CDYL Antibody (e.g., ab5188) is a rabbit polyclonal antibody that recognizes the CDYL protein across multiple species, including humans, mice, rats, and vervet monkeys . CDYL is a chromodomain-containing transcriptional corepressor that interacts with histone-modifying enzymes (e.g., G9a, SETDB1, EZH2) and chromatin-associated complexes (e.g., CAF-1, PRC2) to regulate repressive histone marks such as H3K9me2/3 and H3K27me3 .
The antibody detects both endogenous and overexpressed CDYL, with validation data showing specific binding in transfected COS-7 cells expressing HA-CDYL-FLAG constructs .
CDYL Antibody has been utilized in diverse experimental contexts:
Chromatin Interaction Studies: Identified CDYL’s association with CAF-1 and MCM complexes during DNA replication, facilitating histone H3 deposition .
Neuronal Excitability Research: Demonstrated CDYL’s role in suppressing epileptogenesis by regulating ion channel genes (e.g., SCN8A) via histone modifications .
Transcriptional Repression Analysis: Revealed CDYL’s interaction with PRC2 to propagate H3K27me3 marks, forming a feedback loop for epigenetic silencing .
CDYL bridges CAF-1 and MCM complexes at replication forks, enabling the transfer of repressive histone marks (H3K9me2/3, H3K27me2/3) to newly synthesized chromatin .
Depletion of CDYL disrupts S-phase progression and sensitizes cells to DNA damage .
Knockdown of CDYL in hippocampal neurons lowers action potential thresholds and increases neuronal excitability, implicating it in epilepsy .
Overexpression of CDYL in transgenic mice reduces neuronal firing by modulating ion channel expression (e.g., SCN8A, GABRB3) .
CDYL recruits histone deacetylases (HDACs) and PRC2 to repress target genes such as E-cadherin and BDNF .
Genome-wide ChIP-seq analysis identified 16,756 CDYL-binding sites, primarily in gene bodies and regulatory regions of ion channels and synaptic proteins .
Western Blot: Detects multiple isoforms (60–80 kDa) due to splice variants or post-translational modifications .
ChIP Specificity: Validated in SY5Y cells, where CDYL binding at the SCN8A intron correlates with H3K9me3 enrichment .
Limitations: Cross-reactivity with non-target proteins may occur in tissues with low CDYL expression.
CDYL is a chromatin reader protein that recognizes and binds to histone H3 trimethylated at lysine 9 (H3K9me3), dimethylated at lysine 27 (H3K27me2), and trimethylated at lysine 27 (H3K27me3). It is a component of multimeric repressive chromatin complexes, essential for the inheritance and restoration of repressive histone marks, thereby maintaining epigenetic stability. CDYL is crucial for the chromatin targeting and maximal enzymatic activity of Polycomb repressive complex 2 (PRC2), acting as a positive regulator by bridging pre-existing H3K27me3 and newly recruited PRC2 on adjacent nucleosomes. It also functions as a corepressor for REST, facilitating histone-lysine N-methyltransferase EHMT2 recruitment and H3K9me2 at REST target genes to repress transcription. CDYL is involved in X chromosome inactivation in females, recruited to Xist RNA-coated X chromosomes to facilitate H3K9me2 propagation via EHMT2 anchoring. Furthermore, CDYL promotes EZH2 accumulation and H3K27me3 methylation at DNA double-strand breaks (DSBs), facilitating transcriptional repression at damage sites and homology-directed DSB repair. It plays a critical role in neuronal migration during brain development by repressing RHOA expression and contributes to the inhibition of intrinsic neuronal excitability and epileptogenesis by repressing SCN8A. Beyond its chromatin reader function, CDYL acts as a hydrolase, exhibiting crotonyl-CoA hydratase activity. This activity converts crotonyl-CoA to β-hydroxybutyryl-CoA, negatively regulating histone crotonylation. This regulation of histone crotonylation, along with H3K27 trimethylation, is implicated in stress-induced depression-like behaviors, potentially via VGF expression modulation. Variants lacking the chromo domain, or possessing an N-terminal extension, show impaired binding to H3K9me3, H3K27me2, and H3K27me3. CDYL also negatively regulates isoform 2 by displacing it from chromatin.
CDYL is a chromodomain-containing transcriptional corepressor that plays multiple roles in chromatin modification and gene regulation. The protein functions primarily as a chromatin reader that recognizes and binds to specific histone modifications, particularly H3K9me3, H3K27me2, and H3K27me3 .
CDYL has several critical biological functions including:
Functioning as a transcriptional co-repressor by recruiting histone deacetylases (HDACs)
Participating in multimeric repressive chromatin complexes to preserve epigenetic landscapes
Regulating neuronal excitability and nociception (pain sensing)
Promoting chemoresistance in certain cancers, such as small cell lung cancer
CDYL accomplishes these functions through its dual domain structure: a chromodomain that binds methylated histones and a CoA-pocket domain that interacts with histone-modifying enzymes like HDACs and EZH2.
Research indicates three main CDYL isoforms (variants) with distinct functions:
These isoforms can be studied using specific primers for PCR amplification. The different variants appear to have specialized functions, as demonstrated by their distinct cellular distributions and binding partners . When designing experiments that target CDYL, researchers should consider which isoform(s) may be relevant to their specific research questions.
CDYL antibodies have been validated for multiple experimental applications according to the available data:
When working with CDYL antibodies, researchers should optimize conditions based on their specific experimental system. For instance, in immunofluorescence assays, CDYL typically shows nuclear localization in a characteristic punctate pattern that may colocalize with heterochromatin markers .
ChIP experiments with CDYL antibodies require specific optimization due to CDYL's function as a chromatin-binding protein:
Crosslinking optimization: Standard 1% formaldehyde for 10 minutes is a good starting point, but optimization may be needed based on your cell type.
Sonication parameters: Aim for chromatin fragments of 200-500bp for optimal CDYL ChIP results.
Antibody selection: Use ChIP-validated antibodies such as ab5188 , which has been cited in multiple publications for this application.
Controls to include:
Analysis of histone marks: Consider performing parallel ChIPs for histone marks associated with CDYL binding (H3K27me3, H3K9me2) to validate your results.
In a recent study, CDYL was found to bind to the intron region of Kcnb1, where it was associated with increased H3K27me3 levels leading to transcriptional silencing . This provides a positive control region that can be used to validate CDYL ChIP experiments.
CDYL functions as a bridge between chromatin and histone-modifying enzymes. Several methodological approaches can be used to study these interactions:
Co-immunoprecipitation (Co-IP):
GST pull-down assays:
Immunofluorescence co-localization:
In vivo validation:
These interactions are functionally significant, as CDYL's binding to HDACs is mutually exclusive with its CoA-binding activity, suggesting a regulatory mechanism for CDYL function switching between transcriptional repression and other potential roles .
To investigate CDYL's role in gene silencing, researchers can employ several complementary approaches:
ChIP-seq analysis:
Knockout/knockdown validation:
Histone modification analysis:
Functional assays:
In one study, researchers identified that CDYL transcriptionally silences Kv2.1 by recruiting H3K27me3 activity at its intron region, demonstrating how these approaches can be integrated to establish CDYL's role in specific gene silencing mechanisms .
Recent advances have led to the development of specific CDYL antagonists that can be used to probe CDYL function:
Available inhibitors:
Selectivity profile:
Assessing inhibitor efficacy:
Time-resolved fluorescence resonance energy transfer (TR-FRET) assays can be used to measure binding of compounds to the CDYL chromodomain .
Functional assays such as gene expression analysis, ChIP for H3K27me3 at CDYL target genes, or phenotypic assays (e.g., neuronal excitability) can assess the cellular effects of CDYL inhibition.
Experimental design considerations:
Include appropriate controls (vehicle, inactive analog like UNC7394)
Perform dose-response studies to establish IC50 values in cellular contexts
Validate on-target effects through genetic approaches (e.g., comparing inhibitor effects in wild-type vs. CDYL knockout cells)
These tools enable researchers to probe CDYL function pharmacologically, potentially offering new therapeutic strategies for conditions where CDYL dysfunction plays a role.
Researchers frequently encounter several technical issues when working with CDYL antibodies:
Nuclear protein extraction efficiency:
Specificity across CDYL isoforms:
Cross-reactivity with related proteins:
Fixation sensitivity in immunofluorescence:
Batch-to-batch variability:
Thorough validation of CDYL antibodies is critical for reliable experimental results:
Western blot validation:
Genetic validation approaches:
Compare signal in wild-type vs. CDYL knockout or knockdown samples
Overexpression of tagged CDYL constructs to confirm antibody detection capability
Peptide competition assays:
Pre-incubate antibody with immunizing peptide before application to samples
A specific antibody will show diminished signal when pre-blocked with its target peptide
Cross-validation with different antibodies:
Application-specific validation:
Proper validation ensures experimental reproducibility and reliability when working with CDYL antibodies.
Appropriate experimental controls are essential when investigating CDYL function across different biological systems:
Cell type-specific expression controls:
Subcellular localization controls:
Genetic modification controls:
Experimental readout controls:
For transcriptional repression assays, include positive controls with known repressors
For HDAC recruitment studies, include other HDAC-interacting proteins as comparisons
Animal model controls:
In one study examining CDYL's role in pain, researchers used a conditional knockout mouse model (Cdyl cKO) and included comprehensive experimental controls to demonstrate that CDYL deficiency reduced nociception through regulation of Kv2.1 channel expression .
Recent research has revealed several disease-relevant functions of CDYL that may have therapeutic implications:
Cancer biology:
CDYL is upregulated in chemoresistant small cell lung cancer (SCLC) tissues and correlates with advanced clinical stage and poor prognosis
Mechanistically, CDYL promotes SCLC chemoresistance by silencing CDKN1C through recruitment of EZH2 and regulation of H3K27me3
Potential therapeutic approach: CDYL inhibition might sensitize resistant tumors to chemotherapy
Neurological disorders:
CDYL deficiency affects neuronal excitability and reduces susceptibility to epilepsy
Mechanistically, CDYL transcriptionally silences Kv2.1 potassium channels by recruiting H3K27me3 activity
Potential therapeutic approach: CDYL antagonists like UNC6261 might have applications in pain management and epilepsy treatment
Neurodevelopmental processes:
Reproductive biology:
These emerging roles highlight the importance of developing specific modulators of CDYL function for both research tools and potential therapeutic applications.
CDYL and CDYL2 are related chromodomain proteins with distinct but overlapping functions:
Structural and functional differences:
Experimental methods to distinguish between CDYL and CDYL2:
Inhibitor selectivity:
Response to stimuli:
Different expression changes or subcellular relocalization in response to specific stimuli may help distinguish functional roles
For example, in neuropathic pain models, examining changes in both CDYL and CDYL2 could reveal different roles
Understanding the distinct functions of these related proteins will require careful experimental design with appropriate controls to ensure specificity.
Several innovative approaches are advancing our understanding of CDYL function in chromatin biology:
CUT&RUN and CUT&Tag:
These techniques offer advantages over traditional ChIP-seq for mapping CDYL binding sites with higher resolution and lower background
They require fewer cells and can provide more precise footprinting of CDYL-chromatin interactions
Proximity labeling approaches:
BioID or APEX2 fusions with CDYL can identify protein interaction networks in living cells
This approach could reveal cell type-specific or context-dependent CDYL interactors beyond known partners like HDAC1/2 and EZH2
Single-cell technologies:
Single-cell RNA-seq combined with CDYL perturbation can reveal cell type-specific responses
Single-cell ATAC-seq after CDYL modulation can map chromatin accessibility changes
CRISPR-based approaches:
CRISPRi targeted to CDYL binding sites can help establish functional importance
CRISPR activation/repression of CDYL can reveal dose-dependent effects
CRISPR screens for synthetic lethality with CDYL inhibition can identify potential combination therapeutic targets
Live-cell imaging of chromatin dynamics:
FRAP (Fluorescence Recovery After Photobleaching) with fluorescently tagged CDYL can assess dynamics of chromatin binding
Real-time visualization of CDYL recruitment during processes like DNA damage response or X chromosome inactivation
Chemical biology tools:
These emerging methodologies will help advance our understanding of CDYL's complex roles in chromatin regulation and may lead to novel therapeutic strategies targeting CDYL-dependent processes.