Acetyl-Histone H3.1 (K14) Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Structure and Production

The antibody is produced via recombinant methods, which involve cloning heavy and light chain genes from immunoreactive rabbits into expression vectors . Key features include:

FeatureDetail
Antibody TypeRabbit recombinant monoclonal (e.g., clones EP964Y, JU43-26, D4B9)
TargetAcetylated lysine 14 (H3K14ac) on histone H3.1
Species ReactivityHuman, mouse, rat, monkey (validated in multiple studies)
Production BenefitsBatch-to-batch consistency, animal-free formulation, enhanced specificity

Recombinant production ensures reproducibility and avoids lot-to-lot variability, making it ideal for large-scale studies .

Applications and Validation

This antibody is validated for diverse techniques, with optimized dilutions and protocols:

Application Table

TechniqueDilution RangeKey ValidationsSources
Western Blot (WB)1:500–1:2000Detects a 15 kDa band in human, mouse, and rat lysates .
Immunofluorescence1:50–1:500 (ICC/IF)Nuclear staining in HeLa cells; compatible with TSA-treated samples .
Immunohistochemistry1:50–1:500 (IHC)Automated staining on Leica BOND™ RX platform; validated on human endometrium carcinoma .
ChIP/ChIP-Seq5–20 µg per reactionEffective in chromatin immunoprecipitation assays .

Critical Validation Data:

  • Peptide Array Testing: Demonstrated high specificity for H3K14ac peptides over non-acetylated or other acetylated lysines (e.g., H3K9ac) .

  • Negative Controls: Secondary antibody-only experiments confirm minimal background signal .

Functional Significance of H3K14 Acetylation

H3K14 acetylation is a hallmark of active chromatin regions and is catalyzed by histone acetyltransferases (HATs) such as Elongator . Key roles include:

FunctionMechanismBiological Impact
Transcriptional ActivationPromotes chromatin relaxation, enabling transcription factor access .Upregulates genes involved in growth and survival.
Epigenetic MemoryMaintains active chromatin states across cell divisions .Critical for cellular identity and differentiation.
Disease AssociationDysregulation linked to cancers, neurological disorders, and metabolic diseases .Biomarker potential for diagnostic applications.

Research Findings:

  • Elongator Complex: Holo-Elongator preferentially acetylates H3K14 and H4K8, with nucleosomal substrates enhancing H4K8 acetylation .

  • Gene Regulation: ChIP-Seq studies show H3K14ac enrichment at promoters of actively transcribed genes, particularly in regions regulated by RNA polymerase II .

Challenges and Considerations

  • Cross-Reactivity: While specific to H3K14ac, confirm absence of reactivity with other acetylated lysines (e.g., H3K9ac) using peptide arrays .

  • Storage: Typically stable at -20°C for 12 months; aliquot upon delivery .

  • Species Limitations: Limited reactivity with non-human primates or other species unless homology is confirmed .

Product Specs

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

The development of the acetyl-Histone H3.1 (K14) recombinant monoclonal antibody commences with the cloning of genes encoding the HIST1H3A antibody, encompassing both heavy and light chains. These cloned genes are then integrated into an expression vector, which is subsequently introduced into host cells through transfection. The host cells are cultivated to produce and secrete the antibody. Subsequently, the antibody undergoes purification using affinity chromatography to ensure its purity and efficacy. Rigorous testing then validates its functionality in diverse applications, including ELISA, WB, ICC, and IF, enabling precise detection of human and rat HIST1H3A proteins acetylated at K14.

Acetylation of histone H3.1 at lysine 14 (K14) plays a pivotal role in transcriptional activation, chromatin accessibility, cellular identity, epigenetic memory, and coordinated gene regulation. This modification has implications in various diseases.

Form
Liquid
Lead Time
Generally, we can dispatch products within 1-3 business days after receiving your order. Delivery timelines may vary depending on the purchasing method or location. Kindly consult your local distributors for specific delivery timeframes.
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 is a core component of the nucleosome. Nucleosomes wrap and compact DNA into chromatin, limiting DNA accessibility to cellular machinery that utilize DNA as a template. Histones thus play a central role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is regulated by a complex set of post-translational modifications of histones, known as the histone code, and nucleosome remodeling.
Gene References Into Functions
  1. Research suggests a mechanism for epigenetic regulation in cancer through 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 aid in determining whether tumors are heterochronous. PMID: 29482987
  3. This research reports that JMJD5, a Jumonji C (JmjC) domain-containing protein, acts as a Cathepsin L-type protease that mediates histone H3 N-tail proteolytic cleavage under stress conditions leading to a DNA damage response. PMID: 28982940
  4. Findings suggest that the Ki-67 antigen proliferative index has significant limitations and that phosphohistone H3 (PHH3) is an alternative proliferative marker. PMID: 29040195
  5. This research identifies cytokine-induced histone 3 lysine 27 trimethylation as a mechanism that stabilizes gene silencing in macrophages. PMID: 27653678
  6. This data indicates that, in the early developing human brain, HIST1H3B comprises the largest proportion of H3.1 transcripts among H3.1 isoforms. PMID: 27251074
  7. This series of 47 diffuse midline gliomas demonstrated that the histone H3-K27M mutation was mutually exclusive with IDH1-R132H mutation and EGFR amplification. It rarely co-occurred with BRAF-V600E mutation and was commonly associated with p53 overexpression, ATRX loss, and monosomy 10. PMID: 26517431
  8. Data show that histone chaperone HIRA co-localizes with viral genomes, binds to incoming viral 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) where it functions 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 presents, for the first time, 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) which is correlated with poor outcome and was shown to influence EZH2 function. PMID: 27135271
  13. H3F3A K27M mutation in adult cerebellar HGG is not uncommon. 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. SPOP-containing complex regulates SETD2 stability and H3K36me3-coupled alternative splicing. PMID: 27614073
  17. Data 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) requires molecular recognition of secondary structure motifs within the H3 tail and could represent 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 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. On the other hand, 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 real substrate for GzmA in vivo in the Raji cells treated by 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 important for 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 utilized 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

Show More

Hide All

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 Acetyl-Histone H3.1 (K14) and why is it significant in epigenetic research?

Acetyl-Histone H3.1 (K14) refers to the specific post-translational modification where the lysine residue at position 14 of histone H3.1 is acetylated. This modification is significant in epigenetics because it plays a crucial role in chromatin remodeling and gene expression regulation. Histone H3 is one of the core components of the nucleosome, which consists of 147 base pairs of DNA wrapped around an octamer of core histone proteins .

Lysine acetylation neutralizes the positive charge of histone proteins, weakening the interaction between histones and negatively charged DNA. This modification is generally associated with transcriptionally active chromatin regions. Specifically, H3K14 acetylation is considered a marker of transcriptionally active genes and is performed by histone acetyltransferases (HATs) such as CBP/p300 .

Research has demonstrated that H3K14 acetylation influences chromatin structure by increasing the α-helical content of the H3 tail . This structural change affects how the histone tail interacts with nucleosomal DNA, potentially making DNA more accessible to transcription factors and the transcriptional machinery.

How does the structure of histone H3.1 differ from other H3 variants, and what implications does this have for K14 acetylation?

Histone H3.1 is one of several H3 variants, including H3.2 and H3.3. The H3.1 variant is encoded by genes including HIST1H3A, with aliases such as H3/A, H3C10, H3C11, and others . While H3 variants share high sequence similarity, their expression patterns, incorporation into chromatin, and functions differ significantly.

H3.1 is primarily expressed during S phase and incorporated into chromatin during DNA replication, whereas H3.3 can be incorporated throughout the cell cycle in a replication-independent manner. These differences in chromatin deposition may influence the distribution and functional outcomes of K14 acetylation on different H3 variants.

The amino acid sequence surrounding K14 is conserved among H3 variants, with the full sequence including: "ARTKQTARKSTGGKAPRKQLATKAARK..." . Despite this conservation, K14 acetylation may have variant-specific functions due to differences in chromatin localization and association with distinct protein complexes. Computer simulations have shown that K14 acetylation increases the α-helical content of the H3 tail, suggesting a structural mechanism by which this modification alters chromatin architecture .

What is the relationship between H3K14 acetylation and other histone modifications?

Histone modifications rarely function in isolation; instead, they form complex patterns that collectively influence chromatin structure and function. H3K14 acetylation often co-occurs with other active chromatin marks, including:

  • H3K9 acetylation: Often found together with H3K14ac in promoter regions of active genes

  • H3K4 methylation: H3K4me3 at promoters frequently correlates with H3K14ac

  • H3S10 phosphorylation: Can enhance acetylation at H3K14 by certain HATs

These combinations form part of the "histone code" that regulates gene expression. Importantly, specific antibodies must be validated for selective recognition of H3K14ac without cross-reactivity to other modifications. In particular, antibodies targeting H3K14ac should be tested for potential cross-reactivity with acetylated lysines at positions 9, 18, 23, 27, and others .

The relationship between these modifications can be experimentally determined using sequential ChIP (ChIP-reChIP) or mass spectrometry approaches, which can reveal the co-occurrence of multiple modifications on the same histone tail.

How does K14 acetylation of histone H3.1 influence the tail conformation and subsequent chromatin dynamics?

Molecular dynamics (MD) simulations have provided valuable insights into how K14 acetylation affects H3 tail conformation. Research has demonstrated that although the H3 tail is intrinsically disordered, K14 acetylation significantly increases the α-helical content of specific regions within the tail . This conformational change has several important implications for chromatin dynamics:

  • The increased helical structure alters how the tail interacts with nucleosomal DNA, potentially reducing electrostatic interactions

  • Acetylation-induced conformational changes may expose or mask binding sites for chromatin-associated proteins

  • These structural alterations affect higher-order chromatin folding and compaction

Computer simulations using enhanced sampling methods such as adaptive lambda square dynamics (ALSD) have shown that while the H3 tail has no specific native conformation within the nucleosome, it does exhibit conformational preferences. Residues 2-12 and 17-28 demonstrate high helix content in the H3 tail . When K14 is acetylated, this helical propensity increases, changing how the tail interacts with both nucleosomal DNA and adjacent nucleosomes.

The conformational changes induced by K14 acetylation likely alter the accessibility of the modified residue to reader proteins that specifically recognize this modification, thereby influencing downstream signaling events and transcriptional outcomes.

What are the challenges in distinguishing the specific functions of H3.1K14ac from other acetylation sites on H3?

Distinguishing the specific functions of H3.1K14ac from other acetylation sites presents several significant challenges:

  • Antibody specificity issues: Many antibodies show cross-reactivity with multiple acetylation sites. For instance, pan-acetyl antibodies recognize multiple acetylated lysines on H3 . Even site-specific antibodies require rigorous validation to ensure they don't cross-react with similar epitopes, such as H3K9ac or H3K18ac .

  • Redundancy and compensation: Knockout or mutation of a single acetylation site often leads to compensatory increases in acetylation at other sites, complicating the interpretation of phenotypes.

  • Context-dependent functions: The same modification may have different functions depending on genomic context, cell type, or developmental stage.

  • Technical limitations: Traditional ChIP experiments have limited resolution and cannot easily distinguish between modifications that co-occur within the same nucleosome but on different H3 molecules.

To address these challenges, researchers employ several sophisticated approaches:

  • Using highly specific monoclonal antibodies with demonstrated lack of cross-reactivity to other acetylation sites

  • Employing mass spectrometry to quantitatively measure multiple modifications simultaneously

  • Utilizing genetic approaches like lysine-to-arginine mutations that prevent acetylation at specific sites

  • Developing new techniques like CUT&RUN, CUT&Tag, and single-molecule approaches to study modifications with higher resolution

How do experimental conditions affect the detection of H3.1K14ac using recombinant monoclonal antibodies?

The reliable detection of H3.1K14ac using recombinant monoclonal antibodies is highly dependent on experimental conditions, which must be carefully optimized:

Fixation and Epitope Accessibility:

  • Overfixation with formaldehyde can mask epitopes and reduce antibody binding

  • Chromatin preparation methods significantly impact epitope accessibility

  • Heat-induced epitope retrieval may be necessary for some applications, especially IHC and IF

Buffer Conditions:

  • Salt concentration affects antibody-epitope interactions

  • Detergent types and concentrations influence background signal

  • pH variations can alter antibody specificity and binding efficiency

Post-translational Modification Status:

  • Neighboring modifications can create steric hindrance affecting antibody binding

  • Competing modifications at K14 (methylation, ubiquitination) may mask the acetylation

Table 1: Optimization Conditions for H3.1K14ac Detection in Different Applications

ApplicationRecommended DilutionBuffer ConditionsCritical Parameters
Western Blot1:500 - 1:1000TBST with 5% BSAAcid extraction of histones crucial
ChIP5μg per 5-10μg chromatinPBS with 0.1% Triton X-100Sonication conditions affect epitope exposure
ICC/IF1:50 - 1:200PBS with 1-3% BSAFixation time and permeabilization critical
ELISA1μg/mLCarbonate buffer pH 9.6Blocking agent selection affects S/N ratio

For experiments like ChIP, treatment of cells with HDAC inhibitors (e.g., sodium butyrate) prior to fixation can enhance detection of acetylation marks by preventing their removal . Similarly, for Western blotting, extraction methods significantly impact results, with acid extraction methods generally preferred for histone analysis over standard RIPA buffer extractions.

What are the optimal protocols for using Acetyl-Histone H3.1 (K14) Recombinant Monoclonal Antibody in ChIP and ChIP-seq experiments?

Chromatin immunoprecipitation (ChIP) using Acetyl-Histone H3.1 (K14) Recombinant Monoclonal Antibody requires careful optimization to ensure specificity and sensitivity. The following protocol outline incorporates best practices:

Sample Preparation:

  • Treat cells with HDAC inhibitors (e.g., sodium butyrate 5-10mM for 4 hours) to preserve acetylation marks

  • Cross-link with 1% formaldehyde for 10 minutes at room temperature

  • Quench with 125mM glycine for 5 minutes

  • Wash cells in cold PBS containing protease inhibitors

Chromatin Preparation:

  • Lyse cells in appropriate buffer with protease inhibitors

  • Sonicate chromatin to fragments of 200-500bp (optimal size for sequencing)

  • Check sonication efficiency by reversing cross-links on a small aliquot and analyzing by gel electrophoresis

  • Pre-clear chromatin with protein A/G beads

Immunoprecipitation:

  • Use 5μg of Acetyl-Histone H3.1 (K14) antibody per 5-10μg of chromatin

  • Include appropriate controls (IgG negative control, total H3 for normalization)

  • Incubate overnight at 4°C with rotation

  • Add pre-blocked protein A/G beads and incubate for 2-3 hours

  • Wash stringently (low salt, high salt, LiCl, and TE washes)

DNA Recovery and Analysis:

  • Elute bound chromatin and reverse cross-links (65°C overnight)

  • Treat with RNase A and Proteinase K

  • Purify DNA using column-based methods

  • Quantify by qPCR or prepare libraries for sequencing

Critical Quality Control Measures:

  • Validate antibody specificity using peptide competition assays

  • Include spike-in controls (e.g., Drosophila chromatin) for quantitative normalization

  • Assess enrichment at known positive regions (actively transcribed genes) and negative regions (silent genes)

For ChIP-seq specifically, consider using the CUT&Tag-IT approach as an alternative to traditional ChIP-seq for higher signal-to-noise ratio and lower input requirements .

How can researchers troubleshoot and optimize Western blot experiments using Acetyl-Histone H3.1 (K14) antibodies?

Western blot analysis of H3K14ac requires careful consideration of several factors to ensure reliable results:

Common Issues and Solutions:

  • Weak or No Signal

    • Ensure proper histone extraction (acid extraction recommended)

    • Optimize antibody concentration (start with 1:500 dilution)

    • Increase protein loading (10-15μg of histone extract)

    • Use enhanced chemiluminescence (ECL) detection systems

  • High Background

    • Increase blocking time/concentration (5% BSA in TBST preferred over milk)

    • Add 0.05-0.1% SDS to washing buffer

    • Reduce primary antibody concentration

    • Ensure thorough washing (4-5 times, 5-10 minutes each)

  • Multiple Bands

    • Verify extraction quality (acid extraction minimizes contamination)

    • Check for degradation (add protease inhibitors)

    • Test specificity with peptide competition assays

Optimization Protocol:

Table 2: Systematic Optimization Parameters for Western Blot Detection of H3K14ac

ParameterTest RangeEvaluation Method
Protein Amount5-20μgBand intensity vs. background
Primary Antibody Dilution1:250 - 1:2000Signal-to-noise ratio
Secondary Antibody Dilution1:2000 - 1:10000Background level
Blocking AgentBSA vs. MilkBackground level
Incubation Time1h RT vs. Overnight 4°CSignal intensity
HDAC Inhibitor TreatmentNone vs. Sodium ButyrateSignal intensity

Special Considerations:

  • PVDF membranes generally perform better than nitrocellulose for histone detection

  • Include controls such as untreated vs. HDAC inhibitor-treated samples to verify specificity

  • Consider stripping and reprobing with total H3 antibody for normalization

  • For quantitative analysis, use a ladder of recombinant H3K14ac standards of known concentration

How can researchers validate the specificity of Acetyl-Histone H3.1 (K14) antibodies for their experimental systems?

Validating antibody specificity is essential for reliable interpretation of histone modification data. A comprehensive validation approach includes:

Peptide Competition Assays:

  • Pre-incubate antibody with increasing concentrations of acetylated K14 peptide

  • In parallel, pre-incubate with non-acetylated K14 peptide and irrelevant acetylated peptides (e.g., H3K9ac)

  • Compare binding in Western blot or ChIP to confirm specific blocking only with the target peptide

Peptide Array Analysis:

  • Test antibody against arrays containing various histone modifications

  • Calculate specificity factors (ratio of signal at target modification vs. other modifications)

  • Establish cross-reactivity profiles with similar modifications

Genetic Validation:

  • Use CRISPR/Cas9 to generate K14R mutants (prevents acetylation)

  • Compare antibody signal in wild-type vs. mutant cells

  • Use HAT/HDAC inhibitors or knockdowns to modulate acetylation levels

Orthogonal Techniques:

  • Confirm findings with mass spectrometry-based approaches

  • Use multiple antibodies from different sources targeting the same modification

  • Compare results with genetic reporter systems when available

Table 3: Specificity Analysis of Representative H3K14ac Antibodies

Antibody TypeCross-reactivity ProfileSpecificity Factor*Best Applications
Recombinant MonoclonalNo detection of K9ac, K18ac, K23ac>100ChIP-seq, Western blot
PolyclonalMinor cross-reactivity with K9ac (5-10%)15-20Western blot, IHC
Synthetic FabHighly specific, no cross-reactivity>200All applications

*Specificity factor represents the ratio of the average intensity of all spots containing the target PTM divided by the average intensity of all spots lacking that PTM in peptide array analysis .

Researchers should also be aware that neighboring modifications can affect antibody binding. For example, phosphorylation at S10 or methylation at R8 might impact the recognition of K14ac by certain antibodies. Therefore, validating the antibody in the specific experimental context is crucial.

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.