HIST1H1E (Ab-16) Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Characteristics

The HIST1H1E (Ab-16) antibody (Catalog #CAC15241) is a rabbit-derived polyclonal IgG antibody validated for multiple applications . Key specifications include:

ParameterDetails
TargetHistone H1.4 (HIST1H1E)
ImmunogenSynthetic peptide around lysine 16 (K16) of human Histone H1.4
Host SpeciesRabbit
ReactivityHuman (Homo sapiens)
ApplicationsELISA, Western Blot (WB), Immunohistochemistry (IHC), Immunofluorescence (IF)
Recommended DilutionsWB: 1:50–1:500; IHC: 1:20–1:200; ELISA: 1:20,000–1:40,000
Concentration1 mg/mL
Storage-20°C in 50% glycerol, 0.01M PBS (pH 7.4), 0.03% Proclin 300 preservative

This antibody exhibits high specificity for the C-terminal domain of HIST1H1E, a region critical for chromatin binding and compaction .

Biological Context of HIST1H1E

HIST1H1E encodes histone H1.4, a replication-dependent linker histone that:

  • Binds linker DNA between nucleosomes to stabilize higher-order chromatin structures .

  • Regulates gene transcription by modulating chromatin accessibility and DNA methylation .

  • Impacts cellular senescence: Frameshift mutations in its C-terminal tail disrupt chromatin dynamics, leading to premature aging phenotypes .

Disease Mechanisms

  • Cancer: Heterozygous HIST1H1E mutations are recurrent in diffuse large B-cell lymphoma (DLBCL). These mutations drive chromatin decompaction, epigenetic reprogramming, and derepression of stem cell genes, enhancing tumorigenesis .

  • Aging: Mutant HIST1H1E proteins induce replicative senescence and accelerated aging by altering chromatin methylation profiles (e.g., H3K36me2 gain, H3K27me3 loss) .

Technical Validation

  • Western Blot: Detects HIST1H1E at ~22 kDa in HeLa, MCF-7, and U87 cell lysates .

  • Immunohistochemistry: Validated in paraffin-embedded human melanoma and colon cancer tissues at 1:100 dilution .

  • Functional Studies: Used to assess HIST1H1E’s role in chromatin remodeling via knockdown or mutation models .

Key Research Findings

Study FocusKey InsightsMethodology
LymphomagenesisHIST1H1E loss in mice enhances germinal center B-cell self-renewal, promoting aggressive lymphoma .CRISPR/Cas9 knockout, ChIP-seq, RNA-seq
Cellular SenescenceMutant HIST1H1E alleles reduce proliferation and induce senescence via disrupted chromatin compaction .RT-PCR, methylation profiling, flow cytometry
Epigenetic DysregulationH1.4 deficiency increases H3K36me2 and decreases H3K27me3, altering transcriptional landscapes .Western blot, histone modification assays

Comparative Analysis of HIST1H1E Antibodies

SupplierCatalog #ClonalityApplicationsCross-Reactivity
BiomatikCAC15241PolyclonalWB, IHC, IF, ELISAHuman-specific
Abcamab3596PolyclonalWB, ChIPHuman
EMD MilliporeMAB1276MonoclonalIHC (paraffin)Rat, human

Clinical and Therapeutic Implications

  • Biomarker Potential: HIST1H1E expression levels correlate with disease progression in lymphoma and age-related disorders .

  • Therapeutic Targeting: Small molecules restoring H1.4 function or compensating for chromatin decompaction are under investigation .

Limitations and Considerations

  • Species Specificity: Most antibodies, including Ab-16, are validated only for human samples .

  • Post-Translational Modifications: Phosphorylation (e.g., Thr18) or acetylation (e.g., Lys16) may require specialized antibodies for detection .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days of receiving it. Delivery times may vary depending on the purchasing method or location. 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 protein plays a critical role in chromatin structure by binding to linker DNA between nucleosomes. This interaction forms the macromolecular structure known as the chromatin fiber. Histones H1 are essential for condensing nucleosome chains into higher-order structured fibers. Furthermore, they act as regulators of individual gene transcription through mechanisms such as chromatin remodeling, nucleosome spacing, and DNA methylation.
Gene References Into Functions
  1. Studies have demonstrated the presence of histones H1.2 and H1.4 in MDA-MB-231 metastatic breast cancer cells. Notably, phosphorylation at S173 of histone H1.2 and at S172, S187, T18, T146, and T154 of H1.4 is significantly elevated during the M phase of the cell cycle, indicating a cell cycle-dependent regulation of these modifications. PMID: 26209608
  2. Research has identified and confirmed alterations in the HIST1H1E protein within the postsynaptic density in individuals with schizophrenia. PMID: 25048004
  3. The N-terminal domain of H1 has been shown to be a critical determinant of the affinity and specificity of H1-chromatin interactions. PMID: 22425985
  4. Phosphorylation of H1.4 at S35, mediated by protein kinase A (PKA), results in the dissociation of H1.4 from mitotic chromatin. However, this phosphorylation is also crucial for specific mitotic functions. PMID: 21852232
  5. Allele-specific underacetylation of histone H4 downstream from the promoter is associated with X-inactivation in human cells. PMID: 12498347
  6. The lysine residue adjacent to the phosphorylation site found on the serine residue on the H1.4 peptide KARKSAGAAKR has also been shown to be methylated, raising the question of whether the hypothesized "methyl/phos" switch could be extended to linker histones. PMID: 15595731
  7. Dynamic Histone H1 Isotype 4 Methylation and Demethylation by Histone Lysine Methyltransferase G9a/KMT1C and the Jumonji Domain-containing JMJD2/KDM4 Proteins PMID: 19144645

Show More

Hide All

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 HIST1H1E and what biological role does it play?

HIST1H1E encodes histone H1.4 protein, a critical member of the histone H1 family that mediates interactions between DNA and nucleosomes. This protein binds to linker DNA between nucleosomes, forming the macromolecular structure known as the chromatin fiber. HIST1H1E is essential for the condensation of nucleosome chains into higher-order structured fibers and significantly impacts genome organization . Beyond its structural role, HIST1H1E acts as a regulator of individual gene transcription through chromatin remodeling, nucleosome spacing, and DNA methylation, making it a critical epigenetic modifier . This protein's function in chromatin dynamics makes it an important target for research involving gene expression regulation and epigenetic mechanisms.

How does the HIST1H1E (Ab-16) antibody differ from other histone H1 antibodies?

The HIST1H1E (Ab-16) antibody is specifically designed to recognize the region surrounding Lysine 16 on the histone H1.4 protein encoded by the HIST1H1E gene . This specificity distinguishes it from other histone H1 antibodies that might target different family members (H1A, H1B, H1C) or different epitopes. The antibody is raised in rabbits as a polyclonal preparation, offering broad epitope recognition while maintaining specificity for the Lys-16 region . Unlike antibodies targeting modified histones (such as acetylated or methylated variants), the HIST1H1E (Ab-16) antibody recognizes the native protein regardless of its modification status, providing researchers with a tool to study total HIST1H1E protein levels and distribution . This contrasts with modification-specific antibodies like Acetyl-HIST1H1E (K16) or Di-methyl-HIST1H1E (K16) that recognize only specific post-translational modifications at this position .

What applications have been validated for the HIST1H1E (Ab-16) antibody?

The HIST1H1E (Ab-16) antibody has been validated for multiple research applications, making it versatile for epigenetic studies. Validated applications include:

  • ELISA (Enzyme-Linked Immunosorbent Assay): For quantitative detection of HIST1H1E protein in solution

  • Western Blot (WB): For detection of denatured HIST1H1E in protein extracts

  • Immunohistochemistry (IHC): For visualization of HIST1H1E in tissue sections

  • Immunofluorescence (IF): For subcellular localization studies with recommended dilution of 1:50-1:200

  • Chromatin Immunoprecipitation (ChIP): For investigating HIST1H1E interactions with DNA and associated proteins

This multi-application validation makes the antibody suitable for comprehensive research projects investigating both the presence and function of HIST1H1E in various experimental contexts.

How should researchers optimize ChIP protocols when using HIST1H1E (Ab-16) antibody?

When optimizing Chromatin Immunoprecipitation (ChIP) protocols with HIST1H1E (Ab-16) antibody, researchers should consider several critical factors:

  • Crosslinking optimization: Since HIST1H1E is a linker histone that may have more dynamic binding compared to core histones, crosslinking time should be carefully titrated. Start with standard 1% formaldehyde for 10 minutes, but consider testing 5-15 minute ranges to determine optimal crosslinking for your specific cell type .

  • Sonication parameters: HIST1H1E associated chromatin may require different fragmentation conditions. Aim for chromatin fragments between 200-500bp, adjusting sonication cycles as needed. Monitor fragmentation efficiency using agarose gel electrophoresis.

  • Antibody concentration: Begin with 2-5μg of HIST1H1E (Ab-16) antibody per ChIP reaction. The polyclonal nature of this antibody may require optimization compared to monoclonal antibodies in some systems .

  • Wash stringency: Optimize salt concentration in wash buffers to reduce background while maintaining specific HIST1H1E binding. A sequential washing approach with increasing stringency is recommended.

  • Validation controls: Always include a non-immune IgG control and a positive control locus known to be associated with HIST1H1E to validate ChIP efficiency.

For maximum reproducibility, standardize cell numbers, crosslinking conditions, and antibody batches across experiments. Additionally, consider parallel ChIP experiments with antibodies targeting known HIST1H1E-interacting proteins to validate and extend your findings.

What are the recommended sample preparation procedures for immunofluorescence with HIST1H1E (Ab-16) antibody?

For successful immunofluorescence studies with HIST1H1E (Ab-16) antibody, the following sample preparation procedures are recommended:

  • Fixation protocol: Most histone studies benefit from paraformaldehyde fixation (4% PFA for 15-20 minutes at room temperature). For HIST1H1E specifically, avoid methanol fixation as it may disrupt nuclear architecture and epitope accessibility .

  • Permeabilization optimization: Use 0.2% Triton X-100 for 10 minutes to ensure nuclear penetration while preserving chromatin structure. Over-permeabilization can lead to signal loss, so time should be carefully controlled.

  • Blocking conditions: Use 3-5% BSA or normal serum (from the species of the secondary antibody) in PBS for at least 1 hour to reduce non-specific binding.

  • Antibody dilution: Start with the manufacturer's recommended dilution of 1:50-1:200 for HIST1H1E (Ab-16) antibody . Optimize based on signal-to-noise ratio in your specific cell type.

  • Nuclear counterstaining: Include DAPI or other nuclear stains to contextualize HIST1H1E distribution within nuclear architecture.

  • Controls: Always include a negative control (primary antibody omitted) and, if possible, a HIST1H1E-depleted sample as positive controls.

  • Antigen retrieval: For tissue sections or challenging samples, consider citrate buffer (pH 6.0) heat-mediated antigen retrieval to improve epitope accessibility.

The signal localization should be primarily nuclear with potentially distinctive patterns corresponding to euchromatin versus heterochromatin regions. Document acquisition parameters carefully to enable quantitative comparisons between experimental conditions.

How can researchers validate the specificity of HIST1H1E (Ab-16) antibody in their experimental system?

Validating antibody specificity is crucial for reliable research. For HIST1H1E (Ab-16) antibody, implement these validation strategies:

  • Genetic knockdown/knockout controls: Perform siRNA knockdown or CRISPR/Cas9 knockout of HIST1H1E and confirm reduced signal in Western blot, IF, or ChIP experiments using the antibody. This provides the strongest evidence for specificity.

  • Peptide competition assay: Pre-incubate the antibody with excess immunizing peptide (sequence around Lys-16 of HIST1H1E) prior to application in your experiment. Specific signals should be significantly reduced or eliminated.

  • Parallel detection with alternative antibodies: Compare results using independent antibodies targeting different epitopes of HIST1H1E. Consistent detection patterns increase confidence in specificity.

  • Recombinant protein controls: Test antibody reactivity against recombinant HIST1H1E and related histone H1 family members to confirm isoform specificity.

  • Mass spectrometry validation: For ChIP or immunoprecipitation experiments, analyze immunoprecipitated proteins by mass spectrometry to confirm enrichment of HIST1H1E.

  • Cross-reactivity testing: Evaluate potential cross-reactivity with other histone H1 variants, particularly in species with high sequence homology around the Lys-16 epitope region .

  • Orthogonal method correlation: Correlate antibody-based findings with orthogonal methods like RNA-seq (for gene expression) or ATAC-seq (for chromatin accessibility) at loci identified in ChIP experiments.

Document all validation experiments thoroughly, as they strengthen the reliability of your research findings and may be required for publication.

How can HIST1H1E (Ab-16) antibody be utilized to study pathological conditions associated with HIST1H1E mutations?

HIST1H1E (Ab-16) antibody can be instrumental in studying the pathological mechanisms underlying HIST1H1E Syndrome (previously called Rahman Syndrome) and related disorders. Researchers can employ this antibody in several sophisticated approaches:

  • Patient-derived cell models: In cells derived from patients with HIST1H1E truncating mutations, use the antibody to assess protein localization and abundance through immunofluorescence and western blotting. Compare with wild-type controls to identify aberrant distribution patterns .

  • Chromatin accessibility mapping: Combine ChIP-seq using HIST1H1E (Ab-16) with ATAC-seq or DNase-seq to correlate HIST1H1E binding with altered chromatin accessibility in disease models. This approach can reveal how mutations affect genome-wide chromatin organization.

  • Protein-protein interaction studies: Employ co-immunoprecipitation with HIST1H1E (Ab-16) followed by mass spectrometry to identify altered protein interaction networks in pathological conditions, potentially revealing disrupted epigenetic regulatory complexes.

  • Comparative epigenetic profiling: Use the antibody in ChIP-seq experiments comparing wild-type and mutant HIST1H1E binding patterns, correlating with histone modification profiles (H3K27me3, H3K4me3, etc.) to understand how HIST1H1E mutations alter the epigenetic landscape.

  • Developmental timing studies: In models of HIST1H1E Syndrome, which presents with developmental abnormalities, use the antibody to track HIST1H1E dynamics during differentiation processes to identify critical developmental windows affected by mutations .

Research has shown that patients with HIST1H1E Syndrome present with symptoms including mild to moderate intellectual disability, dysmorphic features, macrocephaly (in 5/6 patients), hypotonia (in 3/6 patients), and global developmental delay . These clinical manifestations likely result from disrupted chromatin regulation during development, making the HIST1H1E (Ab-16) antibody valuable for mechanistic investigations.

What approaches can be used to study HIST1H1E post-translational modifications in conjunction with HIST1H1E (Ab-16) antibody?

Studying HIST1H1E post-translational modifications (PTMs) in relation to total HIST1H1E distribution requires sophisticated methodological approaches:

  • Sequential ChIP (Re-ChIP): Perform initial ChIP with HIST1H1E (Ab-16) to capture total HIST1H1E, followed by a second round of immunoprecipitation with PTM-specific antibodies (acetyl-HIST1H1E, di-methyl-HIST1H1E) . This identifies genomic regions where HIST1H1E bears specific modifications.

  • Multiplexed immunofluorescence: Combine HIST1H1E (Ab-16) with modification-specific antibodies such as Acetyl-HIST1H1E (K16), Acetyl-HIST1H1E (K51), Acetyl-HIST1H1E (K63), or Di-methyl-HIST1H1E (K16) in multi-color immunofluorescence to visualize the nuclear distribution of distinctly modified HIST1H1E pools .

  • Comparative ChIP-seq analysis: Generate parallel ChIP-seq datasets using HIST1H1E (Ab-16) and modification-specific antibodies, then computationally compare binding profiles to identify regions enriched for specific modifications.

  • Mass spectrometry-based quantification: Immunoprecipitate HIST1H1E using the Ab-16 antibody, then analyze by mass spectrometry to quantify the stoichiometry of various PTMs under different experimental conditions.

  • Proximity ligation assay (PLA): Combine HIST1H1E (Ab-16) with modification-specific antibodies in PLA to quantify the co-occurrence of total HIST1H1E and specific modifications at the single-cell level.

The table below summarizes available antibodies for studying HIST1H1E modifications:

ModificationAntibodyApplicationTarget SiteFunction
UnmodifiedHIST1H1E (Ab-16)ELISA, IF, WB, IHCLys-16 regionTotal HIST1H1E detection
AcetylationAcetyl-HIST1H1E (K16)ELISA, IF, ChIPLysine 16Chromatin decompaction
AcetylationAcetyl-HIST1H1E (K51)ELISA, IF, ChIPLysine 51Transcriptional regulation
AcetylationAcetyl-HIST1H1E (K63)ELISA, IF, ChIPLysine 63DNA damage response
MethylationDi-methyl-HIST1H1E (K16)ELISA, IF, ChIPLysine 16Heterochromatin formation

This combinatorial approach enables researchers to understand how different PTMs influence HIST1H1E function and chromatin organization in various biological contexts .

How can HIST1H1E (Ab-16) antibody be integrated into multi-omics research approaches?

Integrating HIST1H1E (Ab-16) antibody into multi-omics research frameworks can provide comprehensive insights into chromatin biology:

  • ChIP-seq with multi-omics integration: Perform ChIP-seq with HIST1H1E (Ab-16) and integrate with:

    • RNA-seq data to correlate HIST1H1E binding with transcriptional outcomes

    • ATAC-seq or DNase-seq to relate HIST1H1E occupancy with chromatin accessibility

    • Hi-C or other chromosome conformation capture techniques to connect HIST1H1E distribution with 3D genome organization

    • DNA methylation profiles (WGBS, RRBS) to examine relationships between HIST1H1E and DNA methylation patterns

  • Single-cell multi-modal analysis: Apply HIST1H1E (Ab-16) in CUT&Tag or single-cell ChIP-seq protocols, then integrate with single-cell RNA-seq and ATAC-seq data to reveal cell-type-specific functions of HIST1H1E in heterogeneous populations.

  • Spatial omics integration: Combine immunofluorescence using HIST1H1E (Ab-16) with spatial transcriptomics to correlate HIST1H1E nuclear distribution patterns with local gene expression in tissue contexts.

  • Dynamic functional genomics: Implement HIST1H1E (Ab-16) in time-resolved ChIP-seq experiments following perturbation (e.g., differentiation signals, stress responses), integrating with time-course transcriptomics and proteomics to map dynamic regulatory networks.

  • Proteome-wide interaction networks: Use HIST1H1E (Ab-16) for immunoprecipitation coupled with mass spectrometry (IP-MS) to identify protein interaction partners, then integrate with ChIP-seq data to create comprehensive maps of HIST1H1E-centered regulatory complexes.

This multi-omics approach can particularly benefit research into HIST1H1E Syndrome by simultaneously capturing aberrations across multiple regulatory layers. For example, integrating HIST1H1E binding patterns with transcriptome and chromatin accessibility data in patient-derived cells could identify dysregulated pathways contributing to neurodevelopmental phenotypes observed in affected individuals .

What are common technical challenges when using HIST1H1E (Ab-16) antibody in ChIP experiments and how can they be addressed?

Researchers frequently encounter several challenges when performing ChIP with HIST1H1E (Ab-16) antibody:

  • High background signal: This can result from insufficient blocking or antibody cross-reactivity.

    • Solution: Increase blocking time (2 hours minimum), use more stringent washing conditions, and pre-clear chromatin with protein A/G beads before adding the antibody. Consider adding 0.1% BSA to wash buffers to reduce non-specific binding.

  • Low enrichment at expected targets: May indicate epitope masking or antibody inefficiency.

    • Solution: Optimize crosslinking conditions (try shorter crosslinking times of 5-8 minutes), ensure complete cell lysis, and increase antibody concentration. Consider native ChIP (without crosslinking) as HIST1H1E may be sensitive to formaldehyde-induced epitope masking.

  • Inconsistent results between replicates: Often reflects variability in chromatin preparation.

    • Solution: Standardize cell collection protocols, crosslinking conditions, and sonication parameters. Monitor sonication efficiency for each experiment using agarose gel electrophoresis to ensure consistent fragment sizes.

  • Reduced signal in specific cell types: May indicate cell-type-specific chromatin organization affecting epitope accessibility.

    • Solution: Customize lysis and sonication protocols for each cell type. Consider testing alternative antibodies targeting different HIST1H1E epitopes to identify the most suitable one for your specific cell type.

  • PCR inhibition in ChIP-qPCR: Often due to contaminants in immunoprecipitated material.

    • Solution: Include additional purification steps after ChIP, dilute template further in qPCR reactions, or use commercial ChIP DNA purification kits specifically designed to remove inhibitors.

  • Poor signal-to-noise ratio in ChIP-seq: Results in low-quality peak calling.

    • Solution: Increase sequencing depth (minimum 20 million uniquely mapped reads), optimize peak calling parameters for linker histones, and consider using more stringent antibody concentrations to reduce non-specific binding.

A systematic approach to troubleshooting, documenting all parameters, and including appropriate controls will help overcome these technical challenges and ensure reliable results.

How should researchers interpret conflicting results between HIST1H1E (Ab-16) antibody data and other histone H1 variant antibodies?

When confronted with discrepancies between results obtained with HIST1H1E (Ab-16) antibody and other histone H1 variant antibodies, researchers should undertake a systematic analytical approach:

  • Epitope accessibility analysis: Different antibodies target distinct epitopes that may be differentially accessible in various chromatin states. Map the exact epitope locations for each antibody and assess whether chromatin conformation might affect their accessibility under your experimental conditions.

  • Antibody specificity validation: Conduct comprehensive cross-reactivity testing against all histone H1 variants using recombinant proteins. Western blot analysis with recombinant H1 variants can determine if antibodies have unexpected cross-reactivity that explains the discrepancies.

  • Modification interference: Post-translational modifications near the epitope may interfere with antibody binding. Since HIST1H1E (Ab-16) targets the region around Lysine 16, modifications like acetylation or methylation at this residue could affect recognition . Compare your results with those using modification-specific antibodies.

  • Technical vs. biological variation: Determine whether discrepancies represent technical artifacts or genuine biological differences by analyzing:

    • Technical: Batch effects, protocol differences, antibody lot variations

    • Biological: Cell cycle stage, differentiation state, stress response conditions

  • Complementary methodologies: Validate findings using non-antibody based approaches such as:

    • Mass spectrometry of chromatin fractions

    • CRISPR tagging of endogenous HIST1H1E

    • RNA-seq correlation with ChIP-seq data

  • Isoform expression analysis: Quantify the relative expression levels of different H1 variants in your system using RT-qPCR or RNA-seq. Discrepancies may reflect genuine biological differences in variant distribution rather than technical issues.

When reporting conflicting results, transparently document all validation steps and acknowledge the limitations of antibody-based approaches. Consider the possibility that discrepancies may reveal novel biological insights about histone variant dynamics rather than simply representing technical artifacts.

How does sample preparation affect HIST1H1E epitope recognition by the Ab-16 antibody?

Sample preparation significantly impacts HIST1H1E epitope recognition by the Ab-16 antibody. Researchers should consider these critical factors:

  • Fixation effects: Different fixatives can dramatically alter epitope accessibility.

    • Paraformaldehyde (4%, 10 min) typically preserves HIST1H1E epitopes without excessive crosslinking

    • Methanol fixation may expose the epitope but disrupt nuclear architecture

    • Over-fixation (>20 min with 4% PFA) can mask the Lys-16 region through excessive crosslinking

    Recommendation: Optimize fixation time with a time-course experiment (5, 10, 15, 20 min) for your specific cell type.

  • Chromatin state preservation: The natively condensed state of chromatin may limit antibody accessibility.

    • Solution: Include a controlled chromatin relaxation step (5-10mM sodium butyrate treatment for 4-6 hours before fixation) to improve epitope exposure while maintaining physiological relevance.

  • Cell lysis conditions: Harsh lysis buffers may denature the epitope.

    • Recommendation: Use gentle lysis conditions (0.1% NP-40 in PBS for initial lysis, followed by nuclear lysis buffer) for applications requiring native protein conformation.

  • pH and ionic strength effects: The Lys-16 region recognition can be sensitive to pH and salt conditions.

    • Optimal conditions: pH 7.2-7.4 with physiological salt concentration (150mM NaCl)

    • High salt (>300mM NaCl) may disrupt antibody-epitope interactions

    Recommendation: Maintain consistent buffer conditions throughout your experimental workflow.

  • Antigen retrieval requirements: For tissue sections or FFPE samples, heat-induced epitope retrieval dramatically improves detection.

    • Optimal protocol: Citrate buffer (pH 6.0), 95°C for 20 minutes, followed by 20-minute cooling period

  • Protein extraction methods: For western blotting, extraction methods affect epitope preservation.

    • Direct SDS lysis may expose the epitope but denature the protein

    • Acid extraction (0.2N HCl) enriches for histones but may alter epitope conformation

    Recommendation: Use specialized histone extraction protocols that maintain epitope integrity while removing histones from chromatin.

The key to consistent results is standardizing sample preparation protocols and including appropriate controls to monitor epitope accessibility across experimental conditions.

How might HIST1H1E (Ab-16) antibody contribute to understanding the role of HIST1H1E in neurodevelopmental disorders?

The HIST1H1E (Ab-16) antibody offers significant potential for investigating the mechanistic underpinnings of HIST1H1E Syndrome and related neurodevelopmental disorders:

  • Developmental chromatin dynamics: Using the antibody in time-course studies of neural differentiation models could reveal how mutant HIST1H1E affects chromatin reorganization during critical developmental windows. This approach could help explain the intellectual disability and developmental delay observed in patients with HIST1H1E Syndrome .

  • Brain region-specific chromatin regulation: Applying the antibody in spatial transcriptomics and chromatin studies across brain organoids derived from patient iPSCs could identify region-specific vulnerabilities to HIST1H1E dysfunction, potentially explaining the neuroanatomical abnormalities like macrocephaly seen in 5/6 patients .

  • Cell type-specific effects: Single-cell multi-omics approaches incorporating HIST1H1E ChIP-seq could reveal cell type-specific impacts of HIST1H1E mutations during neurodevelopment, potentially identifying the most vulnerable neural populations.

  • Therapeutic target identification: Using HIST1H1E (Ab-16) in ChIP-seq experiments before and after candidate therapeutic interventions could help identify compounds that restore normal chromatin regulation patterns disrupted by HIST1H1E mutations.

  • Genotype-phenotype correlations: Comparing HIST1H1E chromatin binding patterns across cells derived from patients with different HIST1H1E mutations could help explain the phenotypic variability observed in this syndrome, where features like hypotonia affect only a subset of patients (3/6) .

  • Interaction with neurodevelopmental gene networks: ChIP-seq with HIST1H1E (Ab-16) followed by pathway analysis could reveal how HIST1H1E regulates genes involved in neural development, synaptogenesis, and synaptic plasticity, connecting epigenetic dysfunction to cognitive impairment.

This research direction is particularly promising given the recent discovery that heterozygous truncating alterations in HIST1H1E cause a recognizable syndrome with intellectual disability and distinctive physical features , suggesting a critical role for this histone variant in neurodevelopment.

What emerging technologies could enhance the utility of HIST1H1E (Ab-16) antibody for epigenetic research?

Several cutting-edge technologies are poised to dramatically expand the research applications of HIST1H1E (Ab-16) antibody:

  • CUT&Tag and CUT&RUN technologies: These antibody-directed genomic mapping methods offer higher signal-to-noise ratios than traditional ChIP-seq. Adapting HIST1H1E (Ab-16) for these platforms could enable:

    • Ultra-low input chromatin profiling from limited clinical samples

    • Single-cell mapping of HIST1H1E genomic distribution

    • Improved spatial resolution of HIST1H1E binding sites

  • Proximity-based labeling methods: Combining HIST1H1E (Ab-16) with TurboID or APEX2 proximity labeling could revolutionize our understanding of the HIST1H1E interactome by:

    • Capturing transient interactions in living cells

    • Identifying cell-type-specific HIST1H1E protein complexes

    • Mapping the spatial proteome surrounding HIST1H1E binding sites

  • Live-cell imaging technologies: Coupling HIST1H1E (Ab-16) with cell-permeable nanobodies or intrabodies could enable:

    • Real-time tracking of HIST1H1E dynamics during cell cycle progression

    • FRAP (Fluorescence Recovery After Photobleaching) studies to measure HIST1H1E mobility on chromatin

    • Super-resolution visualization of HIST1H1E distribution in relation to chromatin domains

  • Spatial multi-omics integration: Emerging platforms combining immunofluorescence with spatial transcriptomics could allow:

    • Correlation of HIST1H1E localization with local gene expression in tissue context

    • Mapping of tissue-specific HIST1H1E function during development

    • Identification of spatial chromatin organization defects in disease states

  • Microfluidic antibody-based technologies: Microfluidic platforms could enable:

    • High-throughput screening of HIST1H1E interactions with drugs or epigenetic modifiers

    • Parallelized single-cell HIST1H1E functional assays

    • Continuous monitoring of HIST1H1E dynamics in response to stimuli

  • CRISPR-based epigenome editing coupled with antibody detection: This approach could allow:

    • Targeted recruitment or displacement of HIST1H1E at specific genomic loci

    • Functional validation of HIST1H1E binding sites identified by ChIP-seq

    • Creation of synthetic chromatin domains with defined HIST1H1E occupancy

These technological advances hold particular promise for investigating conditions like HIST1H1E Syndrome, where precise mapping of altered chromatin regulation could reveal therapeutic vulnerabilities.

How can computational approaches enhance analysis of data generated using HIST1H1E (Ab-16) antibody?

Advanced computational methods can significantly enhance the value of data generated with HIST1H1E (Ab-16) antibody:

  • Integrated multi-omics data analysis frameworks:

    • Develop computational pipelines that integrate HIST1H1E ChIP-seq with RNA-seq, ATAC-seq, and DNA methylation data

    • Implement machine learning approaches to identify patterns in chromatin features associated with HIST1H1E binding

    • Apply network analysis to connect HIST1H1E binding patterns with transcriptional regulatory networks

  • Sequence motif and structural analysis:

    • Deploy deep learning algorithms to identify subtle DNA sequence preferences for HIST1H1E binding beyond traditional motif analysis

    • Integrate DNA shape features (minor groove width, propeller twist) with sequence data to predict HIST1H1E binding affinities

    • Correlate HIST1H1E binding with 3D genome architectural features from Hi-C data

  • Comparative genomics approaches:

    • Develop cross-species analysis pipelines to identify evolutionarily conserved HIST1H1E functions

    • Create computational methods to compare binding patterns of different H1 variants to identify unique and redundant genomic targets

    • Implement statistical frameworks for differential binding analysis between normal and disease states

  • Single-cell data deconvolution:

    • Apply dimensionality reduction and clustering algorithms to single-cell HIST1H1E binding data

    • Develop trajectory inference methods to map HIST1H1E dynamics during cellular differentiation

    • Create computational tools to integrate single-cell HIST1H1E ChIP-seq with scRNA-seq and scATAC-seq data

  • Predictive modeling of HIST1H1E function:

    • Develop machine learning models to predict transcriptional outcomes based on HIST1H1E binding patterns

    • Create computational frameworks to predict the impact of HIST1H1E mutations on chromatin organization

    • Implement network-based approaches to identify key cellular pathways regulated by HIST1H1E

  • Image analysis enhancements for immunofluorescence data:

    • Apply computer vision algorithms to quantify HIST1H1E nuclear distribution patterns

    • Develop deep learning approaches for automated classification of HIST1H1E localization phenotypes

    • Implement spatial statistics to correlate HIST1H1E distribution with other nuclear features

These computational approaches are particularly valuable for understanding complex disorders like HIST1H1E Syndrome, where integrative analysis could reveal how specific mutations disrupt normal chromatin regulation and lead to developmental abnormalities .

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 2025 TheBiotek. All Rights Reserved.