Phospho-HIST1H3A (Y41) Antibody is a specialized immunological reagent designed to specifically recognize and bind to histone H3 that has been phosphorylated at tyrosine 41 (H3Y41ph). Histone H3 contains three highly conserved tyrosine residues, with Y41 positioned at the N-terminus of the first helix of H3 (the αN1-helix) where DNA enters the nucleosome, juxtaposed to the major groove of the DNA double helix . This strategic position highlights the potential significance of this modification in regulating chromatin structure and function.
The development of specific antibodies against H3Y41ph has enabled researchers to investigate this particular histone modification in various cellular contexts. These antibodies are typically generated by immunizing animals with synthetic phosphopeptides corresponding to the region surrounding Y41 of histone H3 . Their specificity is rigorously verified through various control experiments, including peptide competition assays and tests with mutant histones (such as Y41F mutants) to ensure they exclusively recognize the phosphorylated form of H3Y41 .
A groundbreaking discovery revealed that JAK2, previously known primarily for its cytoplasmic signaling role, functions in the nucleus to directly phosphorylate H3Y41 . This phosphorylation event represents a novel nuclear function for JAK2 outside its established involvement in cytoplasmic signaling cascades. Experimental evidence demonstrates that recombinant JAK2 (rJAK2) specifically phosphorylates histone H3, and this activity is inhibited by the JAK2 inhibitor TG101209 . The specificity of this interaction was confirmed through multiple approaches, including the use of mutant histones and specific inhibitors.
H3Y41 phosphorylation is dynamically regulated in response to various stimuli. Cytokine stimulation of K562 cells with leukaemia inhibitory factor (LIF) activates JAK2 and leads to a concomitant increase in H3Y41ph . Similarly, stimulation with PDGF-BB increases H3Y41ph, and this effect can be blocked by JAK2 inhibitors . The IL3 cytokine, which exclusively signals via JAK2 in murine BaF3 cells, also induces an increase in H3Y41ph, further supporting the role of JAK2 in this pathway .
The prevalence of H3Y41 phosphorylation varies across different cell types. It is more abundant in cell lines with active JAK2 signaling (SET2, HEL, UKE1, and K562) and significantly reduced in cell lines lacking detectable JAK2 (HL60 and γ2A cells) . This pattern suggests a strong correlation between JAK2 activity and H3Y41 phosphorylation, though the presence of some H3Y41ph in JAK2-null cells indicates that other tyrosine kinases may also contribute to this modification .
One of the most significant functional consequences of H3Y41 phosphorylation is its effect on the binding of Heterochromatin Protein 1 alpha (HP1α). HP1α specifically binds to the unmodified region of H3 encompassing amino acids 31-56, and this binding is markedly reduced when the peptide is phosphorylated at H3Y41 . Interestingly, HP1β does not bind to this region, indicating a selective interaction between HP1α and this part of H3 .
The binding of HP1α to the Y41 region of H3 is specifically mediated by its chromoshadow domain (CSD), utilizing an alternative binding domain to its interaction with H3K9me . This represents a second distinct binding site for HP1α on histone H3, separate from the well-characterized interaction with methylated H3K9. Phosphorylation of H3Y41 by JAK2 destabilizes the binding of HP1α to chromatin through this second binding site .
The interplay between H3Y41 phosphorylation and HP1α binding has significant implications for gene expression. Genome-wide expression profiling of cells treated with JAK2 inhibitors has identified genes regulated by JAK2, including the hematopoietic oncogene lmo2 . JAK2 inhibition reduces both H3Y41 phosphorylation at the lmo2 promoter and the expression of lmo2, while simultaneously increasing HP1α binding at the same site . This provides a direct mechanistic link between JAK2 activity, H3Y41 phosphorylation, HP1α localization, and gene expression.
Commercially available Phospho-HIST1H3A (Y41) antibodies are typically polyclonal, purified using Protein A and Antigen Affinity methods, and have high purity (≥95% as determined by SDS-PAGE) . These antibodies are carefully designed to ensure specific recognition of the phosphorylated form of H3Y41 without cross-reactivity to unmodified H3 or other phosphorylated residues.
Phospho-HIST1H3A (Y41) antibodies are versatile tools that can be used in various experimental applications. The following table summarizes common applications and typical working dilutions:
| Application | Typical Dilution | Purpose |
|---|---|---|
| Western Blotting | 1:1000 | Detection of H3Y41ph in total protein extracts |
| Immunoprecipitation | 1:50 | Isolation of H3Y41ph-containing proteins |
| Chromatin Immunoprecipitation (ChIP) | 1:100 | Identification of genomic regions enriched for H3Y41ph |
| Immunofluorescence | 1:100 | Visualization of H3Y41ph distribution in cells |
| Flow Cytometry | 1:25 | Quantification of H3Y41ph in cell populations |
These applications enable researchers to investigate the presence, distribution, and dynamics of H3Y41 phosphorylation in various experimental contexts.
Phospho-HIST1H3A (Y41) antibodies serve as valuable tools for monitoring JAK2 activity in the nucleus. The rapid response of H3Y41ph to JAK2 inhibition (observable within 15 minutes, with an 80% decrease after one hour) makes it an excellent readout for JAK2 nuclear activity . This allows researchers to assess the efficacy of JAK2 inhibitors and study the dynamics of JAK2 signaling in various cellular contexts.
The discovery that H3Y41 phosphorylation affects HP1α binding has opened new avenues for investigating chromatin regulation. Researchers can use Phospho-HIST1H3A (Y41) antibodies to map the genome-wide distribution of this modification and correlate it with gene expression patterns, chromatin accessibility, and other epigenetic marks. This has contributed significantly to our understanding of how signaling pathways can directly influence the epigenetic landscape.
Given the frequent activation of JAK2 by chromosomal translocations or point mutations in hematological malignancies, H3Y41 phosphorylation has emerged as a potential marker and mediator of these diseases . Phospho-HIST1H3A (Y41) antibodies enable researchers to investigate the role of this modification in disease progression and evaluate its potential as a diagnostic marker or therapeutic target.
Histone H3Y41 phosphorylation represents a post-translational modification where a phosphate group is added to tyrosine 41 on histone H3. This tyrosine residue is located at the N-terminus of the first helix of H3 (the αN1-helix) where the DNA enters the nucleosome and is juxtaposed to the major groove of the DNA double helix. This positioning makes it strategically important for regulating DNA-protein interactions. The phosphorylation is primarily mediated by the non-receptor tyrosine kinase JAK2, which has been demonstrated to phosphorylate H3Y41 both in vitro and in vivo .
Histone H3 contains three tyrosine residues that are highly conserved across species, but Y41 has particular significance due to its location within the nucleosome core particle. Unlike other common histone modifications such as methylation or acetylation of lysine residues, tyrosine phosphorylation represents a distinct regulatory mechanism with specific functional consequences for chromatin structure .
H3Y41 phosphorylation differs from other histone H3 phosphorylation sites in several key aspects:
| Phosphorylation Site | Primary Kinase | Biological Function | Cell Cycle Association |
|---|---|---|---|
| H3Y41 | JAK2 | Prevents HP1α binding, regulates transcription | Not strongly cell-cycle dependent |
| H3S10 | Aurora B | Chromosome condensation during mitosis | Mitosis marker |
| H3S28 | Aurora B/MSK1 | Chromosome condensation, transcriptional activation | Mitosis and stimulus-responsive |
Unlike H3S10 and H3S28 phosphorylation, which are primarily associated with mitotic chromosome condensation and segregation, H3Y41 phosphorylation has been linked to transcriptional regulation through its ability to exclude HP1α from chromatin . While phosphorylation at serine residues is often used as mitotic markers, Y41 phosphorylation appears to be more closely associated with signaling-dependent transcriptional regulation rather than cell cycle progression per se .
Several complementary approaches can be employed to detect H3Y41 phosphorylation:
Western Blotting: Western blot remains the primary method for detecting global levels of H3Y41ph in cellular preparations. This approach enables quantitative assessment of H3Y41ph levels in response to experimental manipulations such as cytokine stimulation or JAK2 inhibition. When performing western blots, chromatin preparations are recommended over whole cell lysates to enrich for the histone fraction .
Chromatin Immunoprecipitation (ChIP): ChIP is essential for determining the genomic localization of H3Y41ph. This technique can reveal which specific genomic loci are enriched for this modification, providing insights into genes potentially regulated by this modification. ChIP followed by quantitative PCR (ChIP-qPCR) can be used to assess H3Y41ph enrichment at candidate loci, while ChIP-seq provides genome-wide profiling .
Immunofluorescence: This technique allows visualization of H3Y41ph within the nuclear architecture and can provide information about its spatial distribution relative to other nuclear structures or proteins .
For optimal results, using high-specificity antibodies is critical, with testing across multiple experimental platforms to ensure reliable detection of the H3Y41ph modification without cross-reactivity to similar epitopes .
Designing appropriate controls is crucial for H3Y41 phosphorylation studies:
Pharmacological Controls: JAK2 inhibitors such as TG101209 and AT9283 can serve as negative controls. These inhibitors rapidly reduce H3Y41ph levels, with significant decreases observable within 15 minutes and 80% reduction by one hour. This rapid response makes them valuable tools for confirming JAK2-dependent phosphorylation .
Genetic Controls: Cell lines with different JAK2 activity status provide excellent biological controls. Cell lines with active JAK2 signaling (SET2, HEL, UKE1, and K562) typically show higher baseline H3Y41ph levels compared to cell lines lacking detectable JAK2 (HL60 and γ2A cells). Transfection of JAK2 into JAK2-null cells (γ2A) can be used to demonstrate JAK2-dependent H3Y41 phosphorylation .
Peptide Controls: For antibody validation, using unmodified H3 peptides alongside H3Y41-phosphorylated peptides is essential. Additionally, peptides with H3Y41 mutated to phenylalanine (which cannot be phosphorylated) serve as valuable negative controls for both in vitro kinase assays and antibody specificity testing .
Neighboring Gene Controls: When examining H3Y41ph at specific genomic loci, neighboring genes or loci that are not expected to be regulated by this modification should be included as controls. For instance, housekeeping genes like β2-microglobulin typically show no change in H3Y41ph or HP1α binding following JAK2 inhibition .
H3Y41 phosphorylation is primarily regulated by the JAK2 signaling pathway, which integrates various extracellular signals into epigenetic responses:
Cytokine Signaling: Multiple cytokines that signal through JAK2 can induce H3Y41 phosphorylation. Leukaemia inhibitory factor (LIF) activates JAK2 leading to increased H3Y41ph. Similarly, interleukin-3 (IL3) stimulation of murine BaF3 cells, which exclusively signals via JAK2 in these cells, leads to increased H3Y41ph levels .
Growth Factor Signaling: PDGF-BB (Platelet-Derived Growth Factor-BB) can also induce H3Y41 phosphorylation through JAK2 activation. This effect is blocked by JAK2 inhibitors, confirming the JAK2-dependency of this pathway .
Constitutive JAK2 Activation: Oncogenic JAK2 mutations or chromosomal translocations that lead to constitutive JAK2 activation result in elevated baseline H3Y41ph levels. This is particularly relevant in hematological malignancies where JAK2 mutations are common .
The rapidity with which JAK2 inhibitors reduce H3Y41ph levels (within 15 minutes) strongly suggests direct phosphorylation by nuclear JAK2 rather than through lengthy signaling cascades .
H3Y41 phosphorylation directly impacts chromatin structure through its effects on HP1α binding:
Proper validation of phospho-specific antibodies is essential for reliable research outcomes. For H3Y41ph antibodies, a multi-faceted approach is recommended:
Peptide Array Assays: These assays systematically test antibody reactivity against known modifications across all histone proteins and assess the effects of neighboring modifications. This approach, similar to that described by Fuchs et al., provides comprehensive specificity data in a single experiment .
Concentration Titration: Testing antibodies at multiple concentrations (typically three different concentrations) helps ensure that observed reactivity patterns are consistent and not concentration-dependent artifacts .
Western Blot Validation: Comparing antibody reactivity in JAK2-positive versus JAK2-negative cell lines, and before and after JAK2 inhibitor treatment, provides biological validation of specificity. A specific antibody should show reduced signal in JAK2-negative cells and after JAK2 inhibition .
In Vitro Kinase Assays: Using recombinant JAK2 to phosphorylate histone substrates (core histones, purified H3, or recombinant H3) followed by detection with the phospho-specific antibody confirms that the antibody recognizes the JAK2-dependent modification .
Mutation Analysis: Testing antibody reactivity against H3 with Y41 mutated to phenylalanine (which cannot be phosphorylated) serves as a critical negative control that confirms epitope specificity .
Several technical challenges may arise when working with H3Y41ph antibodies:
Cross-Reactivity: Antibodies may detect similar but off-target histone modifications, particularly other phosphorylated tyrosine residues. Comprehensive validation using peptide arrays and mutant H3 proteins is essential to confirm specificity .
Epitope Masking: Steric hindrance from modifications on neighboring residues can inhibit antibody binding, leading to false negative results. This is particularly important for histone tails where multiple modifications can coexist in close proximity .
Fixation Sensitivity: Some epitopes, including phosphorylated residues, may be sensitive to fixation conditions. Optimizing fixation protocols (duration, temperature, and fixative composition) can be critical for immunofluorescence or immunohistochemistry applications .
Extraction Efficiency: Complete extraction of histones, particularly those tightly bound to heterochromatin, may require optimized extraction protocols. Insufficient extraction can lead to underrepresentation of certain chromatin compartments .
Rapid Dephosphorylation: Phosphorylation marks can be dynamically regulated by phosphatases. Including phosphatase inhibitors during sample preparation is essential to prevent artifactual loss of signal .
H3Y41 phosphorylation plays a significant role in hematological gene regulation through several mechanisms:
Oncogene Regulation: H3Y41 phosphorylation has been directly linked to the regulation of the hematopoietic oncogene lmo2. JAK2 inhibition reduces both H3Y41ph at the lmo2 promoter and lmo2 expression while simultaneously increasing HP1α binding at the same site. This establishes a direct mechanistic link between JAK2 activity, H3Y41 phosphorylation, and oncogene expression .
JAK2 Mutation Contexts: In hematological malignancies with JAK2 mutations (such as V617F), constitutive JAK2 activation leads to elevated H3Y41ph levels. This potentially contributes to the altered gene expression profiles observed in these cancers by preventing HP1α-mediated gene silencing at specific loci .
Cytokine Responsiveness: Hematopoietic cells rely on cytokine signaling for survival, differentiation, and proliferation. The JAK2-mediated H3Y41 phosphorylation provides a direct nuclear mechanism by which cytokine signals can rapidly modulate gene expression through chromatin remodeling .
The discovery of direct JAK2-mediated histone modification represents a paradigm shift in understanding how cytoplasmic signaling cascades directly influence nuclear events in hematopoietic contexts .
Distinguishing between global and locus-specific H3Y41 phosphorylation requires complementary approaches:
Global Assessment:
Western blotting of chromatin preparations provides quantification of total cellular H3Y41ph levels .
Mass spectrometry of purified histones can provide unbiased, antibody-independent quantification of H3Y41ph relative to other modifications.
Immunofluorescence microscopy allows visualization of nuclear distribution patterns and potential colocalization with other nuclear markers.
Locus-Specific Assessment:
ChIP-qPCR targeting specific genomic regions allows quantitative comparison of H3Y41ph enrichment at candidate loci before and after experimental manipulation .
ChIP-seq provides genome-wide profiles of H3Y41ph distribution, enabling identification of all genomic loci enriched for this modification.
CUT&RUN or CUT&Tag techniques may offer higher resolution and lower background compared to traditional ChIP for mapping H3Y41ph genome-wide.
Integrated Analysis:
Combining global assessments with locus-specific techniques allows researchers to determine whether changes in H3Y41ph are uniformly distributed or concentrated at specific genomic regions.
Integration with transcriptomic data (RNA-seq) can reveal correlations between H3Y41ph changes and gene expression alterations, providing functional context.
Sequential ChIP (re-ChIP) approaches can identify genomic regions where H3Y41ph co-occurs with other histone modifications of interest .
Cell type-specific factors significantly impact H3Y41 phosphorylation patterns through multiple mechanisms:
JAK2 Expression Levels: Cell lines with active JAK2 signaling (SET2, HEL, UKE1, and K562) show higher baseline H3Y41ph compared to cell lines lacking detectable JAK2 (HL60 and γ2A cells). This indicates that the basic machinery for this modification varies considerably between cell types .
Cytokine Receptor Expression: Different cell types express distinct patterns of cytokine receptors that signal through JAK2. This leads to cell type-specific responses to cytokine stimulation in terms of H3Y41 phosphorylation dynamics .
Chromatin Landscape: The pre-existing chromatin landscape, including other histone modifications and chromatin accessibility, likely influences which genomic regions are susceptible to H3Y41 phosphorylation in a cell type-specific manner.
Nuclear JAK2 Localization: The mechanism by which JAK2 translocates to the nucleus and its subnuclear distribution may differ between cell types, contributing to distinct patterns of H3Y41ph .
Understanding these cell type-specific factors is critical for interpreting H3Y41ph data and extrapolating findings between different experimental systems or tissues.
The crosstalk between H3Y41 phosphorylation and other histone modifications represents a complex and important area of investigation:
Independent HP1α Binding Sites: H3Y41ph affects HP1α binding independently from the well-characterized H3K9me3-HP1α interaction. This suggests that HP1α recruitment can be regulated through multiple, potentially synergistic mechanisms .
Validation Challenges: The presence of neighboring modifications can affect antibody recognition of H3Y41ph, making validation critical for studies examining modification crosstalk. Peptide array assays that test antibody binding in the context of various neighboring modifications are essential for reliable interpretation .
Functional Consequences: The combination of H3Y41ph with other modifications may lead to distinct functional outcomes beyond what each modification would cause independently. For example, the interplay between activating modifications (like H3K4me3) and H3Y41ph may create unique regulatory states .
Methodological Approaches: Studying modification crosstalk requires specialized approaches:
Sequential ChIP to identify co-occurrence of modifications at specific loci
Mass spectrometry to identify combinatorial modifications on the same histone tail
Synthetic peptides with defined modification patterns to study binding of effector proteins
Understanding this crosstalk is essential for developing a comprehensive model of how histone modifications collectively regulate chromatin structure and function.