Tri-methyl-Histone H4 (K20) Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Biological Context

Histone H4 lysine 20 trimethylation (H4K20me3) is a conserved epigenetic mark enriched in pericentric heterochromatin and silenced genomic regions. This modification is catalyzed by Suv4-20h methyltransferases and plays roles in DNA repair, replication timing, and chromatin compaction . The tri-methyl-Histone H4 (K20) recombinant monoclonal antibody is engineered to specifically recognize this modification, excluding mono- or dimethylated states .

Antibody Development and Characterization

Recombinant monoclonal antibodies targeting H4K20me3 are produced using advanced cloning and hybridoma technologies. Key steps include:

  • Immunogen Design: A synthetic peptide corresponding to residues 15–25 of human histone H4, trimethylated at K20 (e.g., sequence AKRHR(Kme3)VLRDN) .

  • Host and Clonality: Typically generated in rabbits or mice, with monoclonal clones (e.g., clone 6F8-D9 in mice or EPR17001(2) in rabbits) .

  • Validation: Rigorous specificity testing via dot blot, Western blot (WB), and immunofluorescence (IF), with no cross-reactivity to unmodified H4 or other methylation states .

Key Applications

ApplicationDetailsRecommended Dilution
Western Blot (WB)Detects ~11 kDa band in nuclear extracts (e.g., HeLa, NIH 3T3 cells) .1:500–1:5000
Immunohistochemistry (IHC)Nuclear staining in paraffin-embedded tissues (e.g., mouse liver, human colon) .1:50–1:8000
Immunofluorescence (IF)Localizes H4K20me3 to heterochromatin foci in fixed cells .1:50–1:500
Chromatin Immunoprecipitation (ChIP)Maps H4K20me3 enrichment at pericentric repeats .Vendor-specific protocols

Research Findings Enabled by This Antibody

  • Heterochromatin Regulation:
    H4K20me3 colocalizes with H3K9me3 at pericentric heterochromatin. RNAi knockdown of Suv4-20h enzymes abolishes H4K20me3, linking it to heterochromatin maintenance .

  • Disease Associations:
    Dysregulated H4K20me3 is observed in cancers, with reduced levels in hepatocellular carcinoma and gliomas .

  • Chromatin Dynamics:
    During DNA repair, H4K20me3 facilitates recruitment of 53BP1 to double-strand breaks, highlighting its role in genome integrity .

Comparative Data Table

ParameterCAB2372 (Assay Genie)CSB-RA010429A20me3HU (Cusabio)ab177190 (Abcam)
Host SpeciesRabbitRabbitRabbit
ClonalityPolyclonalMonoclonalRecombinant Monoclonal
ApplicationsWB, IHC, IF, ELISAWB, ICCChIP, WB, IHC
Dilution Range1:50–1:20001:500–1:50001:1000–1:8000
ImmunogenSynthetic peptideSynthetic peptideRecombinant protein

Technical Considerations

  • Specificity: Validated using peptide competition assays; no cross-reactivity with H4K20me1/2 or unmodified H4 .

  • Storage: Stable at -20°C or -80°C in glycerol-containing buffers .

  • Limitations: Not suitable for diagnostic use; optimal dilution varies by cell type and fixation method .

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Description

This tri-methyl-histone H4 (K20) recombinant monoclonal antibody is produced through a meticulous process. Rabbit genes encoding the HIST1H4A antibody are isolated following immunization with a synthetic peptide derived from the human HIST1H4A protein tri-methylated at K20. These genes are then cloned into specialized expression vectors and introduced into host suspension cells, where they are cultured to induce antibody expression and secretion. The antibody is then purified using affinity chromatography, separating it from the cell culture supernatant. The functionality of the antibody is meticulously validated through ELISA, WB, and ICC tests, confirming its ability to interact effectively with the human HIST1H4A protein tri-methylated at K20.

Tri-methylation of HIST1H4A at K20 is an epigenetic modification linked to gene repression and the formation of repressive chromatin structures such as heterochromatin and silenced gene loci. This modification plays a crucial role in regulating gene expression, maintaining chromatin integrity, and contributing to genome stability.

Form
Liquid
Lead Time
Typically, we can dispatch products within 1-3 working days after receiving your orders. Delivery times may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery time information.
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 nucleosomes. Nucleosomes wrap and compact DNA into chromatin, restricting DNA accessibility to cellular machineries that require DNA as a template. Consequently, histones play a central role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is regulated through a complex set of post-translational modifications of histones, often referred to as the histone code, and nucleosome remodeling.

Gene References Into Functions
  1. Studies have shown that PP32 and SET/TAF-Ibeta proteins inhibit HAT1-mediated H4 acetylation. PMID: 28977641
  2. Research indicates that post-translational modifications of histones, specifically trimethylation of lysine 36 in H3 (H3K36me3) and acetylation of lysine 16 in H4 (H4K16ac), are involved in DNA damage repair. H3K36me3 stimulates H4K16ac upon DNA double-strand break, and this process requires the presence of SETD2, LEDGF, and KAT5. (SETD2 = SET domain containing 2; LEDGF = lens epithelium-derived growth factor; KAT5 = lysine acetyltransferase 5) PMID: 28546430
  3. Data suggest 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 linked to 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 identified as 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 attributed to the epigenetic mechanism of suppression of acetylation of histone H4. PMID: 21973049
  13. 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. Imatinib-induced hemoglobinization and erythroid differentiation in K562 cells are associated with global histone H4. PMID: 20949922
  17. Research reveals that 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. Expression of histone H4 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, potentially through alteration of gene expression. PMID: 12385581
  20. Overexpression of MTA1 protein and acetylation levels 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 play 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 crucial 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. Findings indicate an important role of histone H4 modifications in bronchial carcinogenesis. PMID: 18974389
  34. Results suggest that acetylation of histone H4 K16 during S-phase allows early replicating chromatin domains to acquire the H4K16ac-K20me2 epigenetic label, which 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 a significant 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 supported 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 show 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 the biological significance of Histone H4 K20 tri-methylation?

Histone H4 lysine 20 tri-methylation (H4K20me3) is a post-translational modification that plays a central role in epigenetic regulation. This modification is primarily associated with gene repression and the formation of repressive chromatin structures, such as heterochromatin and silenced gene loci. It is integral to multiple cellular processes, including:

  • Regulation of gene expression through heterochromatin formation

  • Maintenance of genome stability

  • DNA damage response and repair pathways

  • Cell cycle progression and mitotic regulation

  • Chromatin integrity preservation during cellular differentiation

H4K20me3 is a core component of the "histone code" that regulates DNA accessibility to cellular machinery requiring DNA as a template for processes like transcription, replication, and repair . The presence of this modification typically creates a more condensed chromatin structure, limiting access of transcription factors and RNA polymerase machinery to DNA, thereby contributing to gene silencing.

What are the recommended applications for Tri-methyl-Histone H4 (K20) antibodies?

Tri-methyl-Histone H4 (K20) antibodies have been validated for multiple experimental applications, with specific recommendations for optimal dilutions and sample preparations:

ApplicationRecommended DilutionSample Types
Western Blot (WB)1:500-1:5000 (CUSABIO) 1:2000 (Abcam) Cell lysates, nuclear extracts, tissue homogenates
Immunocytochemistry (ICC/IF)1:50-1:300 (CUSABIO) 1:500 (Abcam) Fixed and permeabilized cells
Immunohistochemistry (IHC-P)1:500 (Abcam) Paraffin-embedded tissue sections
Chromatin Immunoprecipitation (ChIP)Varies by protocolCrosslinked chromatin
Dot Blot (DB)1:5000 (Abcam) Purified peptides, nuclear extracts
Immunoprecipitation (IP)Varies by protocolNuclear extracts, whole cell lysates

These antibodies have been successfully applied to samples from human, mouse, and rat origins, with demonstrated reactivity against synthetic peptides containing the H4K20me3 modification . When designing experiments, researchers should consider the specific cellular localization of the target epitope, which is primarily nuclear and associated with heterochromatic regions.

How can researchers validate the specificity of Tri-methyl-Histone H4 (K20) antibodies?

Validation of antibody specificity is critical for accurate experimental interpretation. For Tri-methyl-Histone H4 (K20) antibodies, several validation approaches are recommended:

  • Peptide competition assays: Using synthetic peptides containing H4K20me3, H4K20me2, H4K20me1, and unmodified H4K20 to confirm selective binding to the tri-methylated form.

  • Dot blot analysis: Testing reactivity against a concentration gradient of synthetic peptides with different methylation states (as demonstrated in the dot blot analysis from Abcam showing selective binding to H4K20me3 over H4K20me2, H4K20me1, and unmodified H4K20) .

  • Western blot with nuclear fractions: Confirming specific band at approximately 11 kDa (the predicted size of Histone H4) in nuclear extracts .

  • Immunofluorescence localization: Verifying nuclear localization pattern consistent with heterochromatin distribution, typically visualized as punctate nuclear staining that colocalizes with heterochromatin markers .

  • Knockout/knockdown validation: Comparing antibody signal in wild-type samples versus samples with reduced H4K20me3 levels through knockdown of methyltransferases (such as SUV420H1/H2).

  • Cross-reactivity testing: Evaluating potential binding to other tri-methylated histone lysine residues (e.g., H3K9me3, H3K27me3).

Dot blot analysis has demonstrated that high-quality anti-H4K20me3 antibodies show strong affinity for H4K20me3 with minimal cross-reactivity to H4K20me2, H4K20me1, or unmodified H4K20 peptides, confirming their specificity for the tri-methylation state .

What are the optimal storage and handling conditions for these antibodies?

Proper storage and handling are essential for maintaining antibody activity and specificity. For Tri-methyl-Histone H4 (K20) antibodies, the following conditions are recommended:

  • Storage temperature: Store at 2-8°C for short-term use (up to 1 year from receipt date) .

  • Long-term storage: For periods exceeding 1 year, aliquot and store at -20°C to minimize freeze-thaw cycles.

  • Buffer composition: Typically stored in buffers containing 0.1 M Tris-Glycine (pH 7.4), 150 mM NaCl with 0.05% sodium azide .

  • Freeze-thaw cycles: Minimize repeated freeze-thaw cycles by preparing small aliquots before freezing.

  • Working dilutions: Prepare fresh working dilutions on the day of the experiment.

  • Contamination prevention: Use sterile techniques when handling to prevent microbial contamination.

  • Safety considerations: Be aware that some formulations contain sodium azide, which is a toxic compound and should be handled accordingly .

Following these guidelines will help ensure consistent antibody performance and reproducible experimental results over time.

How does the performance of polyclonal versus recombinant monoclonal anti-H4K20me3 antibodies compare in complex chromatin studies?

The choice between polyclonal and recombinant monoclonal anti-H4K20me3 antibodies significantly impacts experimental outcomes in chromatin studies. Each antibody type offers distinct advantages and limitations:

Polyclonal Antibodies (e.g., Abcam ab227884, Merck 07-463):

  • Epitope recognition: Recognize multiple epitopes on the H4K20me3 modification, potentially increasing signal strength but also increasing the risk of non-specific binding.

  • Batch variation: May exhibit lot-to-lot variability, necessitating validation of each new lot.

  • Sensitivity: Often provide higher sensitivity in applications like Western blotting and immunohistochemistry where signal amplification is beneficial.

  • ChIP applications: Studies using polyclonal anti-H4K20me3 antibodies for ChIP have successfully mapped this modification to heterochromatic regions and silenced gene loci .

Recombinant Monoclonal Antibodies (e.g., CUSABIO CSB-RA010429A20me3HU):

  • Specificity: Provide highly consistent epitope recognition with reduced background, crucial for distinguishing between H4K20me3 and other methylation states.

  • Reproducibility: Offer superior lot-to-lot consistency due to recombinant production methods.

  • Sequential ChIP (Re-ChIP): More suitable for sequential ChIP experiments examining co-occurrence of H4K20me3 with other histone modifications.

  • Production process: Generated through cloning of antibody genes from immunized rabbits into expression vectors, followed by expression in suspension cells and affinity purification .

In genome-wide chromatin profiling studies, the increased specificity of recombinant monoclonal antibodies has provided more precise mapping of H4K20me3 distribution, particularly at repetitive elements like LINE-1 and LTR retrotransposons, where distinguishing specific signals from background is challenging. Comparative analyses have shown that recombinant monoclonal antibodies yield more consistent peak calling in ChIP-seq experiments targeting H4K20me3, particularly at the boundaries between heterochromatin and euchromatin .

What methodological approaches should be considered when investigating changes in H4K20me3 during cellular differentiation or disease progression?

Investigating dynamic changes in H4K20me3 during cellular differentiation or disease progression requires sophisticated methodological approaches:

  • Temporal sampling strategies:

    • Collect samples at multiple timepoints during differentiation/disease progression

    • Include appropriate controls (undifferentiated cells, healthy tissue, isogenic controls)

    • Consider paired samples when possible (e.g., tumor vs. adjacent normal tissue)

  • Multi-omics integration approaches:

    • Combine ChIP-seq for H4K20me3 with RNA-seq to correlate modification patterns with gene expression

    • Integrate DNA methylation data (e.g., from WGBS or RRBS) to examine relationship between H4K20me3 and DNA methylation

    • Include analysis of H4K20me3 methyltransferases (SUV420H1/H2) and demethylases expression/activity

  • Single-cell technologies:

    • Apply single-cell ChIP-seq or CUT&Tag for heterogeneous populations

    • Consider cellular heterogeneity in interpreting bulk sequencing results

    • Use immunofluorescence to visualize cell-to-cell variation in H4K20me3 patterns

  • Genomic context analysis:

    • Examine H4K20me3 enrichment at specific genomic features (promoters, enhancers, repetitive elements)

    • Focus on regions that show dynamic regulation during differentiation

    • Analyze co-occurrence with other heterochromatin marks (H3K9me3, HP1)

Research has revealed significant changes in H4K20me3 patterns during embryonic stem cell differentiation, with naive pluripotent cells showing global hypomethylation of H4K20me3 compared to differentiated cells . Studies in cancer have identified aberrant H4K20me3 patterns, particularly at retrotransposons and specific gene loci like AWT1, which has been associated with acute myeloid leukemia .

The dynamic nature of H4K20me3 has been observed in multiple biological contexts, including:

  • Mouse oocyte development, where H4K20me3 contributes to the DNA methylation landscape

  • Colorectal cancer, where altered H4K20me3 patterns are associated with cancer stemness via STAT3 activation

  • Epstein-Barr virus-mediated B cell transformation, inducing global chromatin changes including H4K20me3 redistribution

How can researchers troubleshoot non-specific binding or weak signals when using Tri-methyl-Histone H4 (K20) antibodies?

Troubleshooting non-specific binding or weak signals with Tri-methyl-Histone H4 (K20) antibodies requires systematic evaluation of experimental conditions:

For non-specific binding issues:

  • Antibody dilution optimization:

    • Test a dilution series (e.g., 1:500, 1:1000, 1:2000, 1:5000) to identify optimal signal-to-noise ratio

    • For Western blot applications, dilutions from 1:500-1:5000 are recommended depending on sample concentration

    • For immunocytochemistry, more concentrated antibody may be required (1:50-1:500)

  • Blocking optimization:

    • Test different blocking agents (BSA, milk, serum)

    • Increase blocking time or concentration to reduce non-specific binding

    • Consider adding 0.1-0.3% Triton X-100 in blocking buffer for better penetration in ICC/IF

  • Cross-reactivity elimination:

    • Pre-adsorb antibody with unmodified histone peptides

    • Use peptide competition assays to confirm specificity

    • Consider dot blot analysis to verify selective binding to H4K20me3 over other methylation states

For weak signal issues:

  • Sample preparation optimization:

    • Ensure proper extraction of nuclear proteins (for Western blot)

    • Verify efficient fixation that preserves epitope structure but allows antibody access

    • For FFPE tissues, optimize antigen retrieval methods (heat vs. enzymatic)

  • Detection system enhancement:

    • Utilize signal amplification systems (HRP-polymer, TSA, etc.)

    • Extend primary antibody incubation time (overnight at 4°C)

    • Reduce washing stringency without compromising specificity

  • Epitope accessibility improvement:

    • For fixed samples, optimize permeabilization conditions

    • For chromatin applications, ensure adequate chromatin fragmentation

    • Consider native ChIP instead of crosslinked ChIP for some applications

Researchers have successfully detected H4K20me3 in paraffin-embedded tissues using 1:500 dilution following heat-mediated antigen retrieval, and in fixed cells using 1:500 dilution with 4% paraformaldehyde fixation for 15 minutes at room temperature .

What technical considerations are important when using Tri-methyl-Histone H4 (K20) antibodies in chromatin immunoprecipitation (ChIP) studies?

Chromatin immunoprecipitation (ChIP) with Tri-methyl-Histone H4 (K20) antibodies requires specific technical considerations to achieve optimal results:

  • Chromatin preparation:

    • Cross-linking conditions: Standard 1% formaldehyde for 10 minutes at room temperature may be insufficient; consider dual crosslinking with EGS followed by formaldehyde for stable capture of histone-DNA interactions

    • Sonication parameters: Optimize to achieve 200-500bp fragments, with careful monitoring to prevent over-sonication which can destroy epitopes

    • Input quality assessment: Verify fragmentation efficiency by agarose gel electrophoresis before proceeding

  • Immunoprecipitation conditions:

    • Antibody amount: Titrate antibody concentration to determine optimal amount (typically 2-5μg per ChIP reaction)

    • Incubation time: Extended incubation (overnight at 4°C) often yields better results for histone modifications

    • Bead selection: Protein A beads are recommended for rabbit host antibodies

    • Pre-clearing steps: Implement to reduce background from non-specific binding to beads

  • Washing and elution:

    • Washing stringency: Balance between removing non-specific interactions and maintaining specific antibody-epitope binding

    • Number of washes: Typically 4-6 washes with increasing stringency

    • Elution conditions: Optimize temperature and buffer composition for efficient release of chromatin-antibody complexes

  • Controls and validation:

    • Include positive control regions known to be enriched for H4K20me3 (e.g., retrotransposons, silenced genes)

    • Include negative control regions known to lack H4K20me3 (e.g., actively transcribed housekeeping genes)

    • Perform IgG control immunoprecipitations to establish background levels

    • Validate ChIP-seq findings with orthogonal methods (e.g., ChIP-qPCR)

  • Data analysis considerations:

    • Peak calling parameters: Adjust to accommodate broad enrichment patterns typical of H4K20me3

    • Genome alignment: Account for repetitive regions where H4K20me3 is often enriched

    • Data normalization: Consider spike-in controls for quantitative comparisons between samples

Research has successfully utilized anti-H4K20me3 antibodies in ChIP studies to map this modification to specific genomic regions, including LINE-1 and LTR retrotransposons , and to investigate its role in mouse oocyte development and embryonic stem cell differentiation .

How does H4K20me3 distribution change during cellular reprogramming and what methodological approaches can accurately track these changes?

Cellular reprogramming induces significant alterations in the H4K20me3 epigenetic landscape, requiring sophisticated methodological approaches to accurately track these changes:

  • Global H4K20me3 dynamics during reprogramming:

    • Naive pluripotent cells exhibit global hypomethylation of H4K20me3 compared to differentiated cells

    • During reprogramming to pluripotency, H4K20me3 levels decrease globally as cells acquire pluripotent characteristics

    • This decrease occurs heterogeneously across the genome, with certain regions retaining H4K20me3 marks (epigenetic memory)

  • Methodological approaches to track H4K20me3 changes:

    a. Quantitative Western blotting:

    • Enables measurement of global H4K20me3 levels relative to total H4

    • Sample standardization using recombinant histones or synthetic peptides for calibration

    • Densitometric analysis using internal loading controls

    b. High-resolution ChIP-seq:

    • CUT&RUN or CUT&Tag methods for improved signal-to-noise ratio in heterochromatic regions

    • ATAC-seq integration to correlate H4K20me3 changes with chromatin accessibility

    • Spike-in normalization (using Drosophila chromatin) for quantitative comparisons between timepoints

    c. Single-cell approaches:

    • scChIP-seq or scCUT&Tag to capture cell-to-cell heterogeneity during reprogramming

    • Immunofluorescence with quantitative image analysis to assess nuclear distribution patterns

    • Flow cytometry with intracellular staining to quantify H4K20me3 levels across cell populations

  • Key genomic regions for targeted analysis:

    • Retrotransposons and repetitive elements often show the most dramatic changes in H4K20me3

    • Developmental gene promoters gain or lose H4K20me3 during lineage commitment

    • Imprinted regions and other epigenetically regulated loci show characteristic patterns

  • Integration with metabolic analysis:

    • α-ketoglutarate levels influence histone methylation states during pluripotency maintenance

    • Monitoring SAM (S-adenosyl methionine) levels as the methyl donor for methyltransferases

    • Assessing activity of H4K20 methyltransferases (SUV420H1/H2) and potential demethylases

Research has demonstrated that during cellular reprogramming, changes in H4K20me3 distribution correlate with changes in DNA methylation patterns, particularly at developmental gene promoters and enhancers . These changes are not simply passive consequences of reprogramming but actively contribute to the acquisition and maintenance of pluripotency. Intracellular α-ketoglutarate has been shown to maintain embryonic stem cell pluripotency through mechanisms that include regulation of histone methylation marks like H4K20me3 .

What are the critical parameters for optimizing Western blot analysis using Tri-methyl-Histone H4 (K20) antibodies?

Optimizing Western blot analysis for H4K20me3 detection requires attention to several critical parameters:

  • Sample preparation optimization:

    • Extraction method: Use specialized histone extraction protocols with high salt buffers or acid extraction to efficiently isolate histones

    • Nuclear enrichment: Separate nuclear and cytoplasmic fractions to concentrate histone proteins

    • Protein quantification: Accurately determine protein concentration to ensure consistent loading

    • Sample volume: Load 20-30μg of nuclear lysate or 5-10μg of purified histones per lane

  • Gel electrophoresis parameters:

    • Gel percentage: Use high percentage (15-18%) SDS-PAGE gels to resolve the small (11 kDa) histone H4 protein

    • Running conditions: Run at lower voltage (80-100V) to prevent sample overheating

    • Ladder selection: Use protein ladders with appropriate small molecular weight markers

  • Transfer optimization:

    • Transfer method: Semi-dry transfer often works well for small proteins like histones

    • Transfer time: Shorter transfer times (15-30 minutes) prevent small proteins from transferring through the membrane

    • Membrane selection: PVDF membranes with 0.2μm pore size (rather than 0.45μm) are recommended for small proteins

  • Antibody incubation conditions:

    • Blocking conditions: 5% BSA in TBST is often more effective than milk-based blockers

    • Primary antibody dilution: Optimize between 1:500-1:5000 based on antibody source and sample concentration

    • Incubation time: Overnight incubation at 4°C often yields better results than shorter incubations

    • Washing stringency: Multiple washes (4-6 times) with TBST to reduce background

  • Detection system selection:

    • Secondary antibody: Anti-rabbit HRP conjugated antibody at 1:2000-1:5000 dilution

    • Exposure time: Multiple exposures to capture optimal signal without saturation

    • Stripping and reprobing: If needed, verify equal loading with anti-total H4 antibody

Western blot analysis using anti-H4K20me3 antibodies should yield a specific band at approximately 11 kDa, corresponding to histone H4. When performed correctly, the signal should be stronger in nuclear extracts compared to whole cell lysates, reflecting the nuclear localization of histones .

How can researchers design multi-parameter immunofluorescence experiments to correlate H4K20me3 with other chromatin marks?

Designing multi-parameter immunofluorescence experiments to correlate H4K20me3 with other chromatin marks requires careful planning:

  • Antibody selection and validation:

    • Host species diversity: Select primary antibodies raised in different host species to enable simultaneous detection

    • Isotype consideration: When antibodies from the same species are unavoidable, use different isotypes and isotype-specific secondary antibodies

    • Validation: Verify each antibody individually before multiplexing

    • Fluorophore selection: Choose fluorophores with minimal spectral overlap

  • Sample preparation protocols:

    • Fixation method: 4% paraformaldehyde for 15 minutes at room temperature preserves nuclear architecture while maintaining epitope accessibility

    • Permeabilization: 0.1-0.3% Triton X-100 for adequate nuclear permeabilization

    • Antigen retrieval: May be necessary for certain epitopes; optimize conditions for compatible retrieval of all target epitopes

    • Blocking: Use species-appropriate normal sera or BSA to reduce non-specific binding

  • Staining sequence optimization:

    • Sequential staining: Consider sequential rather than simultaneous antibody incubation if epitope masking is a concern

    • Primary antibody cocktails: Prepare carefully with optimal dilutions (e.g., 1:500 for H4K20me3)

    • Washing steps: Include extensive washing between steps to reduce background

    • Nuclear counterstain: Include DAPI or Hoechst 33342 to visualize nuclear morphology

  • Co-localization analysis approach:

    • Image acquisition: Obtain z-stacks to capture the three-dimensional distribution of nuclear marks

    • Resolution considerations: Use confocal microscopy for improved spatial resolution

    • Quantitative metrics: Employ Pearson's correlation coefficient, Manders' overlap coefficient, or intensity correlation analysis

    • Spatial relationship analysis: Consider distance-based measurements between different marks

  • Controls and validation:

    • Single antibody controls: Include samples stained with each antibody individually

    • Secondary-only controls: To assess non-specific binding of secondary antibodies

    • Absorption controls: Pre-incubation with specific peptides to verify specificity

    • Biological controls: Include samples known to express or lack specific marks

In successful multi-parameter experiments, H4K20me3 typically shows a punctate nuclear staining pattern that partially co-localizes with heterochromatin markers like H3K9me3 and HP1. When visualized alongside other markers, H4K20me3 (green) can be effectively contrasted with cytoskeletal markers like alpha-tubulin (red) and nuclear counterstains like Hoechst 33342 (blue) .

What are the most reliable normalization approaches for quantitative analysis of H4K20me3 levels in different experimental conditions?

Reliable quantification of H4K20me3 levels across different experimental conditions requires robust normalization approaches:

  • Western blot normalization strategies:

    • Total histone H4 normalization: Probing the same membrane with anti-total H4 antibody after stripping

    • Loading control selection: Traditional loading controls (β-actin, GAPDH) are inappropriate; use total H3 or H4 instead

    • Recombinant protein standards: Include a standard curve of recombinant H4K20me3 peptides for absolute quantification

    • Densitometric analysis: Use linear range of detection for quantification, avoiding saturated signals

  • ChIP-seq/ChIP-qPCR normalization methods:

    • Input normalization: Express enrichment relative to input chromatin (most common approach)

    • Spike-in normalization: Add exogenous chromatin (e.g., Drosophila) as an internal reference

    • Invariant region normalization: Identify genomic regions with stable H4K20me3 levels across conditions

    • Total H4 ChIP normalization: Perform parallel ChIP with anti-total H4 to control for nucleosome occupancy

  • Immunofluorescence quantification approaches:

    • Intensity normalization: Normalize H4K20me3 signal to DAPI intensity or total H4 staining

    • Nuclear segmentation: Proper segmentation of nuclei for accurate signal quantification

    • Single-cell analysis: Quantify on a per-nucleus basis rather than field averages

    • Reference cells: Include internal control cells in the same field when possible

  • ELISA/multiplex bead-based assay normalization:

    • Standard curve fitting: Generate standard curves using synthetic H4K20me3 peptides

    • Balanced sample loading: Equalize total protein or histone content across samples

    • Technical replicates: Include multiple technical replicates to account for assay variation

    • Inter-assay calibrators: Use common samples across multiple plates/experiments as calibrators

  • Mass spectrometry-based approaches:

    • Isotope-labeled internal standards: Use synthetic isotope-labeled peptides corresponding to modified and unmodified forms

    • Retention time normalization: Account for chromatographic variation between runs

    • Modified/unmodified ratio: Express results as ratio of modified to unmodified peptide

    • Total histone normalization: Normalize to total histone levels

For accurate comparison between experimental conditions (e.g., disease vs. healthy, treated vs. untreated), consistent sample processing is critical. Studies comparing H4K20me3 levels between embryonic stem cells and differentiated cells have employed spike-in normalization approaches to account for global differences in modification levels .

How does the distribution of H4K20me3 differ between normal cells and cancer cells, and what methodological approaches best capture these differences?

The distribution of H4K20me3 shows distinctive patterns between normal and cancer cells, with significant implications for genome stability and gene expression:

  • General patterns of H4K20me3 alterations in cancer:

    • Global reduction: Many cancers show global loss of H4K20me3 compared to normal tissues

    • Locus-specific changes: Despite global reduction, certain genomic regions may show increased H4K20me3

    • Heterochromatin disruption: Alterations in H4K20me3 distribution contribute to heterochromatin instability

    • Retrotransposon dysregulation: Loss of H4K20me3 at repetitive elements may lead to their reactivation

  • Cancer-specific H4K20me3 distribution patterns:

    • Colorectal cancer: H4K20me3 redistribution associated with cancer stemness via STAT3 activation and DOT1L induction

    • Hematological malignancies: Hypermethylation of the alternative AWT1 promoter serves as a specific marker for acute myeloid leukemias

    • Epstein-Barr virus-mediated transformation: Induces global chromatin changes including H4K20me3 redistribution in B cells

  • Methodological approaches for comparative analysis:

    • Tissue microarray immunohistochemistry: Enables high-throughput comparison across multiple samples with 1:500 antibody dilution

    • Laser capture microdissection: Isolates specific cell populations from heterogeneous tissues for focused analysis

    • Cell type-specific ChIP-seq: Accounts for cellular heterogeneity in normal tissues and tumors

    • Single-cell approaches: Capture cell-to-cell variation in H4K20me3 distribution within tumors

    • Integrated multi-omics: Correlate H4K20me3 changes with gene expression, DNA methylation, and genetic alterations

  • Technical considerations for cancer studies:

    • Matched normal-tumor pairs: Essential for direct comparison and identification of cancer-specific changes

    • Tumor heterogeneity: Account for intratumoral heterogeneity in sampling and analysis

    • Fixation artifacts: Standardize fixation protocols to minimize variation in antibody performance

    • Quantitative analysis: Develop scoring systems that capture both intensity and distribution patterns

Research has demonstrated that in colorectal cancer, IL-22(+)CD4(+) T cells promote cancer stemness through STAT3 activation and induction of the methyltransferase DOT1L, affecting histone methylation patterns including H4K20me3 . These findings highlight the complex interplay between immune cells, signaling pathways, and epigenetic modifications in cancer progression.

What is the role of H4K20me3 in genome stability, and how can researchers design experiments to investigate this relationship?

H4K20me3 plays a critical role in maintaining genome stability through multiple mechanisms:

  • Established functions of H4K20me3 in genome stability:

    • Heterochromatin maintenance: Stabilizes heterochromatic regions, preventing inappropriate transcription

    • Repetitive element silencing: Suppresses potentially mutagenic retrotransposon activity

    • DNA damage response: Serves as a recognition site for DNA repair proteins

    • Chromosome segregation: Contributes to proper centromere and telomere function

    • Replication timing: Influences the temporal program of DNA replication

  • Experimental approaches to investigate H4K20me3-genome stability relationships:

    a. Genetic manipulation studies:

    • Methyltransferase modulation: SUV420H1/H2 knockdown/knockout to reduce H4K20me3 levels

    • Demethylase overexpression: Force removal of H4K20me3 marks

    • Reader protein manipulation: Disrupt proteins that recognize and bind H4K20me3

    • Assessment metrics: Chromosomal aberrations, micronuclei formation, DNA damage markers

    b. DNA damage response analysis:

    • Double-strand break induction: Use of ionizing radiation or radiomimetic drugs

    • ChIP-seq after damage: Track redistribution of H4K20me3 following DNA damage

    • Co-localization studies: Visualize relationship between H4K20me3 and repair factors (53BP1, BRCA1)

    • Repair kinetics measurement: Assess repair efficiency in cells with altered H4K20me3 levels

    c. Replication stress studies:

    • Replication inhibitors: Aphidicolin or hydroxyurea treatment

    • Pulse-chase labeling: EdU/BrdU incorporation to track replication timing

    • Fork progression analysis: DNA fiber assays in cells with altered H4K20me3

    • Origin activation studies: Measure origin firing in regions with different H4K20me3 levels

  • Technological approaches for mechanistic insights:

    • Proximity ligation assays: Detect physical interactions between H4K20me3 and DNA repair proteins

    • CRISPR-based epigenome editing: Targeted addition or removal of H4K20me3 at specific loci

    • Live-cell imaging: Track dynamics of H4K20me3 and repair factors using fluorescent reporters

    • Genomic scarring analysis: Assess mutation signatures associated with H4K20me3 alterations

  • Integrated analysis frameworks:

    • Multi-scale temporal analysis: Examine immediate, intermediate, and long-term consequences of H4K20me3 alterations

    • Spatial nuclear organization: Correlate H4K20me3 distribution with 3D genome architecture

    • Cell cycle specificity: Determine cell cycle-dependent functions of H4K20me3

    • Tissue-specific variations: Compare H4K20me3-genome stability relationships across different cell types

Studies have shown that H4K20me3 plays a role in maintaining heterochromatin integrity, particularly at repetitive elements like LINE-1 and LTR retrotransposons, which are potential sources of genomic instability when derepressed . Additionally, research has demonstrated connections between aberrant H4K20me3 patterns and genomic instability in cancer contexts .

How does H4K20me3 cooperate with other histone modifications and chromatin regulators to establish repressive chromatin domains?

H4K20me3 functions within a complex network of histone modifications and chromatin regulators to establish and maintain repressive chromatin domains:

  • Co-occurrence with other repressive histone marks:

    • H3K9me3 coordination: H4K20me3 frequently co-occurs with H3K9me3 at constitutive heterochromatin

    • H3K27me3 relationships: More complex association with this Polycomb-associated mark at facultative heterochromatin

    • DNA methylation correlation: Strong association between H4K20me3 and DNA methylation, particularly at repetitive elements and silenced genes

    • Histone deacetylation: Inverse relationship with active marks like H3K9ac and H4K16ac

  • Interaction with chromatin regulator proteins:

    • HP1 proteins: Recognize and bind H3K9me3, stabilizing heterochromatin in regions also enriched for H4K20me3

    • 53BP1 binding: DNA damage response protein that recognizes H4K20me2/3 during double-strand break repair

    • L3MBTL1 interactions: Methyl-lysine reader protein that compacts chromatin through binding to H4K20me3

    • SUV420H1/H2 recruitment: H4K20 methyltransferases that are recruited by HP1, creating a feedback loop

  • Stepwise assembly of repressive domains:

    • Nucleation: Initial targeting of SUV39H1/H2 to specific sequences leads to H3K9me3 deposition

    • Spreading: HP1 proteins bind H3K9me3 and recruit SUV420H1/H2, which establish H4K20me3

    • Reinforcement: H4K20me3 stabilizes the repressive state and helps recruit additional silencing factors

    • Maintenance: Through DNA replication and cell division via interactions with PCNA and CAF-1

  • Experimental approaches to study cooperative interactions:

    • Sequential ChIP (Re-ChIP): Identify genomic regions containing both H4K20me3 and other modifications

    • Proximity ligation assay: Detect physical proximity between differently modified nucleosomes

    • Biochemical reconstitution: In vitro assembly of modified nucleosomes to study higher-order structures

    • Genetic pathway dissection: Systematic perturbation of writers, readers, and erasers of different marks

  • Context-dependent functional outcomes:

    • Developmental contexts: Dynamic interactions during cellular differentiation when chromatin states are established

    • Cell cycle regulation: Fluctuations in the relationships between marks through the cell cycle

    • Stress responses: Altered interactions following cellular stresses that impact chromatin organization

    • Disease states: Disrupted cooperation in cancers and other diseases with epigenetic dysregulation

Research has revealed that during cellular differentiation and development, the establishment of H4K20me3 often follows H3K9me3 deposition, with both marks contributing to stable silencing of developmental genes and repetitive elements . In the context of embryonic stem cells, the maintenance of pluripotency involves precise regulation of these repressive marks, with naive pluripotent cells showing global hypomethylation of H4K20me3 .

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.