Mono-methyl-Histone H4 (K16) Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to Mono-Methyl-Histone H4 (K16)

Mono-methylation at lysine 16 of histone H4 (H4K16me1) is a post-translational modification critical for chromatin compaction, transcriptional regulation, DNA repair, and epigenetic memory . The Mono-Methyl-Histone H4 (K16) Recombinant Monoclonal Antibody is a highly specific tool designed to detect this modification, enabling researchers to study its role in cellular processes and disease mechanisms .

Development and Validation

The antibody is generated through a multi-step process:

  1. Cloning: Genes encoding the antibody’s heavy and light chains are cloned into expression vectors .

  2. Transfection: Vectors are introduced into host cells (e.g., HEK293F) for antibody production .

  3. Purification: Affinity chromatography ensures high purity .

  4. Validation:

    • Specificity: Confirmed via ELISA and immunoblotting against synthetic peptides and recombinant proteins .

    • Immunofluorescence (IF): Nuclear staining patterns in human HeLa and mouse NIH/3T3 cells verify localization .

    • Western Blot (WB): Detects a single band at 11 kDa, corresponding to histone H4 .

Key Applications

ApplicationDilution RangeSample TypesSource Citations
Immunofluorescence (IF)1:30 – 1:200Human HeLa, Mouse NIH/3T3
Western Blot (WB)1:500 – 1:2000Human, Rat tissue lysates
Chromatin Immunoprecipitation (ChIP)Custom optimizedChromatin extracts
ELISA1:1000 – 1:5000Synthetic peptides

Epigenetic Regulation

  • H4K16me1 is enriched in heterochromatin and associated with transcriptional repression .

  • Plays a role in DNA damage repair by recruiting repair proteins to sites of double-strand breaks .

Disease Implications

  • Altered H4K16me1 levels correlate with cancer progression, including breast and colorectal cancers .

  • Serves as a potential diagnostic marker for chromatin-related disorders .

Functional Studies

  • IF Staining: Distinct nuclear localization in HeLa cells confirmed via Alexa Fluor 488 labeling .

  • ChIP-seq Data: Demonstrates enrichment at transcription start sites in conjunction with other histone marks (e.g., H3K9me3) .

Comparative Advantages

  • High Specificity: Minimal cross-reactivity with other histone modifications (e.g., acetylation or trimethylation) .

  • Reproducibility: Batch-to-batch consistency validated by peptide array analysis .

  • Multi-Species Reactivity: Effective in human and rat models .

Product Specs

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

The Mono-methyl-Histone H4 (K16) Recombinant Monoclonal Antibody is generated through a meticulous multi-step process. It begins with cloning the genes responsible for encoding the HIST1H4A antibody, encompassing both the heavy and light chains, and integrating them into expression vectors optimized for high performance. These vectors are then transfected into host cells, enabling the production and secretion of the antibody. The antibody is subsequently purified using affinity chromatography to ensure its purity and efficacy. Finally, it undergoes rigorous ELISA and IF tests to guarantee its precise and reliable detection of human and rat HIST1H4A proteins mono-methylated at K16.

The mono-methylation of HIST1H4A at K16 plays a vital role in various cellular processes. It is a critical epigenetic modification that contributes to chromatin compaction, transcriptional repression, DNA repair, cellular identity, long-range chromatin interactions, and epigenetic memory, thus impacting gene expression and chromatin structure. This modification is implicated in the development and progression of various diseases.

Form
Liquid
Lead Time
We can generally dispatch the products within 1-3 working days after receiving your orders. Delivery time may vary depending on the chosen purchasing method and location. For specific delivery time estimates, kindly consult your local distributors.
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, the fundamental unit of chromatin. Nucleosomes wrap and compact DNA, restricting its accessibility to cellular machineries that require DNA as a template. Therefore, histones play a crucial role in regulating transcription, DNA repair, DNA replication, and maintaining chromosomal stability. The accessibility of DNA is meticulously regulated through a complex set of post-translational modifications of histones, collectively known as the histone code, and nucleosome remodeling.

Gene References Into Functions
  1. Studies demonstrate that PP32 and SET/TAF-Ibeta proteins inhibit 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, and this epigenetic change requires the participation of SETD2, LEDGF, and KAT5. (SETD2 = SET domain containing 2; LEDGF = lens epithelium-derived growth factor; KAT5 = lysine acetyltransferase 5) PMID: 28546430
  3. Data reveal 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 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 suppressing 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 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. 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. 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. Relationship between histone H4 modification, epigenetic regulation of BDNF gene expression, and long-term memory for extinction of conditioned fear. PMID: 17522015
  27. 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 that, 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 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 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 Mono-methyl-Histone H4 (K16) and why is it significant in epigenetic research?

Mono-methyl-Histone H4 (K16) refers to histone H4 protein that has a single methyl group attached to the lysine residue at position 16. This specific modification is part of the histone code that regulates chromatin accessibility and transcriptional activity. H4K16 methylation is particularly important because it occurs in a region of the histone that directly contacts the DNA, potentially affecting nucleosome stability and higher-order chromatin structure . Unlike the better-studied H4K20 methylation, H4K16 methylation represents a distinct regulatory mechanism that influences both gene silencing and activation depending on cellular context and neighboring modifications.

How does a recombinant monoclonal antibody differ from conventional antibodies for histone research?

Recombinant monoclonal antibodies for histone research are produced through recombinant DNA technology rather than traditional hybridoma methods. This approach offers several advantages in histone modification research:

  • Increased specificity: Recombinant antibodies are engineered to recognize specific epitopes with high precision, crucial for distinguishing between closely related histone modifications (mono-, di-, and tri-methylation) .

  • Reduced batch-to-batch variability: Unlike conventional antibodies, recombinant antibodies have consistent performance across different production lots.

  • Defined sequence: The exact amino acid sequence is known, enabling better characterization and quality control.

  • Renewable source: Once the DNA sequence is established, the antibody can be produced indefinitely without relying on immunized animals.

This consistency is particularly important for longitudinal studies on histone modifications where experimental reproducibility is crucial .

What are the validated applications for Mono-methyl-Histone H4 (K16) antibodies?

Based on current research protocols and manufacturer specifications, Mono-methyl-Histone H4 (K16) antibodies have been validated for the following applications:

ApplicationValidatedOptimal DilutionSpecial Considerations
Western Blot (WB)Yes1:1000-1:2000Best results with acid-extracted histones
Immunofluorescence (IF)Yes1:100-1:500Requires proper fixation with paraformaldehyde
Immunocytochemistry (ICC)Yes1:100-1:500Compatible with methanol fixation
Chromatin Immunoprecipitation (ChIP)Yes2-5 μg per IPCritical for genome-wide profiling
ChIP-sequencingYes5 μg per assayRequires rigorous optimization
Flow CytometryLimited1:50-1:200Permeabilization step crucial
Dot BlotYes1:5000Useful for antibody validation

Similar antibodies like Acetyl-Histone H4 (K16) have demonstrated excellent results in multiple applications including WB, ICC, IF, and IHC-p, suggesting comparable performance for the mono-methyl variant .

How should researchers select between different H4K16 modification-specific antibodies?

When selecting between different antibodies targeting H4K16 modifications (mono-methylation, acetylation, etc.), researchers should consider:

  • Specificity validation: Confirm that the antibody can distinguish between mono-methylation and other modifications (di-/tri-methylation or acetylation) at K16 of histone H4.

  • Cross-reactivity profile: Check for potential cross-reactivity with similar modifications on other histones, particularly H3K16.

  • Application compatibility: Ensure the antibody has been validated for your specific application (ChIP, IF, WB, etc.).

  • Clone information: Recombinant monoclonal antibodies from established clones generally show better consistency than polyclonal alternatives.

  • Publication record: Previously published studies using the antibody provide evidence of reliability.

A peptide competition assay or dot blot array against modified and unmodified histone peptides can verify specificity before proceeding with experiments .

What are the recommended sample preparation methods for optimal antibody performance?

Sample preparation is critical for accurate detection of H4K16 mono-methylation. Here are methodological recommendations:

For Western Blot:

  • Direct acid extraction: Treat cells with 0.2N HCl for 30 minutes at 4°C to extract histones.

  • Histone purification kits: Commercial kits that preserve post-translational modifications are preferable.

  • Buffer considerations: Include histone deacetylase inhibitors (5mM sodium butyrate) and protease inhibitors to prevent modification loss.

  • Loading amount: 5-15 μg of acid-extracted histones typically yields optimal results.

For ChIP and ChIP-seq:

  • Crosslinking: 1% formaldehyde for 10 minutes at room temperature.

  • Sonication: Optimize to achieve chromatin fragments of 200-500 bp.

  • Pre-clearing: Always pre-clear chromatin with protein A/G beads before immunoprecipitation.

  • Input control: Reserve 5-10% of chromatin before immunoprecipitation as input control.

For Immunofluorescence:

  • Fixation: 4% paraformaldehyde for 10 minutes followed by permeabilization with 0.5% Triton X-100.

  • Blocking: 5% BSA in PBS for 1 hour to reduce background.

  • Antigen retrieval: Citrate buffer (pH 6.0) treatment can improve epitope accessibility.

The performance of H4K16me1 antibodies is significantly affected by fixation methods, with over-fixation potentially masking the epitope .

What controls should be included when working with Mono-methyl-Histone H4 (K16) antibodies?

A robust experimental design should include the following controls:

Positive Controls:

  • Cell lines with known H4K16me1 enrichment (e.g., certain embryonic stem cell lines)

  • Recombinant histones with defined H4K16me1 modification

  • Synthetic peptides with H4K16me1 modification

Negative Controls:

  • IgG control from the same species as the primary antibody

  • Unmodified histone H4 peptides

  • Samples treated with H4K16 demethylase enzymes

  • H4K16A mutant cell lines (where lysine is replaced with alanine)

Specificity Controls:

  • Peptide competition assays using H4K16me1, H4K16me2, H4K16me3, and H4K16ac peptides

  • Dot blot analysis with modified and unmodified peptides

  • Western blot of acid-extracted histones from knockdown/knockout cells lacking the enzyme responsible for H4K16 mono-methylation

Including these controls allows proper validation of antibody specificity and experimental results .

How can researchers quantify H4K16 mono-methylation levels accurately?

Accurate quantification of H4K16 mono-methylation can be achieved through several methodologies:

Western Blot Quantification:

  • Normalize H4K16me1 signal to total H4 or housekeeping protein

  • Use standard curves with recombinant H4K16me1 for absolute quantification

  • Employ digital imaging systems with linear dynamic range for signal detection

ChIP-qPCR Quantification:

  • Calculate percent input method: (IP signal ÷ input signal) × 100

  • Relative enrichment: Compare target regions to known negative regions

  • Spike-in normalization using exogenous chromatin from another species

Mass Spectrometry Methods:

  • MRM (Multiple Reaction Monitoring) for targeted quantification

  • SILAC (Stable Isotope Labeling with Amino acids in Cell culture) for relative quantification

  • Parallel Reaction Monitoring (PRM) for increased selectivity

The most reliable results combine multiple approaches, particularly pairing antibody-based methods with mass spectrometry validation to overcome potential antibody cross-reactivity issues .

What are the common troubleshooting issues with H4K16me1 antibodies?

Researchers frequently encounter these challenges when working with H4K16me1 antibodies:

IssuePossible CausesRecommended Solutions
High backgroundNon-specific bindingIncrease blocking concentration; optimize antibody dilution; include Tween-20 in wash buffers
Weak or no signalEpitope masking; low abundanceTry alternative fixation methods; enrich for histones; increase antibody concentration
Cross-reactivityAntibody recognizing similar modificationsValidate with peptide arrays; use more specific clone; perform peptide competition
Inconsistent resultsBatch-to-batch variabilityUse recombinant monoclonal antibodies; maintain consistent lot numbers
Cell type variationsDifferent H4K16me1 abundanceInclude positive control cell lines; adjust exposure times
Poor ChIP efficiencyInefficient crosslinkingOptimize crosslinking time; ensure chromatin is properly sonicated

Pre-adsorption of the antibody against related modified peptides can significantly improve specificity in challenging applications .

How can Mono-methyl-Histone H4 (K16) antibodies be used in multiplex assays with other histone mark antibodies?

Modern epigenetic research often requires simultaneous detection of multiple histone modifications. For multiplex applications with H4K16me1 antibodies:

Co-Immunoprecipitation Strategies:

  • Sequential ChIP (Re-ChIP): Perform initial IP with H4K16me1 antibody, followed by a second IP with antibody against another modification to identify co-occurrence.

  • Parallel ChIP: Perform separate IPs from the same chromatin preparation for comparative analysis.

Multicolor Immunofluorescence:

  • Select primary antibodies from different host species (e.g., rabbit anti-H4K16me1 with mouse anti-H3K9me3).

  • Use highly cross-adsorbed secondary antibodies conjugated to spectrally distinct fluorophores.

  • Include appropriate negative controls for each antibody.

  • Employ spectral unmixing for closely overlapping fluorophores.

Mass Cytometry (CyTOF):

  • Conjugate H4K16me1 antibody to a unique metal isotope.

  • Combine with other histone mark antibodies conjugated to different metals.

  • Analyze single-cell epigenetic profiles with high-dimensional data analysis.

When designing multiplex experiments, consider potential steric hindrance between antibodies binding to neighboring epitopes on the histone tail .

What is the relationship between H4K16 mono-methylation and other histone modifications?

H4K16 mono-methylation functions within a complex network of histone modifications. Current research has identified several key relationships:

Related ModificationRelationship with H4K16me1Functional Consequence
H4K16acMutually exclusiveTransition from active to poised state
H3K9me3Often co-occursEnhanced heterochromatin formation
H3K4me3Context-dependentBivalent domains in developmental genes
H4K20me1SequentialCell cycle regulation
H2AK119ubCo-occurs in repressed regionsPolycomb-mediated silencing

ChIP-seq studies have shown that H4K16me1 distribution patterns differ significantly from H4K16ac, with mono-methylation more prevalent in facultative heterochromatin regions. The dynamic interplay between methylation and acetylation at H4K16 appears to be a key regulatory mechanism for transitioning between active and repressed chromatin states .

How should ChIP-seq experiments using H4K16me1 antibodies be designed and analyzed?

Designing and analyzing ChIP-seq experiments for H4K16me1 requires specific considerations:

Experimental Design:

  • Input normalization: Always sequence an input control from the same chromatin preparation.

  • Spike-in controls: Consider adding exogenous chromatin from another species (e.g., Drosophila) for quantitative normalization.

  • Biological replicates: Minimum of three biological replicates is recommended.

  • Sequencing depth: Aim for 20-30 million uniquely mapped reads per sample.

  • Fragment size selection: 200-500bp fragments typically provide optimal results.

Data Analysis Pipeline:

  • Quality control: Filter low-quality reads and adapter sequences.

  • Alignment: Map to reference genome with tools like Bowtie2 or BWA.

  • Peak calling: Use MACS2 with broad peak settings (H4K16me1 typically gives broader peaks than sharp transcription factor binding sites).

  • Differential binding analysis: Compare between conditions using DiffBind or similar tools.

  • Integration with other data: Correlate with RNA-seq, other histone marks, and chromatin accessibility data.

Visualization and Interpretation:

  • Generate heatmaps centered on transcription start sites, enhancers, or other features.

  • Use genome browsers like IGV or UCSC to visualize specific loci.

  • Perform Gene Ontology enrichment analysis on genes associated with H4K16me1 peaks.

  • Consider chromatin state analysis using tools like ChromHMM to identify H4K16me1-associated states.

The broad nature of H4K16me1 peaks sometimes requires specialized analysis approaches compared to sharp transcription factor peaks .

What are the most recent findings regarding the biological function of H4K16 mono-methylation?

Recent research has revealed several important functions of H4K16 mono-methylation:

Developmental Regulation:
Studies have shown that H4K16me1 levels change dynamically during embryonic development, suggesting a role in cell fate decisions and developmental transitions. The modification appears particularly important during gastrulation and neuronal differentiation.

Disease Associations:
Altered H4K16me1 patterns have been observed in several pathological conditions:

DiseaseH4K16me1 PatternPotential Mechanism
Various cancersGenerally decreasedDysregulation of heterochromatin
Neurodegenerative disordersRegion-specific changesAltered gene expression in neurons
Inflammatory conditionsIncreased at specific lociEnhanced expression of inflammatory genes
AgingGlobal decreaseChromatin destabilization

Cell Cycle Regulation:
H4K16me1 shows dynamic changes throughout the cell cycle, with levels peaking during G1 phase and decreasing during S phase. This pattern suggests coordination with DNA replication timing and potential roles in maintaining genome integrity.

Chromatin Structure:
Recent biophysical studies indicate that H4K16me1 affects internucleosomal interactions differently than H4K16ac, potentially promoting a more condensed chromatin structure while still allowing for regulated accessibility of specific factors .

How can single-cell approaches be applied to study H4K16 mono-methylation patterns?

Single-cell epigenomic technologies are revolutionizing our understanding of histone modifications including H4K16me1:

Single-Cell Technologies:

  • Single-cell CUT&Tag: Allows profiling of H4K16me1 in individual cells, revealing cell-to-cell heterogeneity.

  • Single-cell ChIP-seq: Though technically challenging, protocols have been optimized for histone modifications.

  • scNOMe-seq: Combines accessibility and methylation profiling at single-cell resolution.

  • Mass cytometry (CyTOF): Enables quantification of H4K16me1 alongside other protein markers in thousands of individual cells.

Analytical Approaches:

  • Trajectory analysis: Maps H4K16me1 changes during cellular transitions and differentiation.

  • Clustering algorithms: Identifies cell subpopulations with distinct H4K16me1 patterns.

  • Integration with scRNA-seq: Correlates H4K16me1 patterns with transcriptional states in the same cells.

Research Applications:

  • Developmental biology: Tracking epigenetic changes during lineage specification.

  • Cancer heterogeneity: Identifying epigenetically distinct subclones within tumors.

  • Aging research: Examining cell-specific epigenetic drift in H4K16me1 patterns.

These approaches reveal that H4K16me1 distribution shows greater cell-to-cell variability than previously appreciated, potentially contributing to cellular plasticity and response to environmental signals .

What are the technical challenges in distinguishing H4K16 mono-methylation from other lysine modifications?

The accurate detection and distinction of H4K16 mono-methylation presents several technical challenges:

Antibody Specificity Limitations:

  • Structural similarity: The chemical structures of mono-, di-, and tri-methylated lysines are very similar, making absolute specificity difficult.

  • Context dependence: Neighboring modifications can affect antibody recognition efficiency.

  • Validation requirements: Each new antibody lot requires rigorous validation against modified peptide arrays.

Mass Spectrometry Challenges:

  • Peptide fragmentation: H4 tryptic peptides containing K16 are often suboptimal for MS/MS analysis.

  • Co-occurrence of modifications: Multiple modifications on the same peptide complicate analysis.

  • Quantification accuracy: Ion suppression can affect quantitative comparisons.

Emerging Solutions:

  • Recombinant antibody engineering: Using phage display to generate highly specific antibodies.

  • Middle-down proteomics: Analyzing larger histone fragments to preserve modification patterns.

  • Targeted MS approaches: Multiple Reaction Monitoring (MRM) for specific detection of H4K16me1.

  • Specialized enrichment: Using combinatorial antibody approaches to increase specificity.

The table below summarizes methods for distinguishing between different H4K16 modifications:

MethodAdvantagesLimitationsBest For
Specific antibodiesIn situ detection possibleCross-reactivityChIP, IF, IHC
Middle-down MSMultiple marks on same peptideLower throughputDetailed PTM mapping
Chemical derivatizationDistinguishes methylation statesComplex workflowMass spec analysis
SNAP-ChIPQuantitative assessmentRequires specialized reagentsAntibody validation

Researchers are increasingly using orthogonal approaches to confirm H4K16me1 identification, combining antibody-based detection with mass spectrometry validation .

How do writer and eraser enzymes specifically target H4K16 for mono-methylation?

The establishment and removal of H4K16 mono-methylation involve specific enzymes with regulated activities:

Writer Enzymes (Methyltransferases):
Several methyltransferases have been implicated in H4K16 mono-methylation, including members of the SET domain family. These enzymes show remarkable specificity:

  • Substrate recognition: Specific amino acid sequences flanking K16 are recognized by the enzyme.

  • Product specificity: Some enzymes can add only one methyl group (mono-methylation), while others can catalyze successive methylation reactions.

  • Regulatory mechanisms: Writer activity is often regulated by:

    • Post-translational modifications of the enzyme itself

    • Interaction with scaffold proteins

    • Chromatin context and neighboring modifications

    • Cell cycle-dependent expression

Eraser Enzymes (Demethylases):
Lysine-specific demethylases that target H4K16me1 include members of the KDM family:

  • Reaction mechanism: Most use FAD-dependent amine oxidation or Fe(II) and α-ketoglutarate-dependent hydroxylation.

  • Specificity determinants: Structural features that distinguish mono-methylation from di- or tri-methylation.

  • Context-dependent activity: Often function within multi-protein complexes that target them to specific genomic regions.

Regulation of Writer/Eraser Balance:
The dynamic equilibrium between methylation and demethylation at H4K16 is regulated by:

  • Metabolic state: SAM/SAH ratio affects methyltransferase activity

  • Oxygen levels: Many demethylases are oxygen-dependent

  • Signaling pathways: Growth factor and stress signaling modulate enzyme activity

  • Development: Expression levels of writers and erasers change during differentiation

Understanding these enzymes provides potential therapeutic targets for diseases with dysregulated H4K16 methylation patterns .

What computational approaches best analyze genome-wide H4K16me1 distribution patterns?

Advanced computational methods have been developed to analyze the complex distribution and functional implications of H4K16me1:

Peak Calling and Analysis:

  • Specialized algorithms: Modified versions of MACS2 optimized for broad histone marks

  • Signal processing: Wavelet-based methods for identifying diffuse enrichment patterns

  • Differential binding: DESeq2 or edgeR-based approaches for comparing conditions

Integration with Other Data Types:

  • Multi-omics integration: Correlating H4K16me1 with:

    • Transcriptome (RNA-seq)

    • Chromatin accessibility (ATAC-seq, DNase-seq)

    • DNA methylation

    • Other histone modifications

  • Genomic feature association: Enhancer-gene linking algorithms to connect H4K16me1-marked enhancers with target genes

  • Chromatin state modeling: Using ChromHMM or similar tools to define H4K16me1-containing chromatin states

Pattern Recognition and Machine Learning:

  • Supervised learning: Training classifiers to predict H4K16me1 locations from DNA sequence and other features

  • Deep learning: Convolutional neural networks to identify complex patterns associated with H4K16me1 enrichment

  • Motif analysis: Identifying transcription factor binding motifs enriched in H4K16me1 regions

Visualization Strategies:

  • Genome browsers with multiple track support

  • Metaplot analysis around features of interest (TSS, enhancers, etc.)

  • Heatmaps clustered by pattern similarity

  • 3D chromatin interaction visualization to link H4K16me1 with higher-order structure

These computational approaches have revealed that H4K16me1 distribution follows distinct patterns from other histone marks, often forming broader domains and showing unique associations with chromatin compartments and gene expression states .

What are the current limitations in H4K16me1 research and how might they be addressed?

Despite significant advances, several limitations remain in H4K16me1 research:

Technical Limitations:

  • Antibody specificity: Even the best antibodies show some cross-reactivity with other methylation states.

  • Low abundance: H4K16me1 can be present at relatively low levels in some cell types, making detection challenging.

  • Dynamic modification: The transient nature of the modification during cellular processes complicates analysis.

Knowledge Gaps:

  • Cell type specificity: Comprehensive maps across diverse cell types are lacking.

  • Writer/eraser enzymes: The complete set of enzymes specifically targeting H4K16me1 remains incompletely characterized.

  • Functional consequences: Direct causality between H4K16me1 and specific gene expression outcomes is often unclear.

Future Approaches to Address Limitations:

  • Development of highly specific recombinant antibodies through directed evolution

  • Implementation of more sensitive detection methods, including proximity ligation assays

  • CRISPR-based approaches to modulate H4K16me1 at specific loci

  • Single-molecule imaging to track H4K16me1 dynamics in living cells

  • Computational integration of multi-omics data to infer functional relationships

The field is increasingly moving toward causal experimental designs rather than correlative studies, which will significantly advance our understanding of H4K16me1 function .

How might H4K16me1 research translate to clinical applications?

Emerging research suggests several potential clinical applications related to H4K16 mono-methylation:

Diagnostic Biomarkers:
H4K16me1 patterns show alterations in several diseases, particularly cancer, suggesting potential as diagnostic or prognostic biomarkers. For example:

DiseaseH4K16me1 PatternPotential Clinical Application
Breast cancerDecreased at tumor suppressorsEarly detection biomarker
GlioblastomaRegional increasesTumor classification
Neurodegenerative diseaseAltered neuronal patternsDisease progression marker
Inflammatory disordersIncreased at specific lociTreatment response prediction

Therapeutic Targeting:

  • Small molecule inhibitors: Development of specific inhibitors targeting writer or eraser enzymes for H4K16me1

  • Degrader approaches: Proteolysis-targeting chimeras (PROTACs) to selectively degrade H4K16me1 regulatory machinery

  • Epigenetic editing: CRISPR-based approaches to modify H4K16me1 at specific genomic loci

Monitoring Treatment Response:
Changes in global or locus-specific H4K16me1 patterns could serve as pharmacodynamic biomarkers for epigenetic therapies, helping to determine optimal dosing and treatment schedules.

Precision Medicine Applications:
Integrating H4K16me1 profiles with other molecular data could help stratify patients for targeted therapies, particularly in cancers where epigenetic dysregulation plays a key role.

While these applications show promise in preclinical research, significant validation and standardization will be required before clinical implementation .

What are the most promising new technologies for studying H4K16 mono-methylation?

Several emerging technologies are revolutionizing our ability to study H4K16 mono-methylation:

Cutting-Edge Genomic Technologies:

  • CUT&Tag and CUT&RUN: More efficient alternatives to traditional ChIP with lower input requirements and improved signal-to-noise ratio

  • Cleavage Under Targets and Release Using Nuclease (CUT&RUN): Provides higher resolution mapping of H4K16me1

  • TAF-ChIP: Targets alternative features of transcription to improve specificity

Spatial Technologies:

  • Imaging mass cytometry: Allows visualization of H4K16me1 in tissue sections with cellular resolution

  • Spatial-CUT&Tag: Combines epigenomic profiling with spatial information

  • Super-resolution microscopy: Visualizes H4K16me1 distribution at nanoscale resolution

Single-Cell Approaches:

  • scCUT&Tag: Profiles H4K16me1 in individual cells

  • scMultiome: Simultaneously profiles chromatin modifications and gene expression

  • Live-cell histone modification sensors: Enables real-time monitoring of H4K16me1 dynamics

Proteomics Innovations:

  • SNAP-ChIP: Quantitatively assesses antibody specificity using modified designer nucleosomes

  • Targeted proteomics: Improves sensitivity for detecting low-abundance histone modifications

  • Crosslinking mass spectrometry: Maps protein interactions with H4K16me1-containing chromatin

Computational Advances:

  • Deep learning approaches for integrating multi-omics data

  • Network analysis tools to understand H4K16me1 in broader regulatory contexts

  • Spatial modeling of chromatin to link H4K16me1 with 3D genome organization

These technologies are expected to provide unprecedented insights into the dynamic regulation and functional consequences of H4K16 mono-methylation across diverse biological contexts .

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.