Phospho-Histone H1.4 (T17) Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to Phospho-Histone H1.4 (T17) Recombinant Monoclonal Antibody

Phospho-Histone H1.4 (T17) Recombinant Monoclonal Antibody is a specialized immunological reagent designed to recognize and bind specifically to histone H1.4 protein when it is phosphorylated at the threonine 17 (T17) position. This antibody serves as a crucial tool in epigenetic research, enabling scientists to study the specific post-translational modification of histone H1.4, which plays important roles in chromatin organization and gene regulation. The specificity of this antibody allows researchers to distinguish between phosphorylated and non-phosphorylated forms of the protein, offering valuable insights into the dynamic processes of chromatin remodeling and gene expression regulation.

Histone H1.4 is one of the eleven human H1 isoforms and represents one of the most widely expressed somatic H1 variations with significant levels of expression in many cell types . As a linker histone, H1.4 binds to linker DNA between nucleosomes, contributing to the formation of higher-order chromatin structures and thereby influencing cellular processes ranging from gene expression to DNA replication and repair. Understanding the modifications of histone H1.4, particularly phosphorylation events, provides critical insights into the mechanisms controlling chromatin dynamics.

Recombinant monoclonal antibodies against phospho-Histone H1.4 (T17) are produced using advanced recombinant DNA technology, ensuring consistent performance across experimental uses. These antibodies are isolated from tissue culture supernatant containing vectors of the human phospho-Histone H1.4 (T17) monoclonal antibody gene and undergo affinity-chromatography purification . This production method offers significant advantages over traditional antibody production, including superior lot-to-lot consistency, continuous supply, and elimination of animal-derived variability .

Biological Function of Histone H1.4 and its T17 Phosphorylation

Histone H1.4 serves critical functions in the organization and regulation of chromatin structure. As a member of the linker histone family, H1.4 binds to linker DNA between nucleosomes forming the macromolecular structure known as the chromatin fiber . These histones are necessary for the condensation of nucleosome chains into higher-order structured fibers, which is essential for proper chromosome organization during both interphase and cell division. Beyond this structural role, histone H1.4 also acts as a regulator of individual gene transcription through its involvement in chromatin remodeling, nucleosome spacing, and DNA methylation processes .

Phosphorylation of histone H1 variants, including H1.4, has been recognized as one of the most prominent post-translational modifications affecting their function. The modification status of H1.4 changes dynamically during various cellular processes, including the cell cycle progression, transcriptional activation, and in response to various signaling pathways . The amino and carboxy terminal tails of histone H1 variants are among the most abundantly post-translationally modified sequences in the cell, with multiple simultaneous modifications frequently occurring .

While much research has focused on other phosphorylation sites such as serine 187 (S187), which has been associated with gene activation, the threonine 17 (T17) phosphorylation represents another important regulatory site . Research has revealed conflicting evidence regarding the functional consequences of histone H1 phosphorylation. Some studies suggest that phosphorylation can cause H1 release from chromatin, leading to transcriptional activation of certain genes, while other evidence indicates that phosphorylated H1 may remain associated with active promoters and potentially play a direct role in transcription activation . This apparent contradiction suggests that different H1 isoforms and their specifically modified versions are not functionally redundant but may play distinct roles in chromatin regulation depending on the cellular context and the specific modification site.

Applications in Research

Phospho-Histone H1.4 (T17) Recombinant Monoclonal Antibody serves as a valuable tool in various research applications, allowing scientists to investigate the presence, distribution, and dynamics of this specific histone modification in different experimental contexts. The major applications include:

Western Blotting (WB)

Western blotting represents one of the primary applications for these antibodies, allowing researchers to detect and quantify phosphorylated H1.4 in cell or tissue lysates. Commercial antibodies are typically used at dilutions ranging from 1:300 to 1:5000 for this application . Western blotting has been successfully employed to analyze extracts from various cell lines, demonstrating the specificity of these antibodies for the phosphorylated form of histone H1.4. This technique enables researchers to monitor changes in T17 phosphorylation levels under different experimental conditions, such as during cell cycle progression or in response to various treatments that affect chromatin structure.

Immunohistochemistry (IHC)

Immunohistochemistry allows visualization of the distribution of phosphorylated H1.4 in tissue sections, providing insights into the spatial localization of this modification within tissues and organs. The commercial antibodies are typically used at dilutions of approximately 1:50 to 1:500 for this application . Published validation studies have demonstrated successful staining in paraffin-embedded tissues, including both normal tissues and cancer samples such as colon carcinoma . This application is particularly valuable for understanding the role of histone H1.4 phosphorylation in tissue-specific gene regulation and in pathological conditions.

Immunofluorescence (IF)

Immunofluorescence techniques employing these antibodies (typically at dilutions of 1:30 to 1:200) allow examination of the subcellular localization of phosphorylated H1.4 within individual cells . Published validation studies have shown successful staining in various cell lines, including NIH-3T3 cells and colorectal cancer (CRC) cells . Immunofluorescence has revealed important information about the nuclear distribution of phosphorylated histones during different cell cycle stages and transcriptional states, contributing to our understanding of the dynamic regulation of chromatin structure.

Enzyme-Linked Immunosorbent Assay (ELISA)

Some of these antibodies, particularly the one from Cusabio (CSB-RA010380A17phHU), have been validated for use in ELISA applications . This technique provides a quantitative method to measure phosphorylated H1.4 levels in biological samples, allowing for high-throughput screening and quantitative analysis of this modification under various experimental conditions.

Chromatin Immunoprecipitation (ChIP)

While not explicitly validated for all commercial antibodies in the search results, some histone H1.4 antibodies have been used for chromatin immunoprecipitation studies . This technique allows researchers to identify genomic regions where phosphorylated H1.4 is bound, providing valuable insights into the role of this modification in gene regulation. ChIP studies using phospho-specific antibodies have been instrumental in connecting histone modifications to specific gene regulatory elements and understanding their functional significance.

Validation Studies and Quality Control

Commercial Phospho-Histone H1.4 (T17) antibodies undergo rigorous validation to ensure specificity and reliability across different experimental applications. These validation studies are essential for confirming that the antibodies specifically recognize the phosphorylated form of histone H1.4 at threonine 17 without cross-reactivity to non-phosphorylated forms or other similar phosphorylation sites.

Western Blot Validation

Western blot validation typically involves testing with extracts from various cell lines to confirm that the antibody detects bands of the expected molecular weight (approximately 30 kDa) . For phospho-specific antibodies, validation often includes comparative analysis with samples treated with phosphatase to remove phosphorylation or with samples from cells where the phosphorylation has been induced or inhibited through specific treatments. The search results indicate that commercial antibodies have been validated using extracts of various cells, with secondary detection using HRP-conjugated anti-rabbit IgG antibodies .

Immunohistochemistry Validation

Validation for immunohistochemistry applications typically involves testing on paraffin-embedded tissue sections from both human and animal sources. The search results indicate successful validation in various tissues, including human and mouse colon tissues and human colon carcinoma samples . These studies typically use defined antibody dilutions (often 1:100) and imaging with appropriate magnification (e.g., 40x lens) to demonstrate specific nuclear staining patterns consistent with the expected localization of histone proteins.

Immunofluorescence Validation

Immunofluorescence validation involves testing in cell lines such as NIH-3T3 or CRC cells to confirm nuclear localization and specificity of the staining pattern . Controls often include competing phosphopeptides or dephosphorylation treatments to demonstrate phospho-specificity. The nuclear staining pattern observed in these validation studies confirms the expected localization of histone H1.4 and provides evidence for the specificity of the antibody for the phosphorylated form.

Quality control measures for these antibodies ensure that each lot meets stringent standards for purity, specificity, activity, and consistency between lots. The recombinant nature of many of these antibodies offers particular advantages for quality control, as the production process can be more precisely controlled compared to traditional antibody production methods. This results in greater lot-to-lot consistency, which is crucial for reproducible research results across different experiments and laboratories.

Buffer Formulations

Commercial antibodies are typically supplied in specialized buffer formulations that help maintain stability during storage. These often include:

  • Phosphate-buffered saline (PBS) as the base buffer

  • Bovine serum albumin (BSA) at approximately 1% concentration to stabilize the antibody protein

  • Glycerol at 40-50% concentration to prevent freezing at -20°C, which would damage the antibody structure

  • Sodium azide (0.02-0.05%) as a preservative to prevent microbial contamination

Dilution and Application Recommendations

When preparing working dilutions of the antibody for specific applications, it is important to follow the manufacturer's recommendations:

  • For western blotting, dilutions typically range from 1:300 to 1:5000

  • For immunohistochemistry, dilutions of 1:50 to 1:500 are commonly used

  • For immunofluorescence, dilutions of 1:30 to 1:200 are often recommended

Working dilutions should be prepared fresh before use and may benefit from the addition of appropriate blocking agents (such as BSA or normal serum) to reduce non-specific binding. When working with phospho-specific antibodies, the inclusion of phosphatase inhibitors in sample preparation buffers is often advisable to preserve the phosphorylation state of the target protein.

Product Specs

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

This recombinant monoclonal antibody specifically recognizes human Histone H1.4 phosphorylated at threonine 17 (p-Histone H1.4 (T17)). It was produced and purified from the tissue culture supernatant (TCS) of cell lines expressing a vector encoding the human p-Histone H1.4 (T17) monoclonal antibody gene. Purification was achieved via affinity chromatography. The antibody is of rabbit IgG isotype and is suitable for ELISA, immunohistochemistry (IHC), and immunofluorescence (IF) applications using human samples.

Linker histone H1, including H1.4 (one of eleven human isoforms and a widely expressed somatic variant), plays a crucial role in maintaining and establishing higher-order chromatin structure. Phosphorylation is a key post-translational modification of H1. This anti-p-Histone H1.4 (T17) antibody is a valuable tool for detecting and localizing Histone H1.4 phosphorylated at Thr17.

Form
Liquid
Lead Time
Orders typically ship within 1-3 business days. Delivery times may vary depending on shipping method and destination. Please consult your local distributor for specific delivery timelines.
Synonyms
H1 histone family member 4 antibody; H1.4 antibody; H14_HUMAN antibody; H1E antibody; H1F4 antibody; Hist1h1e antibody; Histone 1 H1e antibody; Histone cluster 1 H1e antibody; Histone H1 antibody; Histone H1.4 antibody; Histone H1B antibody; MGC116819 antibody
Target Names
HIST1H1E
Uniprot No.

Target Background

Function

Histone H1 proteins bind to linker DNA between nucleosomes, contributing to the formation of the chromatin fiber. They are essential for the compaction of nucleosome chains into higher-order structures and also regulate gene transcription through mechanisms involving chromatin remodeling, nucleosome spacing, and DNA methylation.

Gene References Into Functions
  1. Histones H1.2 and H1.4 were identified in MDA-MB-231 metastatic breast cancer cells. Phosphorylation at specific residues (H1.2 S173; H1.4 S172, S187, T18, T146, and T154) showed a significant increase during the M phase, indicating cell cycle dependency. PMID: 26209608
  2. This study identified and characterized changes in HIST1H1E protein within the postsynaptic density in schizophrenia. PMID: 25048004
  3. The N-terminal domain of H1 significantly influences the affinity and specificity of H1-chromatin interactions. PMID: 22425985
  4. PKA-mediated phosphorylation of H1.4 at Serine 35 (S35) leads to its dissociation from mitotic chromatin, suggesting a role in specific mitotic processes. PMID: 21852232
  5. Allele-specific underacetylation of histone H4 downstream of promoters is linked to X-inactivation in human cells. PMID: 12498347
  6. Methylation of a lysine residue adjacent to a phosphorylated serine residue on the H1.4 peptide (KARKSAGAAKR) was observed, raising questions about the potential interplay between methylation and phosphorylation in linker histones. PMID: 15595731
  7. Dynamic methylation and demethylation of Histone H1 isotype 4 are regulated by G9a/KMT1C and JMJD2/KDM4 proteins, respectively. PMID: 19144645
Database Links

HGNC: 4718

OMIM: 142220

KEGG: hsa:3008

STRING: 9606.ENSP00000307705

UniGene: Hs.248133

Involvement In Disease
Rahman syndrome (RMNS)
Protein Families
Histone H1/H5 family
Subcellular Location
Nucleus. Chromosome. Note=Mainly localizes in heterochromatin. Dysplays a punctuate staining pattern in the nucleus.

Q&A

What is Phospho-Histone H1.4 (T17) and what is its role in chromatin biology?

Phospho-Histone H1.4 (T17) refers to the phosphorylation of the threonine residue at position 17 in the H1.4 histone variant. Histone H1.4 belongs to the linker histone family, which binds to linker DNA between nucleosomes and plays essential roles in the formation and maintenance of higher-order chromatin structure. These linker histones are necessary for the condensation of nucleosome chains into higher-order structured fibers and act as regulators of individual gene transcription through involvement in chromatin remodeling, nucleosome spacing, and DNA methylation . The specific phosphorylation at T17 creates a unique modification state that may mediate particular chromatin functions during cell cycle progression or in response to cellular signaling pathways.

How does Phospho-Histone H1.4 (T17) differ from other histone H1 phosphorylation sites?

Histone H1.4 contains multiple phosphorylation sites, each potentially mediating distinct functions. The T17 phosphorylation occurs in the N-terminal domain, which is one of the unstructured tails of H1 that undergoes extensive post-translational modifications. This differs from other well-characterized sites such as phosphorylation at S187 in the C-terminal domain, which has been associated with active promoters , or T146 phosphorylation, which has been identified on condensed mitotic chromatin . Unlike some phosphorylation events, such as S35 phosphorylation which has been shown to cause removal of H1.4 from chromatin, the specific function of T17 phosphorylation may involve more nuanced roles in chromatin regulation . The amino and carboxy terminal tails of histone H1 variants are among the most abundantly post-translationally modified sequences in the cell, with multiple PTMs frequently occurring simultaneously on a single H1 molecule .

What are the common tools for detecting Phospho-Histone H1.4 (T17) in biological samples?

The primary tools for detecting Phospho-Histone H1.4 (T17) are specific antibodies designed to recognize this modification. Multiple commercial antibodies are available as either rabbit monoclonal or polyclonal antibodies. For instance, the recombinant monoclonal antibody from Abcam (ab188294, clone EPR18087) specifically detects both Histone H1.3 (phospho T17) and Histone H1.4 (phospho T17) . Other options include the rabbit monoclonal antibody from Boster Bio (catalog #P06652, clone DGH-8) and Bioss's monoclonal antibody (catalog #bsm-52267R, clone 3A4) . These antibodies have been validated for multiple applications including Western blot (WB), immunohistochemistry (IHC), immunocytochemistry (ICC), and immunofluorescence (IF), with demonstrated reactivity in human, mouse, and rat samples .

What are the recommended protocols for using Phospho-Histone H1.4 (T17) antibodies in Western blot analyses?

For Western blot applications using Phospho-Histone H1.4 (T17) antibodies, researchers should follow these optimized protocols:

  • Sample preparation: Extract histones using acid extraction methods, which are particularly important for enriching histone proteins. For cell cultures, treatment with colcemid has been used to enhance phosphorylation signals .

  • Protein loading and separation: Load approximately 10 μg of histone-enriched protein extract per lane on SDS-PAGE gels. The observed molecular weight for Histone H1.4 is approximately 36 kDa, although the calculated molecular weight is 21.865 kDa—this discrepancy is common for highly basic proteins like histones .

  • Antibody dilution: The recommended dilution ratios vary by manufacturer:

    • Abcam's EPR18087 antibody: 1/5000 dilution

    • Other antibodies may require different dilutions (typically 1/200 to 1/1000)

  • Blocking: Use 5% non-fat dry milk in TBST as blocking/dilution buffer .

  • Controls: Include both untreated and treated (e.g., colcemid-treated for mitotic enrichment) samples to demonstrate specificity of the phosphorylation signal .

The antibody should detect a single band at approximately 36 kDa in samples where H1.4 T17 phosphorylation is present.

How can Phospho-Histone H1.4 (T17) antibodies be used effectively in immunofluorescence studies?

For optimal immunofluorescence applications with Phospho-Histone H1.4 (T17) antibodies, researchers should:

  • Fixation: Use 4% paraformaldehyde for cell fixation, followed by permeabilization with 0.1-0.2% Triton X-100.

  • Blocking: Block with either 1-5% BSA or 5-10% normal serum from the species in which the secondary antibody was raised.

  • Primary antibody incubation: Dilute the antibody according to manufacturer recommendations (typically 1/100 to 1/500) in blocking buffer and incubate overnight at 4°C.

  • Nuclear counterstain: Use DAPI or other nuclear dyes to visualize the nuclei, which helps in correlating the phospho-histone signal with chromatin.

  • Microscopy: Confocal microscopy is preferable for detailed nuclear localization studies.

  • Controls: Include both positive controls (such as mitotic cells, which typically show enriched histone phosphorylation) and negative controls (antibody specificity controls, such as phosphatase-treated samples) .

This approach allows for the visualization of the nuclear distribution pattern of Phospho-Histone H1.4 (T17), which can provide insights into its functional role during different cell cycle stages or in response to various treatments.

What sample preparation techniques are optimal for preserving Phospho-Histone H1.4 (T17) in tissue samples for IHC?

For immunohistochemistry (IHC) applications preserving Phospho-Histone H1.4 (T17) in tissue samples, researchers should follow these guidelines:

  • Fixation: Formalin-fixed, paraffin-embedded (FFPE) tissue samples have been successfully used with these antibodies . Fixation should be performed promptly after tissue collection to prevent degradation of phospho-epitopes.

  • Antigen retrieval: Heat-induced epitope retrieval (HIER) is typically necessary for FFPE samples. This often involves citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) heating, with optimization required for specific antibodies.

  • Phosphatase inhibitors: Include phosphatase inhibitors (e.g., sodium fluoride, sodium orthovanadate) in all buffers used during sample preparation to preserve phospho-epitopes.

  • Section thickness: Optimal results are typically obtained with 4-5 μm thick sections.

  • Antibody dilution: A dilution of 1/200 has been reported for some antibodies in IHC-P applications .

  • Detection system: DAB (3,3'-diaminobenzidine) staining has been successfully used for visualizing the antibody binding in tissue sections .

Researchers should validate the specific conditions with positive control tissues known to express the phosphorylated form of Histone H1.4. Brain tissue has been used successfully as seen in the immunohistochemical analysis of rat brain using anti-Histone H1.4 (phospho T18) antibody .

How can researchers distinguish between the signals of different phosphorylated H1 variants?

Distinguishing between signals from different phosphorylated H1 variants presents a significant challenge due to sequence similarity and the presence of multiple modifications. Researchers should employ these strategies:

  • Antibody selection: Choose antibodies with demonstrated specificity for the particular phosphorylation site. For example, some antibodies recognize both Histone H1.3 (phospho T17) and Histone H1.4 (phospho T17) due to sequence similarity , while others may be more specific to one variant.

  • Peptide competition assays: Perform peptide competition experiments using phosphorylated and non-phosphorylated peptides corresponding to the region of interest to confirm antibody specificity.

  • Mass spectrometry validation: Use mass spectrometry (MS) as a complementary approach to antibody-based detection. MS has become widely used to analyze histone H1 variants as it can bypass the limitations of immunological reagents, although even MS has limitations when analyzing histone H1 .

  • Knockout/knockdown controls: When possible, use genetic approaches (siRNA, CRISPR) to deplete specific H1 variants and confirm signal specificity.

  • Multiple antibodies: Use multiple antibodies from different sources that recognize the same modification to corroborate findings.

  • Consider modification combinations: Be aware that the amino and carboxy terminal tails of histone H1 variants frequently contain multiple simultaneous PTMs, which can affect antibody binding and specificity .

What are common pitfalls in interpreting results from Phospho-Histone H1.4 (T17) experiments?

When interpreting results from Phospho-Histone H1.4 (T17) experiments, researchers should be aware of these common pitfalls:

  • Antibody cross-reactivity: Many H1 phospho-specific antibodies may cross-react with similar phosphorylation sites on different H1 variants due to sequence homology. For instance, some antibodies recognize both H1.3 (phospho T17) and H1.4 (phospho T17) .

  • PTM combinatorial effects: The presence of multiple PTMs on histone H1 may affect antibody recognition of the T17 phosphorylation site. Multiple simultaneous PTMs on histone H1 are regularly identified, which can complicate interpretation .

  • Dynamic phosphorylation: Histone phosphorylation is highly dynamic and can change rapidly in response to cellular conditions, making timing of sample collection critical.

  • Observed molecular weight discrepancies: The observed molecular weight of Histone H1.4 (approximately 36 kDa) differs significantly from the calculated molecular weight (21.865 kDa), which is common for histones but may confuse interpretation of Western blot results .

  • Context-dependent functions: The function of T17 phosphorylation may vary depending on cell type, cell cycle stage, or in response to different stimuli, making generalizations difficult.

  • Technical variations: Differences in sample preparation, antibody lots, and detection methods can all contribute to variability in results across experiments.

How can researchers verify the specificity of their Phospho-Histone H1.4 (T17) antibody?

To verify the specificity of Phospho-Histone H1.4 (T17) antibodies, researchers should implement the following validation strategies:

  • Peptide competition assays: Perform pre-absorption of the antibody with the phosphorylated peptide used as the immunogen to confirm that this eliminates the signal.

  • Phosphatase treatment controls: Treat samples with lambda phosphatase to remove phosphate groups and confirm the loss of antibody binding.

  • Multiple application testing: Validate the antibody across multiple applications (WB, IHC, IF) to ensure consistent results. Commercial antibodies are often tested in multiple applications (WB, IHC, ICC/IF) with known positive and negative samples to ensure specificity and high affinity .

  • ENCODE guideline compliance: Follow ENCODE (Encyclopedia of DNA Elements) guidelines for antibody validation. Some commercially available antibodies, such as the purified pS187 H1.4 antibody, have been tested for specificity in accordance with these guidelines .

  • Knockout/knockdown controls: When possible, use samples from knockdown or knockout systems to confirm specificity.

  • Stimulation experiments: Use treatments known to increase or decrease the specific phosphorylation (e.g., kinase inhibitors, cell cycle synchronization) to demonstrate the expected changes in signal intensity.

  • Comparison with other antibodies: Compare results with those obtained using antibodies from different sources or those recognizing different epitopes of the same protein.

How does Phospho-Histone H1.4 (T17) contribute to chromatin dynamics during the cell cycle?

The phosphorylation of Histone H1.4 at threonine 17 represents one of many cell cycle-dependent histone modifications that regulate chromatin structure and function. While the specific role of T17 phosphorylation has not been fully elucidated in the provided search results, we can make informed inferences based on related research:

  • Cell cycle regulation: Histone H1 phosphorylation generally increases during the cell cycle, reaching maximum levels during mitosis. Phosphorylation at specific sites, such as T146 in H1.4, has been identified on condensed mitotic chromatin by immunofluorescence . The T17 phosphorylation may similarly contribute to cell cycle-dependent chromatin condensation or relaxation.

  • Chromatin binding dynamics: Different phosphorylation states of H1 can affect its binding affinity for chromatin. For example, protein kinase A-induced phosphorylation at S35 causes removal of H1.4 from chromatin . The T17 phosphorylation may similarly modulate H1.4's association with DNA, potentially affecting chromatin accessibility during different cell cycle phases.

  • Higher-order structure formation: Histone H1 proteins are necessary for the condensation of nucleosome chains into higher-order structured fibers . Phosphorylation at T17 may regulate this process, potentially facilitating the formation or disassembly of higher-order chromatin structures during cell division.

  • Integration with other histone modifications: T17 phosphorylation likely functions in concert with other histone modifications to orchestrate chromatin structural changes throughout the cell cycle.

What is the relationship between Phospho-Histone H1.4 (T17) and gene expression regulation?

The relationship between Phospho-Histone H1.4 (T17) and gene expression regulation appears complex and context-dependent, based on our understanding of histone H1 biology:

  • Contradictory to traditional views: While histone H1 has traditionally been viewed as a general repressor of transcription, recent evidence suggests that linker histones and their modified forms have more nuanced roles than previously understood and may even play roles in gene activation . For instance, when phosphorylated at serine 187 in the C-terminal domain, H1.4 is enriched at active promoters—directly contrasting with previous reports suggesting phosphorylation of H1 leads to its dissociation from chromatin .

  • Site-specific effects: Different phosphorylation sites on H1.4 may have distinct effects on gene expression. While the specific role of T17 phosphorylation is not explicitly detailed in the search results, its location in the N-terminal domain suggests it may affect interactions with DNA or chromatin-associated proteins differently than C-terminal phosphorylation sites.

  • Regulatory mechanisms: H1.4 acts as a regulator of individual gene transcription through multiple mechanisms:

    • Chromatin remodeling

    • Nucleosome spacing

    • DNA methylation

  • Gene-specific effects: The impact of Phospho-Histone H1.4 (T17) on gene expression likely varies depending on genomic context, cell type, and the presence of other chromatin modifications.

  • Integration with signaling pathways: Histone phosphorylation often serves as an endpoint for various cellular signaling cascades, potentially linking external stimuli to changes in gene expression through modification of chromatin structure.

What are the challenges in studying the kinases and phosphatases that regulate Phospho-Histone H1.4 (T17) levels?

Studying the enzymes that regulate Phospho-Histone H1.4 (T17) levels presents several significant challenges:

  • Multiple kinases and overlapping specificity: Various kinases may be capable of phosphorylating H1.4 at T17, potentially with overlapping specificity. Identifying the specific kinase responsible in different cellular contexts requires careful experimental design. For comparison, other H1.4 phosphorylation sites have identified kinases: Aurora B kinase for S27 phosphorylation, and protein kinase A for S35 phosphorylation .

  • Temporal and spatial regulation: The activity of relevant kinases and phosphatases may be temporally and spatially regulated, making it difficult to capture the dynamic nature of T17 phosphorylation.

  • Antibody limitations: Limitations of immunological reagents make it challenging to specifically detect the T17 phosphorylation. Antibodies must be generated with distinct combinations of localized PTMs to retain specificity for the epitopes of interest, due to the presence of multiple simultaneous PTMs on histone H1 .

  • Mass spectrometry challenges: Even mass spectrometry, which has become widely used to analyze histone H1 variants, has limitations when analyzing histone H1 due to the complexity of modifications .

  • Context-dependent regulation: The regulation of T17 phosphorylation may differ depending on cell type, cell cycle stage, or in response to various stimuli, requiring extensive experimental conditions to fully characterize.

  • Integration with other modifications: The presence of other post-translational modifications on H1.4 may influence the ability of kinases and phosphatases to access and modify the T17 residue, adding another layer of complexity.

What methodological approaches can help reveal the functional significance of Phospho-Histone H1.4 (T17) in different cellular contexts?

To elucidate the functional significance of Phospho-Histone H1.4 (T17) across cellular contexts, researchers should consider these methodological approaches:

  • Genome-wide mapping: ChIP-seq (Chromatin Immunoprecipitation followed by sequencing) using highly specific Phospho-Histone H1.4 (T17) antibodies can reveal the genomic distribution of this modification. This approach has been used successfully for other H1.4 modifications, such as pS187H1.4, which showed enrichment at active promoters in estradiol-responsive MCF7 cells .

  • Phosphomimetic and phospho-null mutations: Generate cell lines expressing H1.4 with T17 mutations to either prevent phosphorylation (T17A) or mimic constitutive phosphorylation (T17E/T17D), then analyze effects on chromatin structure and gene expression.

  • Inducible systems: Develop systems where T17 phosphorylation can be induced or inhibited to study immediate effects on chromatin and transcription.

  • Proteomics approaches: Use proximity labeling or pull-down experiments to identify proteins that specifically interact with Phospho-Histone H1.4 (T17).

  • Combination with other techniques: Integrate ChIP-seq data with RNA-seq, ATAC-seq, and Hi-C to correlate T17 phosphorylation with gene expression, chromatin accessibility, and three-dimensional genome organization.

  • Single-cell analyses: Apply single-cell techniques to understand cell-to-cell variation in T17 phosphorylation and its functional consequences.

  • In vitro reconstitution: Reconstitute nucleosomes and higher-order chromatin structures with H1.4 containing or lacking T17 phosphorylation to study direct effects on chromatin compaction.

  • Super-resolution microscopy: Use advanced imaging techniques to visualize the nuclear distribution of Phospho-Histone H1.4 (T17) with high precision and correlate with chromatin states.

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.