Phospho-Histone H3.1 (S1) Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

To create the phospho-histone H3.1 (S1) recombinant monoclonal antibody, genes encoding the HIST1H3A antibody are first cloned, encompassing both heavy and light chains. These cloned genes are then inserted into expression vectors, which are introduced into host cells via transfection. The host cells are responsible for the production and secretion of the antibody. Affinity chromatography is employed to ensure the antibody's purity, after which it undergoes rigorous functionality testing in various applications, including ELISA, WB, ICC, and IF, enabling accurate detection of the human HIST1H3A protein phosphorylated at S1.

Phosphorylation at H3.1 S1 can promote chromatin condensation thus leading to gene repression. During mitosis, histone H3.1 S1 phosphorylation plays a role in chromosome condensation and segregation. It helps ensure accurate cell division by regulating chromosomal architecture. Phosphorylated H3.1 S1 is also associated with DNA damage response and repair processes. H3.1 S1 phosphorylation is also linked to cell cycle regulation, epigenetic signaling, and coordinated gene regulation.

Product Specs

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

The phospho-histone H3.1 (S1) recombinant monoclonal antibody is meticulously crafted through a process involving gene cloning, expression vector insertion, and cell transfection. This antibody is specifically designed to recognize the human HIST1H3A protein when phosphorylated at serine 1. The genes encoding the HIST1H3A antibody, encompassing both heavy and light chains, are first cloned. These cloned genes are then inserted into expression vectors, which are subsequently introduced into host cells via transfection. The host cells then produce and secrete the antibody, which undergoes affinity chromatography for purification. Subsequently, the antibody undergoes rigorous functionality testing in various applications, including ELISA, Western blotting, immunocytochemistry, and immunofluorescence, ensuring its accuracy in detecting the human HIST1H3A protein phosphorylated at S1.

Phosphorylation at H3.1 S1 is a key regulator of chromatin condensation, which in turn affects gene expression. During mitosis, histone H3.1 S1 phosphorylation plays a crucial role in chromosome condensation and segregation, ensuring accurate cell division by regulating chromosomal architecture. Phosphorylated H3.1 S1 is also actively involved in DNA damage response and repair processes. Furthermore, H3.1 S1 phosphorylation is intricately linked to cell cycle regulation, epigenetic signaling, and coordinated gene regulation.

Form
Liquid
Lead Time
We are typically able to ship products within 1-3 business days of receiving your order. Delivery times may vary depending on the chosen purchase method and location. For specific delivery timeframes, please consult your local distributors.
Synonyms
H3 histone family member E pseudogene antibody; H3 histone family; member A antibody; H3/A antibody; H31_HUMAN antibody; H3F3 antibody; H3FA antibody; Hist1h3a antibody; HIST1H3B antibody; HIST1H3C antibody; HIST1H3D antibody; HIST1H3E antibody; HIST1H3F antibody; HIST1H3G antibody; HIST1H3H antibody; HIST1H3I antibody; HIST1H3J antibody; HIST3H3 antibody; histone 1; H3a antibody; Histone cluster 1; H3a antibody; Histone H3 3 pseudogene antibody; Histone H3.1 antibody; Histone H3/a antibody; Histone H3/b antibody; Histone H3/c antibody; Histone H3/d antibody; Histone H3/f antibody; Histone H3/h antibody; Histone H3/i antibody; Histone H3/j antibody; Histone H3/k antibody; Histone H3/l antibody
Target Names
Uniprot No.

Target Background

Function

Histone H3.1 serves as a core component of the nucleosome, a fundamental structure responsible for wrapping and compacting DNA into chromatin. This compaction restricts DNA accessibility to cellular machineries that require DNA as a template for various processes. Consequently, histones play a pivotal role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. The accessibility of DNA is dynamically regulated by a complex interplay of post-translational modifications of histones, collectively known as the histone code, along with nucleosome remodeling.

Gene References Into Functions
  1. Research suggests a mechanism for epigenetic regulation in cancer involving the induction of E3 ubiquitin ligase NEDD4-dependent histone H3 ubiquitination. PMID: 28300060
  2. The identification of increased expression of H3K27me3 during a patient's clinical course can be helpful for determining whether the tumors are heterochronous. PMID: 29482987
  3. This study reports that JMJD5, a Jumonji C (JmjC) domain-containing protein, acts as a Cathepsin L-type protease mediating histone H3 N-tail proteolytic cleavage under stress conditions that trigger a DNA damage response. PMID: 28982940
  4. Data suggest that the Ki-67 antigen proliferative index has significant limitations, and phosphohistone H3 (PHH3) presents a viable alternative as a proliferative marker. PMID: 29040195
  5. These findings identify cytokine-induced histone 3 lysine 27 trimethylation as a mechanism that stabilizes gene silencing in macrophages. PMID: 27653678
  6. This study demonstrates that, in the early developing human brain, HIST1H3B constitutes the largest proportion of H3.1 transcripts among H3.1 isoforms. PMID: 27251074
  7. This series of 47 diffuse midline gliomas revealed that the histone H3-K27M mutation was mutually exclusive with IDH1-R132H mutation and EGFR amplification, rarely co-occurred with BRAF-V600E mutation, and was commonly associated with p53 overexpression, ATRX loss, and monosomy 10. Notably, among these K27M+ diffuse midline gliomas. PMID: 26517431
  8. Data indicate that histone chaperone HIRA co-localizes with viral genomes, binds to incoming viral particles, and deposits histone H3.3 onto these. PMID: 28981850
  9. These experiments demonstrated that PHF13 binds specifically to DNA and to two types of histone H3 methyl tags (lysine 4-tri-methyl or lysine 4-di-methyl), functioning as a transcriptional co-regulator. PMID: 27223324
  10. Hemi-methylated CpGs DNA recognition activates UHRF1 ubiquitylation towards multiple lysines on the H3 tail adjacent to the UHRF1 histone-binding site. PMID: 27595565
  11. This study provides, for the first time, a comprehensive description of the MR imaging features of pediatric diffuse midline gliomas with histone H3 K27M mutation. PMID: 28183840
  12. Approximately 30% of pediatric high-grade gliomas (pedHGG), including GBM and DIPG, harbor a lysine 27 mutation (K27M) in histone 3.3 (H3.3). This mutation is correlated with poor prognosis and has been shown to influence EZH2 function. PMID: 27135271
  13. The H3F3A K27M mutation is not uncommon in adult cerebellar HGG. PMID: 28547652
  14. Data show that lysyl oxidase-like 2 (LOXL2) is a histone modifier enzyme that removes trimethylated lysine 4 (K4) in histone H3 (H3K4me3) through an amino-oxidase reaction. PMID: 27735137
  15. Histone H3 lysine 9 (H3K9) acetylation was most prevalent when the Dbf4 transcription level was highest, whereas the H3K9me3 level was greatest during and just after replication. PMID: 27341472
  16. The SPOP-containing complex regulates SETD2 stability and H3K36me3-coupled alternative splicing. PMID: 27614073
  17. These findings suggest that binding of the helical tail of histone 3 (H3) with PHD ('plant homeodomain') fingers of BAZ2A or BAZ2B (bromodomain adjacent to zinc finger domain 2A or 2B) necessitates molecular recognition of secondary structure motifs within the H3 tail, potentially representing an additional layer of regulation in epigenetic processes. PMID: 28341809
  18. The results demonstrate a novel mechanism by which Kdm4d regulates DNA replication by reducing the H3K9me3 level to facilitate the formation of the preinitiation complex. PMID: 27679476
  19. Histone H3 modifications caused by traffic-derived airborne particulate matter exposures in leukocytes. PMID: 27918982
  20. A key role of persistent histone H3 serine 10 or serine 28 phosphorylation in chemical carcinogenesis through regulating gene transcription of DNA damage response genes. PMID: 27996159
  21. hTERT promoter mutations are frequent in medulloblastoma and are associated with older patients, prone to recurrence and located in the right cerebellar hemisphere. Conversely, histone 3 mutations do not appear to be present in medulloblastoma. PMID: 27694758
  22. AS1eRNA-driven DNA looping and activating histone modifications promote the expression of DHRS4-AS1 to economically control the DHRS4 gene cluster. PMID: 26864944
  23. Data suggest that nuclear antigen Sp100C is a multifaceted histone H3 methylation and phosphorylation sensor. PMID: 27129259
  24. The authors propose that histone H3 threonine 118 phosphorylation via Aurora-A alters the chromatin structure during specific phases of mitosis to promote timely condensin I and cohesin disassociation, which is essential for effective chromosome segregation. PMID: 26878753
  25. Hemi-methylated DNA opens a closed conformation of UHRF1 to facilitate its H3 histone recognition. PMID: 27045799
  26. Functional importance of H3K9me3 in hypoxia, apoptosis, and repression of APAK. PMID: 25961932
  27. Taken together, the authors verified that histone H3 is a genuine substrate for GzmA in vivo in Raji cells treated with staurosporin. PMID: 26032366
  28. We conclude that circulating H3 levels correlate with mortality in sepsis patients and inversely correlate with antithrombin levels and platelet counts. PMID: 26232351
  29. Data show that double mutations on the residues in the interface (L325A/D328A) decreases the histone H3 H3K4me2/3 demethylation activity of lysine (K)-specific demethylase 5B (KDM5B). PMID: 24952722
  30. Data indicate that minichromosome maintenance protein 2 (MCM2) binding is not required for incorporation of histone H3.1-H4 into chromatin but is essential for the stability of H3.1-H4. PMID: 26167883
  31. Data suggest that histone H3 lysine methylation (H3K4me3) plays a crucial mechanistic role in leukemia stem cell (LSC) maintenance. PMID: 26190263
  32. PIP5K1A modulates ribosomal RNA gene silencing through its interaction with histone H3 lysine 9 trimethylation and heterochromatin protein HP1-alpha. PMID: 26157143
  33. Data indicate that lower-resolution mass spectrometry instruments can be employed for histone post-translational modifications (PTMs) analysis. PMID: 25325711
  34. Data indicate that inhibition of lysine-specific demethylase 1 activity prevented IL-1beta-induced histone H3 lysine 9 (H3K9) demethylation at the microsomal prostaglandin E synthase 1 (mPGES-1) promoter. PMID: 24886859
  35. The authors report that de novo CENP-A assembly and kinetochore formation on human centromeric alphoid DNA arrays are regulated by a histone H3K9 acetyl/methyl balance. PMID: 22473132
Database Links

HGNC: 4766

OMIM: 137800

KEGG: hsa:8350

STRING: 9606.ENSP00000444823

UniGene: Hs.132854

Involvement In Disease
Glioma (GLM)
Protein Families
Histone H3 family
Subcellular Location
Nucleus. Chromosome.

Q&A

What is Histone H3.1 and what biological significance does S1 phosphorylation have?

Histone H3.1 is one of the core components of the nucleosome, the basic unit of chromatin. It belongs to the H3 family of histone proteins and differs from the H3.3 variant by specific amino acid residues. H3.1 is primarily expressed during S-phase and incorporated into chromatin during DNA replication, making it a replication-coupled (RC) histone .

Phosphorylation at serine 1 (S1) of histone H3.1 is associated with:

  • Promotion of chromatin condensation leading to gene repression

  • Regulation of chromosome condensation during mitosis

  • Involvement in DNA damage response and repair mechanisms

  • Contribution to cell cycle regulation and epigenetic signaling

Unlike other phosphorylation sites such as S10 or S28, the S1 phosphorylation appears to be more directly involved in transcriptional silencing rather than activation.

How do recombinant monoclonal antibodies differ from traditional monoclonal antibodies for histone research?

Recombinant monoclonal antibodies offer several advantages over traditional hybridoma-derived antibodies:

FeatureTraditional Monoclonal AntibodiesRecombinant Monoclonal Antibodies
Production methodHybridoma cell cultureIn vitro expression systems with cloned antibody sequences
ConsistencyMay vary between lotsHigher lot-to-lot consistency
SpecificityGoodBetter specificity and sensitivity
FormulationAnimal-derived components may be presentAnimal origin-free formulations possible
CustomizationLimitedCan be engineered for specific applications
ReproducibilitySubject to hybridoma driftSequence-defined, highly reproducible

For histone research specifically, recombinant antibodies can be designed to recognize precise post-translational modifications with minimal cross-reactivity to other histone marks, which is critical when studying the nuanced epigenetic code .

What are the recommended applications and dilutions for Phospho-Histone H3.1 (S1) antibodies?

Based on manufacturer recommendations across multiple providers:

ApplicationRecommended Dilution RangeNotes
Western Blot (WB)1:500-1:5000Lower dilutions (1:500-1:2000) typically yield optimal results
Immunocytochemistry (ICC)1:50-1:500Fixation method can impact results
Immunofluorescence (IF)1:30-1:200May require optimization based on cell type
ELISAVaries by kit/protocolFollow manufacturer's specific recommendations

Optimization is essential as detection sensitivity may vary depending on:

  • Cell type or tissue used

  • Fixation and permeabilization methods

  • Extent of S1 phosphorylation in your experimental system

  • Secondary antibody and detection system employed

How can researchers validate the specificity of Phospho-Histone H3.1 (S1) antibodies?

Rigorous validation of phospho-specific antibodies is crucial to ensure experimental accuracy:

  • Peptide competition assay: Pre-incubate the antibody with phosphorylated and non-phosphorylated peptides corresponding to the target epitope. A specific signal should be blocked only by the phosphorylated peptide.

  • Phosphatase treatment controls: Treat one sample with lambda phosphatase to remove phosphorylation and compare to untreated samples. The signal should disappear in phosphatase-treated samples.

  • Knock-in/knock-out validation: Use cells where the target site has been mutated (S1A) to prevent phosphorylation or use histone H3.1 knockout models as negative controls.

  • Kinase inhibition: Use specific kinase inhibitors that block the phosphorylation of the S1 residue to confirm specificity.

  • Orthogonal techniques: Confirm results using alternative methods such as mass spectrometry to detect the phosphorylation state of H3.1 S1.

  • Positive controls: Include samples known to have high levels of H3.1 S1 phosphorylation, such as cells synchronized at specific cell cycle stages .

What cell culture and sample preparation methods maximize detection of H3.1 S1 phosphorylation?

Optimal detection of H3.1 S1 phosphorylation requires careful sample preparation:

Cell Culture Considerations:

  • Synchronization: Use techniques like thymidine block combined with nocodazole to enrich for mitotic cells when H3.1 phosphorylation is often elevated

  • Stress induction: Various cellular stresses can alter histone phosphorylation patterns

  • Inhibitor treatments: Calyculin A (phosphatase inhibitor) can be used to preserve phosphorylation states

Sample Preparation for Western Blot:

  • Add phosphatase inhibitors (sodium fluoride, sodium orthovanadate, β-glycerophosphate) to all buffers

  • Use acid extraction methods for histone isolation to preserve phosphorylation marks

  • Keep samples cold during processing to minimize phosphatase activity

  • Consider direct lysis in SDS sample buffer for rapid fixation of phosphorylation states

For Immunofluorescence:

  • Quick fixation with 4% paraformaldehyde is often preferred

  • Brief permeabilization with 0.1-0.2% Triton X-100

  • Include phosphatase inhibitors in buffers

  • Consider antigen retrieval methods if necessary

What are the technical differences between detecting H3.1 S1 phosphorylation versus other histone H3 phosphorylation marks?

Different histone H3 phosphorylation sites present unique technical challenges:

Phosphorylation SiteDetection CharacteristicsTechnical Considerations
H3.1 S1Less studied; can be masked by histone tail binding proteinsMay require specific extraction methods; typically lower abundance than S10
H3 S10Widely studied; strong mitotic markerHigh signal in M-phase; serves as a positive control for many experiments
H3 S28Similar dynamics to S10 but less abundantMay require more sensitive detection methods
H3.3 S31Variant-specific mark; not present on H3.1Requires variant-specific antibodies; useful for distinguishing H3 variants
H3 T3Often co-occurs with S10 phosphorylationCan be detected with specific antibodies for combinatorial analysis

For multiplex detection of different modifications:

  • Consider sequential detection with appropriate stripping between antibodies

  • Use fluorescent multiplex Western blotting with differently labeled secondary antibodies

  • For imaging applications, select antibodies from different host species to allow simultaneous detection

How does Histone H3.1 S1 phosphorylation differ functionally from the more well-characterized S10 phosphorylation?

These phosphorylation marks have distinct functions and dynamics in cellular processes:

FeatureH3.1 S1 PhosphorylationH3 S10 Phosphorylation
Cell cycle patternPresent in interphase and mitosisDramatically increases during mitosis
Primary functionGene repression; chromatin condensationMitotic chromosome condensation; gene activation
Associated kinasesMultiple candidates including MSK1/2Aurora B kinase (mitotic); MSK1/2, PKA (interphase)
Interplay with other marksMay prevent methylation/acetylation of nearby residuesDisrupts HP1 binding to adjacent H3K9me3
Gene expression effectsGenerally repressiveContext-dependent: activating in interphase, associated with condensed chromatin in mitosis
ConservationHighly conserved across speciesHighly conserved across species

The spatial and temporal dynamics of these marks reflect their distinct roles in chromatin biology. While S10 phosphorylation is a robust marker of mitosis widely used in cell cycle studies, S1 phosphorylation appears to play more subtle roles in transcriptional regulation and chromatin organization .

What experimental approaches can be used to study the kinases responsible for H3.1 S1 phosphorylation?

Several complementary approaches can identify and characterize kinases that phosphorylate H3.1 at S1:

  • In vitro kinase assays:

    • Incubate purified recombinant H3.1 with candidate kinases

    • Detect phosphorylation using Phospho-H3.1 (S1) antibodies

    • Quantify using Western blot or ELISA-based methods

    • Can be performed with γ-32P-ATP for radiometric analysis

  • Kinase inhibitor screens:

    • Treat cells with panels of specific kinase inhibitors

    • Monitor changes in H3.1 S1 phosphorylation levels

    • Create dose-response curves to identify potential kinases

  • Genetic approaches:

    • CRISPR/Cas9 knockout or knockdown of candidate kinases

    • Overexpression of constitutively active or dominant negative kinase mutants

    • Monitor effects on global H3.1 S1 phosphorylation levels

  • Immunoprecipitation-based methods:

    • Co-immunoprecipitation to identify kinases physically associated with H3.1

    • Phosphorylation of immunoprecipitated H3.1 using cell extracts

    • Mass spectrometry to identify kinases in complex with H3.1

  • HTRF (Homogeneous Time-Resolved Fluorescence) assays:

    • Enables high-throughput screening of kinase activity

    • Uses donor and acceptor fluorophore-labeled antibodies

    • Signal intensity correlates with phosphorylation levels

How can researchers distinguish between H3.1 and H3.3 phosphorylation patterns in experimental systems?

Distinguishing between H3 variants requires specialized techniques due to their high sequence similarity:

  • Variant-specific antibodies:

    • Use antibodies that recognize the unique regions of H3.1 or H3.3

    • H3.3 S31 phosphorylation is a variant-specific mark (not present in H3.1)

    • For H3.1, antibodies targeting regions containing A31 (vs S31 in H3.3) can be used

  • Sequential chromatin immunoprecipitation (ChIP):

    • First IP with variant-specific antibody

    • Second IP with phosphorylation-specific antibody

    • Identifies genomic regions containing specific variant with specific modifications

  • Mass spectrometry approaches:

    • Can distinguish variant-specific peptides and their modifications

    • Enables quantitative analysis of modification stoichiometry

    • Can detect combinatorial modifications not possible with antibodies

  • Stable isotope labeling by amino acids in cell culture (SILAC):

    • Label histones during protein synthesis

    • Monitor turnover rates of different variants and their modifications

    • Particularly useful for studying replication-dependent vs. independent incorporation

  • Exogenous tagged histones:

    • Express tagged versions of H3.1 or H3.3 (HA, FLAG, GFP)

    • Can be immunoprecipitated with tag-specific antibodies

    • Monitor phosphorylation on specific variant using phospho-specific antibodies

What is the relationship between H3.1 S1 phosphorylation and other histone modifications in the context of the histone code?

H3.1 S1 phosphorylation functions within the complex network of histone modifications that constitute the histone code:

  • Cross-talk with adjacent modifications:

    • S1 phosphorylation may prevent methylation or acetylation of neighboring residues (K4, R2)

    • Can influence reader protein binding to nearby modified residues

    • May alter accessibility of the histone tail to other modifying enzymes

  • Combinatorial effects with other modifications:

    • The combination of S1ph with specific lysine methylation states (K4me3, K9me3, K27me3) may signal for distinct functional outcomes

    • Sequential establishment of modifications during cell cycle progression creates specific chromatin states

  • Experimental approaches to study cross-talk:

    • Use recombinant histones with defined modifications for in vitro studies

    • Mass spectrometry to identify co-occurring modifications

    • Sequential ChIP to identify genome regions with multiple modifications

    • Antibodies specifically recognizing combinatorial modifications

  • Functional consequences:

    • S1 phosphorylation may influence the binding of chromatin remodeling complexes

    • Can affect higher-order chromatin structure and accessibility

    • May target specific genomic regions for silencing or activation depending on context

  • Cell cycle dynamics:

    • Progressive, coordinated restoration of histone modifications occurs after DNA replication

    • S1 phosphorylation shows distinct temporal patterns compared to other modifications such as S10 phosphorylation

What are common technical issues when working with Phospho-Histone H3.1 (S1) antibodies and how can they be addressed?

Researchers frequently encounter these challenges when detecting H3.1 S1 phosphorylation:

IssuePossible CausesSolutions
Weak or no signalLow phosphorylation levels; Phosphatase activity during sample preparation; Antibody concentration too lowAdd phosphatase inhibitors to all buffers; Increase antibody concentration; Enrich for cells with high phosphorylation; Consider signal amplification methods
High backgroundNon-specific binding; Concentration too high; Inadequate blockingOptimize antibody dilution; Use alternative blockers (BSA vs milk); Include competitive peptides; Increase washing stringency
Inconsistent resultsPhosphorylation instability; Cell cycle variation; Lot-to-lot antibody variationStandardize sample preparation; Synchronize cells; Use recombinant antibodies with better lot consistency
Cross-reactivityAntibody recognizing other phosphorylation sitesValidate with peptide competition; Use S1A mutant controls; Consider alternative antibody clones
Epitope maskingProtein-protein interactions blocking antibody accessUse alternative extraction methods; Consider different fixation protocols for IF/ICC

For Western blotting specifically:

  • Transfer efficiency: Use transfer methods optimized for low molecular weight proteins

  • Loading controls: Consider using total H3 antibodies or non-histone loading controls

  • Quantification: Normalize phospho-signal to total H3 signal for accurate comparisons

How can researchers optimize detection of phosphorylated H3.1 in fixed tissue samples for immunohistochemistry?

Optimizing phospho-histone detection in tissue samples requires special considerations:

  • Fixation protocols:

    • Overfixation can mask epitopes; consider shorter fixation times (4-24 hours)

    • Freshly prepared 4% paraformaldehyde is often optimal

    • Avoid acidic fixatives that may affect phospho-epitopes

    • Consider perfusion fixation for animal tissues when possible

  • Antigen retrieval methods:

    • Heat-induced epitope retrieval (HIER) in citrate buffer (pH 6.0) or Tris-EDTA (pH 9.0)

    • Optimize time and temperature (typically 95-100°C for 15-30 minutes)

    • Allow slow cooling to room temperature

    • Include phosphatase inhibitors in retrieval buffers

  • Signal amplification:

    • Consider tyramide signal amplification systems

    • Polymer-based detection systems may provide better sensitivity

    • Avoid excessive amplification that can increase background

  • Controls and validation:

    • Include phosphatase-treated sections as negative controls

    • Use tissues known to contain cells with high H3.1 S1 phosphorylation (e.g., proliferating tissues)

    • Consider mitotic cells (identifiable by DAPI) as internal positive controls

  • Quantification approaches:

    • Digital image analysis can provide quantitative data

    • Consider normalization to total H3 staining

    • Set consistent thresholds for positive staining

What emerging technologies offer improved detection of histone phosphorylation beyond traditional methods?

Several advanced technologies are enhancing histone phosphorylation research:

  • Single-cell analysis approaches:

    • Mass cytometry (CyTOF) for multiparameter analysis of histone modifications

    • Single-cell Western blotting for phospho-histone analysis

    • Imaging mass spectrometry for spatial distribution of modifications

  • High-sensitivity detection methods:

    • HTRF (Homogeneous Time-Resolved Fluorescence) assays provide plate-based detection without Western blotting

    • Luminex bead-based multiplex assays for simultaneous detection of multiple phosphorylation sites

    • Digital ELISA platforms offering femtomolar sensitivity

  • Genomic approaches:

    • CUT&RUN and CUT&Tag methods for improved chromatin profiling

    • ChIP-seq combining phospho-specific antibodies with next-generation sequencing

    • CHIC/HiChIP methods combining chromatin conformation with histone modification analysis

  • Live-cell imaging techniques:

    • FRET-based biosensors for real-time monitoring of histone phosphorylation

    • Specific fluorescent probes for visualizing phosphorylated histones

    • Optogenetic tools to manipulate kinase activity with spatial and temporal precision

  • Synthetic biology approaches:

    • Engineered histones with unnatural amino acids at specific positions

    • Chemical methods for generating homogeneously modified histones

    • Designer reader domains for detecting specific modification patterns

How is H3.1 S1 phosphorylation dysregulated in cancer and other diseases?

While H3.1 S1 phosphorylation is less studied than other histone marks in disease contexts, emerging research suggests several important connections:

  • Cancer associations:

    • Altered H3.1 S1 phosphorylation patterns observed in several cancer types

    • May contribute to aberrant gene silencing of tumor suppressors

    • Dysregulation of kinases responsible for S1 phosphorylation often occurs in tumors

    • Potential biomarker for specific cancer subtypes or progression stages

  • Neurodegenerative disorders:

    • Altered histone phosphorylation patterns observed in models of neurodegeneration

    • May contribute to transcriptional dysregulation in affected neurons

    • Could represent potential therapeutic targets

  • Inflammatory conditions:

    • Inflammation-associated signaling can affect histone phosphorylation patterns

    • May contribute to epigenetic reprogramming during chronic inflammation

  • Developmental disorders:

    • Proper histone modification dynamics are essential for development

    • Mutations affecting histone H3.1 or its modifying enzymes linked to developmental abnormalities

  • Experimental approaches:

    • Tissue microarrays with phospho-H3.1 (S1) immunostaining to assess clinical correlations

    • Genetic models manipulating kinases/phosphatases regulating S1 phosphorylation

    • Integration with other epigenetic markers for comprehensive disease profiling

What are the technical considerations for studying H3.1 S1 phosphorylation in patient-derived samples?

Working with clinical specimens presents unique challenges for histone phosphorylation analysis:

  • Sample preservation:

    • Phosphorylation marks are labile and sensitive to post-collection handling

    • Flash freezing or immediate fixation is optimal

    • Document ischemia time and preservation method

    • Consider phosphatase inhibitors during sample collection

  • FFPE tissue considerations:

    • Formalin fixation can affect epitope accessibility

    • Standardize fixation protocols (time, buffer, temperature)

    • Optimize antigen retrieval methods specifically for phospho-epitopes

    • Consider using phosphatase inhibitors in retrieval buffers

  • Controls and normalization:

    • Use matched normal tissues when possible

    • Consider adjacent normal tissue within samples as internal control

    • Normalize phospho-signal to total H3 levels

    • Include phosphatase-treated sections as technical controls

  • Quantification methods:

    • Digital pathology approaches for objective quantification

    • Consider H-score methods (intensity × percentage positive cells)

    • Blinded scoring by multiple observers

    • Use image analysis software for consistent threshold application

  • Integration with other biomarkers:

    • Multiplex immunohistochemistry/immunofluorescence for co-localization studies

    • Correlate with clinical parameters and outcomes

    • Consider laser capture microdissection for cell type-specific analysis

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.