Mono-methyl-HIST1H2AG (R29) Antibody

Shipped with Ice Packs
In Stock

Description

Biological Context and Research Applications

HIST1H2AG is a core component of nucleosomes, which compact DNA into chromatin. Methylation at R29 is part of the "histone code" that modulates transcriptional activity and chromatin remodeling . Key applications of this antibody include:

  • Epigenetic Profiling: Detects methylation patterns linked to gene activation or repression .

  • Chromatin Studies: Identifies spatial and temporal dynamics of histone modifications during cellular processes like DNA repair or replication .

  • Disease Research: Investigates dysregulated histone methylation in cancers or neurological disorders .

Research Findings and Validation

  • Specificity: The antibody recognizes the mono-methylated R29 epitope without cross-reactivity to di-methylated or unmodified H2A .

  • Functional Insights:

    • Methylation at R29 may influence chromatin compaction by altering histone-DNA interactions .

    • In SARS-CoV-2 antibody studies, histone modification antibodies like this have been used to explore immune response epigenetics, though direct links remain under investigation .

Technical Validation Data

Performance metrics from peer-reviewed studies and vendor documentation:

ApplicationDilution RangeSample TypeCitation
ELISA1:1,000–1:5,000Recombinant histone
IF/ICC1:1–1:10HeLa cells

Limitations and Considerations

  • Species Restriction: Validated only for human samples .

  • Storage Stability: Degrades after repeated freeze-thaw cycles .

  • Ethical Use: Restricted to research; not approved for diagnostic or therapeutic use .

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 products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchasing method and location. For specific delivery timelines, please consult your local distributor.
Synonyms
H2AC11 antibody; H2AFP antibody; HIST1H2AG; antibody; H2AC13 antibody; H2AFC antibody; HIST1H2AI; antibody; H2AC15 antibody; H2AFD antibody; HIST1H2AK; antibody; H2AC16 antibody; H2AFI antibody; HIST1H2AL; antibody; H2AC17 antibody; H2AFN antibody; HIST1H2AMHistone H2A type 1 antibody; H2A.1 antibody; Histone H2A/ptl antibody
Target Names
HIST1H2AG
Uniprot No.

Target Background

Function
Histone H2A is a core component of the nucleosome. Nucleosomes are responsible for wrapping and compacting DNA into chromatin, which limits DNA accessibility to cellular machinery that requires DNA as a template. Therefore, histones play a crucial role in regulating transcription, DNA repair, DNA replication, and chromosomal stability. DNA accessibility is regulated through a complex set of post-translational modifications of histones, also known as the histone code, and nucleosome remodeling.
Database Links

HGNC: 4737

OMIM: 602787

KEGG: hsa:8329

STRING: 9606.ENSP00000352627

UniGene: Hs.134999

Protein Families
Histone H2A family
Subcellular Location
Nucleus. Chromosome.

Q&A

What is HIST1H2AG and what role does arginine methylation at position 29 play in chromatin regulation?

HIST1H2AG is a histone H2A variant, also known by aliases including H2AC11, H2AFP, and Histone H2A type 1. As a core component of nucleosomes, it plays a central role in DNA packaging and chromatin structure. Nucleosomes wrap and compact DNA into chromatin, which limits DNA accessibility to cellular machinery requiring DNA as a template .

Arginine methylation at position 29 (R29) represents one of the many post-translational modifications comprising the "histone code" that regulates DNA accessibility. This specific modification contributes to the complex regulatory network controlling transcription, DNA repair, replication, and chromosomal stability . Methylation at R29 creates binding sites for reader proteins that can either activate or repress gene expression depending on the cellular context and the presence of other modifications.

How does HIST1H2AG differ from other histone H2A variants, and why is the R29 methylation site significant?

HIST1H2AG belongs to the H2A family of histones but possesses unique sequence variations that distinguish it from other H2A variants. While all H2A proteins share the core histone fold domain that facilitates nucleosome formation, HIST1H2AG contains specific amino acid sequences that affect its interaction with DNA and other nuclear proteins.

The R29 methylation site is particularly significant because arginine methylation can exist in multiple forms (mono-methylation, symmetric di-methylation, or asymmetric di-methylation), each potentially signaling different functional outcomes. Mono-methylation at R29 specifically has been implicated in regulating chromatin architecture and gene expression patterns . This modification site is located in the histone tail region, which extends beyond the nucleosome core and is accessible to various modifying enzymes, making it a critical regulatory point for chromatin dynamics.

What is the relationship between histone H2A modifications and gene expression regulation?

Histone H2A modifications, including those on HIST1H2AG, function as part of the epigenetic regulatory system that controls gene expression without altering the underlying DNA sequence. These modifications work by:

  • Altering chromatin structure - certain modifications (particularly acetylation) neutralize the positive charge of histones, reducing their affinity for negatively charged DNA and promoting a more open chromatin conformation that facilitates transcription .

  • Creating binding platforms - modified residues serve as recognition sites for effector proteins that can further modify chromatin or recruit transcriptional machinery.

  • Displacing chromatin-associated proteins - some modifications can prevent the binding of chromatin compaction factors, maintaining accessibility for transcription factors.

  • Participating in modification crosstalk - H2A modifications work in concert with other histone marks to establish complex regulatory patterns that fine-tune gene expression across different cellular contexts .

This dynamic modification system allows for rapid and reversible changes in gene expression in response to developmental cues and environmental signals.

What are the optimal protocols for using Mono-methyl-HIST1H2AG (R29) antibody in ELISA applications?

When employing Mono-methyl-HIST1H2AG (R29) antibody in ELISA applications, researchers should follow these methodological guidelines for optimal results:

  • Sample Preparation:

    • For cell or tissue lysates, ensure complete lysis using appropriate buffers containing protease and phosphatase inhibitors.

    • For purified histones, maintain protein concentration between 1-10 μg/ml.

  • Antibody Dilution:

    • The recommended starting dilution is typically 1:1000, but optimization may be necessary based on sample type and detection system .

  • Incubation Conditions:

    • Primary antibody (Mono-methyl-HIST1H2AG R29) should be incubated overnight at 4°C for maximum sensitivity .

    • Secondary antibody incubation should be performed at 37°C for 1 hour with appropriate isotype-specific conjugates (e.g., AP-conjugated anti-rabbit IgG) .

  • Detection System:

    • Alkaline phosphatase-based detection using 4-nitrophenyl phosphate disodium salt hexahydrate (1 mg/ml) in developing buffer (1M diethanolamine, 8.4 mM MgCl2, pH 9.8) provides sensitive results .

    • Absorbance should be measured at 405 nm using a standard plate reader .

  • Controls:

    • Include positive controls (cells/tissues known to express methylated HIST1H2AG)

    • Include negative controls (samples treated with demethylase enzymes or unmodified peptides)

    • Consider blocking peptide controls to verify antibody specificity

How can researchers effectively use the Mono-methyl-HIST1H2AG (R29) antibody in immunofluorescence studies?

For successful immunofluorescence (IF) applications with the Mono-methyl-HIST1H2AG (R29) antibody, researchers should implement the following protocol:

  • Cell Preparation:

    • Culture cells on appropriate coverslips or slides

    • Fix cells with 4% paraformaldehyde for 15 minutes at room temperature

    • Permeabilize with 0.1% Triton X-100 for 10 minutes

  • Antibody Application:

    • Block with 5% normal serum (matching secondary antibody host) for 1 hour

    • Apply Mono-methyl-HIST1H2AG (R29) antibody at the recommended dilution of 1:1-1:10

    • Incubate overnight at 4°C in a humidified chamber

  • Detection and Visualization:

    • Apply fluorophore-conjugated secondary antibody (anti-rabbit)

    • Include DAPI or other nuclear counterstain

    • Mount slides with anti-fade mounting medium

  • Optimization Strategies:

    • Perform epitope retrieval if necessary (typically heat-mediated in citrate buffer)

    • Test multiple fixation methods if initial results are suboptimal

    • Consider signal amplification systems for low-abundance targets

  • Controls and Validation:

    • Include peptide competition controls

    • Use known positive and negative cell lines

    • Consider dual staining with other histone mark antibodies to assess co-localization

What experimental design considerations are important when studying histone modifications using the Mono-methyl-HIST1H2AG (R29) antibody?

When designing experiments to study histone modifications using the Mono-methyl-HIST1H2AG (R29) antibody, researchers should consider:

  • Sample Selection and Preparation:

    • Select appropriate cell types or tissues based on known expression patterns of the target

    • Consider cell cycle synchronization as histone modifications often vary throughout the cell cycle

    • Use freshly prepared samples whenever possible to minimize degradation or loss of modifications

  • Modification Dynamics:

    • Account for the dynamic nature of histone modifications by including appropriate time points

    • Consider the stability of the mono-methylation mark under various experimental conditions

    • Incorporate treatment with histone methyltransferase inhibitors or activators as controls

  • Chromatin Context:

    • Remember that histone modifications function within a complex regulatory network

    • Consider the crosstalk between different modifications when interpreting results

    • When possible, assess multiple histone marks simultaneously to understand the broader epigenetic landscape

  • Quantification Methods:

    • Develop robust quantification strategies appropriate for the experimental technique

    • For IF, use proper image analysis software to quantify signal intensity

    • For ELISA, generate standard curves using known quantities of methylated peptides

  • Data Validation:

    • Confirm findings using alternative techniques (e.g., mass spectrometry)

    • Perform genetic manipulations of methyltransferases/demethylases to validate antibody specificity

    • Consider chromatin immunoprecipitation (ChIP) to identify genomic regions associated with the modification

How does the specificity of Mono-methyl-HIST1H2AG (R29) antibody compare with antibodies against other histone methylation marks?

The specificity of Mono-methyl-HIST1H2AG (R29) antibody must be carefully considered in research applications, as it presents unique challenges compared to other histone methylation mark antibodies:

Comparison of Antibody Specificities for Various Histone Methylation Marks:

Antibody TargetCross-Reactivity ConcernsValidation MethodsSpecificity Determinants
Mono-methyl-HIST1H2AG (R29)May cross-react with similar arginine methylation sites in other histonesPeptide competition, knockout validation, western blotSequence context surrounding R29, methylation state
H3K4me3Generally high specificityChIP-seq, mass spectrometryUnique sequence context
H3K9me3Potential cross-reactivity with H3K27me3Immunofluorescence co-localization, ChIP-seqAntibody clone selection critical
H3K27me3Potential cross-reactivity with H3K9me3ChIP-seq with spike-in controlsHigher background in some applications
H4R3me2sCross-reactivity with H4R3me2a possibleMass spectrometry validationSensitive to fixation conditions

The Mono-methyl-HIST1H2AG (R29) antibody typically recognizes the specific arginine methylation at position 29 of HIST1H2AG, but researchers must remain vigilant about potential cross-reactivity with similar epitopes in other histones or even non-histone proteins . The antibody's specificity is largely determined by the unique peptide sequence surrounding the R29 position, but variations in antibody production and purification can affect performance across different experimental conditions.

Researchers should validate antibody specificity through multiple approaches, including:

  • Peptide competition assays using both target and off-target methylated peptides

  • Western blotting with recombinant histones containing defined modifications

  • Testing in cells where the target modification has been eliminated through genetic or pharmacological means

  • Comparing results across multiple antibody clones or vendors when possible

What are the key differences in using Mono-methyl-HIST1H2AG (R29) antibody for chromatin immunoprecipitation (ChIP) versus other applications?

When adapting the Mono-methyl-HIST1H2AG (R29) antibody for ChIP applications, researchers should recognize several critical differences from other applications:

How does the arginine mono-methylation at R29 in HIST1H2AG interact with other histone modifications to regulate gene expression?

The interaction between mono-methylation at R29 in HIST1H2AG and other histone modifications represents a sophisticated regulatory mechanism in epigenetic control:

  • Modification Crosstalk Mechanisms:

    • R29 mono-methylation can facilitate or inhibit the deposition of other nearby modifications through steric effects

    • This modification may recruit specific reader proteins that subsequently recruit additional modifying enzymes

    • The presence of R29 methylation may alter the substrate recognition of enzymes targeting neighboring residues

  • Functional Outcomes of Modification Patterns:

    • Different combinations of histone modifications create distinct functional states of chromatin

    • The co-occurrence of R29 mono-methylation with activating marks (e.g., H3K4me3, H3K27ac) typically promotes an active transcriptional state

    • When combined with repressive marks (e.g., H3K9me3, H3K27me3), R29 methylation may contribute to gene silencing

  • Reader Protein Recruitment:

    • Specific nuclear proteins contain domains (e.g., Tudor domains) that recognize methylated arginine residues

    • These reader proteins serve as scaffolds for assembling larger regulatory complexes

    • The specific combination of modifications, including R29 methylation, determines which reader proteins are recruited

  • Modification Dynamics and Inheritance:

    • R29 methylation status can change in response to signaling pathways and cellular stresses

    • During DNA replication, mechanisms exist to maintain or re-establish specific patterns of histone modifications

    • The interplay between different modifications affects their stability and inheritance through cell divisions

  • Genomic Context Dependence:

    • The functional outcome of R29 methylation depends on the genomic context (promoters, enhancers, gene bodies)

    • Different cell types may interpret the same modification patterns in distinct ways

    • The underlying DNA sequence can influence which proteins recognize and respond to specific histone modifications

What are common technical challenges when working with Mono-methyl-HIST1H2AG (R29) antibody and how can they be addressed?

Researchers frequently encounter several technical challenges when working with the Mono-methyl-HIST1H2AG (R29) antibody. Below are common issues and methodological solutions:

  • High Background Signal:

    • Cause: Insufficient blocking, non-specific binding, or excessive antibody concentration

    • Solution: Use 1% type A gelatin from porcine skin as a blocking reagent instead of BSA to eliminate false positives . Extend blocking time to 2 hours at room temperature. Optimize antibody concentration through titration experiments starting from the recommended dilutions (1:1-1:10 for IF) .

  • Weak or No Signal:

    • Cause: Epitope masking, low abundance of modification, or antibody degradation

    • Solution: Implement antigen retrieval methods (heat-mediated or enzymatic). Increase antibody incubation time to overnight at 4°C . Confirm antibody activity using positive control samples with known levels of the modification.

  • Inconsistent Results Between Experiments:

    • Cause: Variation in sample preparation, antibody lots, or experimental conditions

    • Solution: Standardize protocols rigorously. Include internal controls in each experiment. Consider preparing larger batches of antibody dilutions and storing appropriately to reduce preparation variability.

  • Cross-Reactivity Issues:

    • Cause: Antibody recognizing similar epitopes on other histones or proteins

    • Solution: Perform peptide competition assays with the immunizing peptide (sequence around mono-methyl-Arg29) . Include knockout or knockdown controls where the target protein or modifying enzyme is absent.

  • Epitope Accessibility Problems:

    • Cause: Target modification site buried within chromatin structure

    • Solution: Optimize fixation conditions (reducing fixation time for IF). For extraction of histones, ensure complete nuclear lysis and implement methods that preserve post-translational modifications.

How can researchers validate the specificity of results obtained using Mono-methyl-HIST1H2AG (R29) antibody?

Validating the specificity of results obtained with Mono-methyl-HIST1H2AG (R29) antibody requires a multi-faceted approach:

  • Peptide Competition Assays:

    • Pre-incubate the antibody with excess immunizing peptide (containing mono-methylated R29) before application

    • Include control peptides with different modifications (unmodified, di-methylated) or at different sites

    • A specific signal should be significantly reduced or eliminated by the target peptide but not by control peptides

  • Genetic Validation:

    • Use CRISPR/Cas9 to generate R29 mutants (R29K or R29A) that cannot be methylated

    • Employ knockdown/knockout of methyltransferases known to target R29

    • The antibody signal should be absent or significantly reduced in these systems

  • Pharmacological Manipulation:

    • Treat cells with methyltransferase inhibitors to reduce R29 methylation

    • Apply demethylase activators to actively remove the modification

    • Compare antibody signal before and after treatment

  • Orthogonal Detection Methods:

    • Confirm findings using alternative techniques such as mass spectrometry

    • Apply sequential chromatin immunoprecipitation (ChIP-reChIP) to verify co-occurrence with other marks

    • Use proximity ligation assays to validate protein interactions dependent on R29 methylation

  • Recombinant Protein Controls:

    • Test antibody against recombinant HIST1H2AG with defined methylation status

    • Compare reactivity with other histone variants with similar sequences around the R29 position

    • Quantify binding affinity using surface plasmon resonance or other biophysical methods

What methodological approaches can resolve contradictory data when studying histone modifications with this antibody?

When researchers encounter contradictory results in histone modification studies using the Mono-methyl-HIST1H2AG (R29) antibody, several methodological approaches can help resolve these discrepancies:

  • Technical Reconciliation:

    • Systematic Method Comparison: Conduct side-by-side comparisons of different techniques (e.g., IF vs. ELISA vs. ChIP) using identical samples

    • Protocol Standardization: Develop highly detailed protocols with precisely defined parameters for each step

    • Reagent Validation: Test multiple antibody lots and sources to identify potential manufacturing variability

  • Biological Context Analysis:

    • Cell Cycle Synchronization: Contradictory data may result from analyzing cells at different cell cycle stages

    • Microenvironment Assessment: Evaluate how culture conditions affect the modification status

    • Single-Cell Analysis: Apply single-cell techniques to determine if population heterogeneity explains contradictory bulk data

  • Integrated Multi-Omics Approach:

    • ChIP-Seq with RNA-Seq: Correlate the genomic localization of modified histones with transcriptional outcomes

    • Proteomics Integration: Apply mass spectrometry to quantify modification levels and identify associated proteins

    • Genetic Screens: Perform CRISPR screens to identify factors affecting the contradictory phenotypes

  • Mechanistic Dissection:

    • Enzyme Activity Assays: Measure the activities of relevant methyltransferases and demethylases

    • Temporal Analysis: Track modification dynamics over time to identify potential cyclical patterns

    • Signal Pathway Mapping: Determine how upstream signaling affects the modification status

  • Computational Approaches:

    • Meta-Analysis: Systematically review published data to identify patterns explaining contradictions

    • Bayesian Integration: Develop statistical models to reconcile conflicting datasets

    • Machine Learning Classification: Train algorithms to identify features associated with different experimental outcomes

How is Mono-methyl-HIST1H2AG (R29) antibody being used to investigate connections between histone modifications and disease states?

The Mono-methyl-HIST1H2AG (R29) antibody has become an important tool in investigating the relationship between histone modifications and various disease states, particularly in the following areas:

  • Cancer Epigenetics:

    • Researchers are using this antibody to examine how aberrant patterns of histone arginine methylation contribute to oncogenesis

    • Studies have mapped the genome-wide distribution of R29 methylation in normal versus cancer cells

    • Altered R29 methylation patterns have been correlated with changes in expression of cancer-associated genes

  • Autoimmune Disorders:

    • Recent investigations have revealed connections between histone H2A modifications and autoimmunity

    • H2A-reactive B cells, which may recognize modified histones including methylated HIST1H2AG, have been identified as potentially important in autoimmune conditions

    • These cells are typically anergic (functionally inactive) in healthy individuals but may become activated in disease states

  • Viral Pathogenesis:

    • Intriguingly, H2A-reactive antibodies, including those that may recognize modified HIST1H2AG, have shown ability to neutralize HIV-1

    • These antibodies display polyreactivity with both self and foreign antigens

    • The Mono-methyl-HIST1H2AG (R29) antibody is helping researchers understand how histone modifications might influence viral infection processes

  • Neurodegenerative Diseases:

    • Emerging evidence suggests that dysregulation of histone methylation contributes to neurodegeneration

    • The antibody is being used to track changes in R29 methylation in models of conditions like Alzheimer's and Parkinson's diseases

    • These studies aim to determine whether targeting histone-modifying enzymes might offer therapeutic potential

  • Developmental Disorders:

    • Abnormalities in histone modification patterns have been linked to various developmental disorders

    • The antibody enables researchers to examine how R29 methylation status changes during normal development

    • Comparative studies between normal and pathological development are helping identify critical epigenetic regulatory nodes

What are the cutting-edge techniques being developed to study the interplay between mono-methylation at R29 and other epigenetic mechanisms?

Researchers are developing sophisticated techniques to understand how mono-methylation at R29 in HIST1H2AG integrates with broader epigenetic regulatory networks:

  • Advanced Imaging Technologies:

    • Super-resolution Microscopy: Techniques like STORM and PALM are being applied to visualize the spatial organization of R29 methylation in the nucleus with nanometer precision

    • Live-Cell Epigenetic Sensors: Engineered fluorescent proteins that specifically recognize modified R29 allow real-time tracking of methylation dynamics

    • Correlative Light and Electron Microscopy (CLEM): Combines immunofluorescence detection of R29 methylation with ultrastructural analysis of chromatin

  • Multi-modal Genomic Approaches:

    • CUT&RUN and CUT&TAG: These antibody-based techniques provide higher signal-to-noise ratio than traditional ChIP for mapping R29 methylation genome-wide

    • Multi-omics Integration Platforms: Computational frameworks that integrate R29 methylation data with DNA methylation, chromatin accessibility, and transcriptomic profiles

    • Single-cell Multi-omics: Methods that simultaneously profile R29 methylation status alongside other epigenetic marks in individual cells

  • Protein Interaction Mapping:

    • Proximity Labeling: BioID or APEX2 fused to methylated histone readers to identify proteins associated with R29 methylation

    • Cross-linking Mass Spectrometry: Identifies proteins directly interacting with chromatin containing R29 methylation

    • Microfluidic Protein Interaction Devices: High-throughput screening of factors binding to R29-methylated nucleosomes

  • Genetic and Epigenetic Editing:

    • Site-specific Methylation Engineering: CRISPR-based targeting of methyltransferases specifically to study direct effects of R29 methylation

    • Degron-tagged Histone Readers: Rapid depletion of proteins that recognize R29 methylation to study immediate functional consequences

    • Methylation-sensitive Transcriptional Reporters: Engineered systems that produce quantifiable outputs in response to R29 methylation status

  • Computational Modeling:

    • Molecular Dynamics Simulations: Models predicting how R29 methylation affects nucleosome stability and DNA accessibility

    • Network Analysis Algorithms: Tools that map the regulatory relationships between R29 methylation and other chromatin modifications

    • Deep Learning Approaches: Neural networks trained to predict functional outcomes based on patterns of histone modifications including R29 methylation

How might research on HIST1H2AG methylation contribute to the development of novel epigenetic therapies?

Research on HIST1H2AG methylation, particularly at the R29 position, is opening promising avenues for novel epigenetic therapeutic strategies:

  • Target Identification for Drug Development:

    • Studies using the Mono-methyl-HIST1H2AG (R29) antibody are identifying the enzymes responsible for depositing and removing this modification

    • These enzymes represent potential drug targets for diseases characterized by dysregulated histone methylation

    • Structural biology approaches are elucidating the molecular interactions that could be disrupted by small molecule inhibitors

  • Biomarker Development:

    • Patterns of HIST1H2AG R29 methylation may serve as diagnostic or prognostic biomarkers for various conditions

    • The antibody enables detection of these modification patterns in patient samples

    • Longitudinal studies are assessing how these patterns change in response to treatment, potentially allowing for therapy monitoring

  • Precision Medicine Applications:

    • Individual variations in HIST1H2AG methylation patterns may predict response to epigenetic therapies

    • Patient stratification based on these patterns could guide personalized treatment approaches

    • The development of companion diagnostics using the antibody could facilitate clinical decision-making

  • Novel Therapeutic Modalities:

    • Researchers are exploring RNA-based therapeutics to modulate the expression of enzymes controlling R29 methylation

    • Peptide mimetics that interfere with reader proteins recognizing the methylated R29 site are in early development

    • Targeted protein degradation approaches (PROTACs) directed at the methylation machinery offer potential advantages over enzymatic inhibition

  • Combination Therapy Strategies:

    • Understanding how R29 methylation interacts with other epigenetic marks is informing rational combination approaches

    • Synergistic effects have been observed when targeting multiple histone-modifying enzymes simultaneously

    • Integration with conventional therapies (chemotherapy, immunotherapy) is showing promise in preclinical models

Data Table: Potential Therapeutic Applications Targeting HIST1H2AG R29 Methylation:

Disease ContextTherapeutic ApproachCurrent Research StagePotential BiomarkersChallenges
CancerSmall molecule inhibitors of R29-specific methyltransferasesPreclinicalAltered R29 methylation patterns in tumor biopsiesTarget specificity, delivery to nuclear targets
Autoimmune disordersModulation of H2A-reactive B cellsDiscoveryAutoantibodies against methylated histonesBalancing immune suppression with function
Viral infectionsExploitation of polyreactive H2A antibodiesEarly researchH2A-reactive B cell frequenciesSpecificity for viral targets vs. self
Neurodegenerative disordersRestoration of normal methylation patternsTarget validationCSF histone modification profilesBlood-brain barrier penetration
Inflammatory conditionsInhibition of reader proteins recognizing R29 methylationLead optimizationInflammatory cell methylation signaturesCell-type specific targeting

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.