoprecipitation (ChIP) - For identifying genomic regions associated with propionylated H4K8

The methodological approach for each application must be optimized based on the specific antibody characteristics. For Western blotting applications, a typical working dilution might be 1:500 to 1:1000 . For immunofluorescence, proper fixation is critical to preserve nuclear architecture while maintaining epitope accessibility. ChIP applications require careful optimization of chromatin fragmentation and antibody binding conditions to ensure specific enrichment of propionylated H4K8-associated genomic regions.

How should I design ChIP experiments using Propionyl-HIST1H4A (K8) Antibody?

Designing effective ChIP experiments with Propionyl-HIST1H4A (K8) Antibody requires careful planning and optimization:

  • Sample Preparation:

    • Crosslink cells with 1% formaldehyde for 10 minutes at room temperature to preserve protein-DNA interactions

    • Quench with 125mM glycine for 5 minutes

    • Lyse cells and isolate nuclei using appropriate buffers

    • Sonicate chromatin to generate fragments of 200-500bp (optimize sonication conditions for your cell type)

  • Immunoprecipitation:

    • Pre-clear chromatin with protein A/G beads

    • Incubate 2-5μg of Propionyl-HIST1H4A (K8) Antibody with chromatin overnight at 4°C

    • Include appropriate controls: IgG negative control and a positive control antibody

    • Add protein A/G beads and incubate for 2-3 hours

    • Wash thoroughly to remove non-specific binding

  • DNA Recovery and Analysis:

    • Reverse crosslinks and purify DNA

    • Analyze by qPCR targeting regions of interest or perform sequencing (ChIP-seq)

Critical validation is necessary to confirm antibody specificity. Research using H4K8 modification antibodies has shown enrichment around transcription start sites , so include these regions in your initial validation experiments. For ChIP-seq analysis, consider using established peak-calling algorithms and create genome browser tracks to visualize the distribution of propionylated H4K8 across the genome.

What controls should I include when using Propionyl-HIST1H4A (K8) Antibody in immunofluorescence studies?

When performing immunofluorescence with Propionyl-HIST1H4A (K8) Antibody, include these essential controls:

  • Primary Antibody Controls:

    • Negative control: Omit primary antibody but include all other steps

    • Isotype control: Use non-specific IgG from the same species (rabbit)

    • Peptide competition: Pre-incubate antibody with propionylated H4K8 peptide to confirm specificity

    • Non-propionylated control: Use cells treated with propionylation inhibitors to confirm signal specificity

  • Sample Controls:

    • Positive control: Include cell types known to have high levels of H4K8 propionylation

    • Treatment comparison: Compare cells before and after treatments known to affect histone propionylation (e.g., propionyl-CoA modulators)

  • Technical Controls:

    • Autofluorescence control: Examine unstained samples to detect any natural fluorescence

    • Secondary antibody control: Omit primary antibody to check for non-specific binding of secondary antibody

Document the pattern of nuclear staining, which should be consistent with chromatin localization. Based on studies of histone modifications, you might expect certain patterns of nuclear distribution that correlate with chromatin states. Typical working dilutions for immunofluorescence might range from 1:100 to 1:500, but optimization is necessary for each specific antibody preparation .

How can I optimize Western blot protocols for detecting Propionyl-HIST1H4A (K8)?

Optimizing Western blot protocols for Propionyl-HIST1H4A (K8) detection requires attention to several specific factors:

  • Sample Preparation:

    • Extract histones using specialized acid extraction methods (e.g., 0.2N HCl extraction)

    • Include deacetylase and propionylation inhibitors in lysis buffers

    • Use freshly prepared samples when possible, as modifications can be lost during storage

  • Gel Electrophoresis:

    • Use high percentage (15-18%) SDS-PAGE gels to resolve the low molecular weight histone proteins

    • Consider using Triton-Acid-Urea (TAU) gels for better separation of modified histones

    • Load appropriate amount of histone sample (typically 5-15μg of acid-extracted histones)

  • Transfer and Detection:

    • Use PVDF membrane with 0.2μm pore size (rather than 0.45μm) for small proteins

    • Optimize transfer conditions: typically 30V overnight at 4°C works well for histones

    • Block with 5% BSA rather than milk (which contains proteins that may cross-react)

    • Use antibody at recommended dilution (typically 1:500 to 1:1000)

    • Include a loading control (total H4 or H3)

  • Validation Steps:

    • Run a peptide competition control

    • Include samples with known levels of H4K8 propionylation

Expected results: Histone H4 is approximately 11kDa in size . The Propionyl-HIST1H4A (K8) Antibody should detect a single band at this molecular weight, with intensity varying based on the degree of propionylation at K8 in your samples.

How can I differentiate between propionylation and acetylation at H4K8 in my experimental data?

Differentiating between propionylation and acetylation at H4K8 is challenging due to their similar chemical structures. Here's a methodological approach:

  • Antibody Specificity Testing:

    • Perform peptide competition assays using propionylated and acetylated H4K8 peptides

    • Test cross-reactivity using dot blots with modified peptides

    • Use modified histone arrays containing different modifications

  • Mass Spectrometry Validation:

    • Employ LC-MS/MS to confirm the presence of propionylation vs. acetylation

    • Look for mass shifts: propionylation adds 56 Da, while acetylation adds 42 Da

    • Use targeted MS approaches to quantify relative abundance of each modification

  • Functional Validation:

    • Manipulate cellular propionyl-CoA and acetyl-CoA levels to shift the modification balance

    • Examine differential enzyme sensitivity: some histone deacetylases might not remove propionyl groups

    • Use specific enzymatic inhibitors to modulate each modification separately

What are the expected genomic distribution patterns of Propionyl-HIST1H4A (K8) based on ChIP-seq data?

Based on studies of histone H4 modifications, we can infer some expected genomic distribution patterns for Propionyl-HIST1H4A (K8):

  • Promoter and Enhancer Regions:

    • Similar to H4K8 acetylation, propionylation is likely enriched around transcription start sites (TSS)

    • Based on evidence that acetylation of H4K8 is enriched around TSS, propionylation may follow similar patterns

    • Expect possible enrichment at enhancer regions, particularly those active in metabolic regulation

  • Gene Bodies:

    • May show differential patterns compared to acetylation

    • Possibly enriched in the gene bodies of metabolically regulated genes

    • Could correlate with exonic rather than intronic regions in actively transcribed genes

  • Correlation with Chromatin States:

    • Likely associated with euchromatin (open chromatin regions)

    • May correlate with other active marks like H3K4me3 and H3K27ac

    • Potentially absent or depleted in heterochromatic regions marked by H3K9me3 or H3K27me3

  • Analysis Methodology:

    • Perform peak calling using MACS2 or similar algorithms

    • Generate heatmaps and aggregate plots centered on TSS

    • Compare distribution with other histone marks using correlation analysis

    • Perform Gene Ontology enrichment analysis on propionylation-associated genes

ChIP-seq analysis has shown that acetylation of H4K8 and H4K16 are enriched around transcription start sites , and propionylation may exhibit similar or distinct patterns depending on its functional role in transcriptional regulation.

How should I integrate Propionyl-HIST1H4A (K8) ChIP-seq data with transcriptome data?

Integrating ChIP-seq data for Propionyl-HIST1H4A (K8) with transcriptome data requires a systematic approach:

  • Experimental Design Considerations:

    • Collect matched samples for ChIP-seq and RNA-seq from the same experimental conditions

    • Include appropriate controls for both assays

    • Consider time course experiments to capture dynamic relationships

  • Data Processing Workflow:

    • Process ChIP-seq data: quality control, alignment, peak calling, signal normalization

    • Process RNA-seq data: quality control, alignment, quantification, differential expression analysis

    • Use consistent genome builds and annotations for both datasets

  • Integration Analysis Methods:

    • Assign ChIP-seq peaks to genes (e.g., by proximity to TSS or within gene bodies)

    • Correlate propionylation signal intensity with gene expression levels

    • Classify genes based on presence/absence of propionylation and expression status

    • Perform gene set enrichment analysis on co-regulated genes

  • Visualization Approaches:

    • Create scatter plots of propionylation signal vs. expression level

    • Generate genome browser tracks showing both propionylation and RNA-seq data

    • Produce heatmaps clustering genes by propionylation pattern and expression

This integrative analysis can reveal whether H4K8 propionylation serves primarily as an activating mark (like many acetylation marks) or has more complex regulatory functions. Based on studies of histone acetylation around transcription start sites , you might expect positive correlation between propionylation signal at promoters and gene expression levels, though the relationship may be more nuanced for specific gene subsets.

How can Propionyl-HIST1H4A (K8) Antibody be used to investigate links between metabolism and epigenetic regulation?

Integrating Propionyl-HIST1H4A (K8) Antibody into metabolic epigenetics research provides a powerful approach to investigate links between cellular metabolism and gene regulation:

  • Experimental Design Strategies:

    • Perform parallel ChIP-seq (with Propionyl-HIST1H4A (K8) Antibody), RNA-seq, and metabolomics on matched samples

    • Apply metabolic perturbations (e.g., propionate supplementation, propionyl-CoA synthetase modulation)

    • Include time course analyses to capture dynamic responses

    • Compare different metabolic states (fed vs. fasted, different carbon sources)

  • Key Metabolic Conditions to Test:

    • Propionate supplementation (direct precursor to propionyl-CoA)

    • Branched-chain amino acid manipulation (source of propionyl-CoA)

    • Odd-chain fatty acid metabolism perturbation

    • Fasting/feeding cycles (alters metabolic flux)

    • Hypoxia (changes metabolic pathways)

  • Analytical Approaches:

    • Monitor changes in global H4K8 propionylation levels via Western blotting

    • Map genomic distribution changes via ChIP-seq

    • Correlate propionylation changes with transcriptomic responses

    • Measure propionyl-CoA/acetyl-CoA ratios and correlate with propionylation

Propionylation is particularly relevant for metabolic studies because propionyl-CoA levels fluctuate with diet, microbiome activity, and metabolic state. This makes H4K8 propionylation a potential sensor that connects environmental inputs to epigenetic regulation. Using Propionyl-HIST1H4A (K8) Antibody allows researchers to track how these metabolic changes affect the epigenome.

What approaches should I use to study Propionyl-HIST1H4A (K8) in different cell types and tissues?

Studying H4K8 propionylation across different biological contexts requires tailored methodological approaches:

  • Tissue and Cell Type Comparison Strategies:

    • Create a tissue panel for Western blot analysis of propionylation levels

    • Perform immunohistochemistry on tissue microarrays using optimized protocols

    • Isolate primary cells from different tissues for comparative ChIP-seq

    • Consider single-cell approaches for heterogeneous tissues

  • Optimization for Challenging Samples:

    • For tissue samples: Optimize crosslinking conditions and chromatin extraction

    • For limited material: Adapt micro-ChIP protocols for Propionyl-HIST1H4A (K8) Antibody

    • For fixed clinical samples: Test epitope retrieval methods to restore antibody reactivity

    • For highly specialized cell types: Develop FACS sorting strategies before ChIP

  • Comparative Analysis Framework:

    • Create standardized protocols to enable direct comparison between samples

    • Include reference cell lines in each experiment batch as technical controls

    • Develop quantitative metrics for propionylation levels

    • Consider tissue-specific expression of propionylation writers/erasers

  • Functional Validation in Different Contexts:

    • Test metabolic sensitivities of propionylation in tissue-specific manner

    • Examine cell type-specific readers of propionylation

    • Assess functional consequences of propionylation disruption

Different cell types may exhibit distinct patterns of H4K8 propionylation reflecting their metabolic preferences and gene expression programs. Metabolically active tissues (liver, muscle, brain) might show higher or more dynamic propionylation patterns than quiescent tissues. The Propionyl-HIST1H4A (K8) Antibody enables mapping these tissue-specific epigenetic landscapes.

How can Propionyl-HIST1H4A (K8) Antibody be used in studies of cell differentiation and development?

Applying Propionyl-HIST1H4A (K8) Antibody in developmental biology research requires specialized approaches:

  • Developmental Time Course Analysis:

    • Track propionylation changes during differentiation processes

    • Compare propionylation patterns across embryonic stages

    • Analyze different cell lineages as they diverge from progenitor cells

    • Correlate with expression of developmental regulators

  • Cell Type-Specific Profiling:

    • Combine with cell sorting to isolate specific progenitor or differentiated populations

    • Use single-cell approaches to capture heterogeneity in propionylation

    • Perform ChIP-seq with Propionyl-HIST1H4A (K8) Antibody across distinct cell types

    • Correlate propionylation patterns with cell fate decisions

  • Functional Studies:

    • Modulate propionylation during critical developmental windows

    • Assess effects on lineage commitment and differentiation potential

    • Test differentiation capacity after inhibiting propionylation writers/erasers

    • Create reporter systems to monitor propionylation dynamics in real-time

During development, cellular metabolism undergoes significant changes that may affect propionyl-CoA availability and consequently H4K8 propionylation. These changes could contribute to cell fate decisions and lineage commitment through epigenetic mechanisms. The Propionyl-HIST1H4A (K8) Antibody provides a tool to investigate these connections between metabolic state changes and epigenetic reprogramming during development.

What are common challenges when using Propionyl-HIST1H4A (K8) Antibody and how can they be addressed?

Researchers working with Propionyl-HIST1H4A (K8) Antibody may encounter several challenges:

  • Specificity Issues:

    • Challenge: Cross-reactivity with acetylated H4K8 due to structural similarity

    • Solution: Perform peptide competition assays with both modifications; optimize antibody concentration; confirm with mass spectrometry

  • Signal Strength Problems:

    • Challenge: Weak signal due to low abundance of propionylation

    • Solution: Increase antibody concentration; optimize incubation conditions; use signal amplification methods; enrich for histones before analysis

  • Background and Non-specific Binding:

    • Challenge: High background in immunofluorescence or Western blots

    • Solution: Optimize blocking (use BSA instead of milk) ; increase washing stringency; pre-absorb antibody; titrate antibody concentration

  • Sample Preparation Issues:

    • Challenge: Loss of modification during extraction

    • Solution: Include propionylation inhibitors in lysis buffers; use fresh samples; optimize extraction protocols to preserve modifications

How should I validate the specificity of my Propionyl-HIST1H4A (K8) Antibody?

Thorough validation of Propionyl-HIST1H4A (K8) Antibody specificity is critical for reliable research findings:

  • Peptide Competition Assays:

    • Test antibody binding with and without pre-incubation with propionylated H4K8 peptide

    • Include acetylated H4K8 peptide to test cross-reactivity

    • Use unmodified H4K8 peptide as a negative control

    • Analyze in Western blot, ELISA, or dot blot format

  • Modified Histone Panel Testing:

    • Test antibody against recombinant histones with defined modifications

    • Include H4 with various modifications (propionylation, acetylation, butyrylation at K8)

    • Test H4 with propionylation at other lysine residues (K5, K12, K16)

    • Test for sensitivity to neighboring modifications

  • Genetic/Enzymatic Validation:

    • Compare samples with knockdown/knockout of propionylation writers

    • Test samples treated with propionylation inhibitors

    • Use in vitro enzymatic assays to create or remove propionylation

    • Validate with mass spectrometry as an orthogonal method

  • Application-Specific Validation:

    • For ChIP: Perform sequential ChIP with general H4 antibody

    • For IF: Include peptide-blocking controls directly on slides

    • For WB: Include multiple controls and ladder of modified recombinant proteins

What are emerging alternatives to antibody-based detection of histone propionylation?

Understanding the limitations of current antibody-based approaches and exploring alternatives is important for advancing research on histone propionylation:

  • Current Antibody Limitations:

    • Potential cross-reactivity with similar modifications (especially acetylation)

    • Batch-to-batch variability affecting reproducibility

    • Limited ability to detect combinatorial modifications on the same histone tail

    • Qualitative rather than truly quantitative results

  • Mass Spectrometry Approaches:

    • Bottom-up, middle-down, and top-down proteomics for histone analysis

    • Targeted MS approaches for specific modification quantification

    • Advantages: Can detect combinatorial modifications; more quantitative

    • Disadvantages: Requires specialized equipment; limited spatial information

  • Chemical Biology Methods:

    • Chemical probes for specific acyl modifications

    • Click chemistry approaches for modification labeling

    • Advantages: Can track dynamics in living cells; less dependent on antibody quality

    • Disadvantages: May require genetic engineering; potential off-target effects

  • Next-Generation Sequencing Based Methods:

    • CUT&RUN or CUT&Tag as alternatives to traditional ChIP

    • Advantages: Requires less material; potentially higher resolution

    • Disadvantages: Still relies on antibody specificity; more complex workflow

While antibodies remain valuable tools, especially for applications like ChIP and immunofluorescence, researchers should consider complementary approaches when designing experiments to study histone propionylation. The ideal strategy often combines multiple technologies to overcome the limitations of any single approach.

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.