CDYL Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to CDYL Antibody

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 .

Key Features of CDYL Antibody

PropertyDetails
Host SpeciesRabbit
ReactivityHuman, Mouse, Rat, Vervet Monkey
ApplicationsWestern Blot (WB), Immunohistochemistry (IHC-P), Chromatin Immunoprecipitation (ChIP), ChIP-seq
Observed Bands60 kDa, 65 kDa, 80 kDa (varies by post-translational modifications)
ValidationConfirmed in transfected COS-7 cells and native tissues

The antibody detects both endogenous and overexpressed CDYL, with validation data showing specific binding in transfected COS-7 cells expressing HA-CDYL-FLAG constructs .

Research Applications and Experimental Validation

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 .

Epigenetic Regulation

  • 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 .

Neurological Function

  • 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) .

Transcriptional Repression Mechanisms

  • 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 .

Technical Validation and Limitations

  • 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.

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Stored at -20°C. Avoid freeze-thaw cycles.
Lead Time
Orders are typically shipped within 1-3 business days of receipt. Delivery times may vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Synonyms
CDY-like antibody; CDY-like autosomal antibody; Cdyl antibody; CDYL: chromodomain protein Y-like antibody; CDYL1 antibody; CDYL1_HUMAN antibody; chromodomain protein Y chromosome-like 6 antibody; Chromodomain protein Y like antibody; Chromodomain Y-like protein antibody; DKFZP586C1622 antibody; MGC131936 antibody; OTTHUMP00000015984 antibody; OTTHUMP00000211185 antibody; OTTHUMP00000211186 antibody; OTTHUMP00000211189 antibody; testis-specific chromodomain Y-like protein antibody; Weakly similar to enoyl-CoA hydratase [H.sapiens] antibody
Target Names
CDYL
Uniprot No.

Target Background

Function

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.

Gene References Into Functions
  1. CDYL's role in maintaining repressive histone marks during replication elucidates epigenetic inheritance and memory. PMID: 28402439
  2. CDYL's negative regulation of histone crotonylation through crotonyl-CoA hydratase activity suggests a role in spermatogenesis. PMID: 28803779
  3. CDYL and EZH2's influence on dendrite morphogenesis provides insight into neurodevelopmental regulation. PMID: 24671995
  4. h-CDYLb and G9a's cooperative involvement in hepatocellular carcinomas. PMID: 23629948
  5. CDYL acts as a molecular bridge between PRC2 and the H3K27me3 mark, forming a positive feedback loop for H3K27me3 establishment and propagation. PMID: 22009739
  6. Multimeric H3K9me3 binding by CDYL1b complexes is essential for efficient chromatin targeting. PMID: 19808672
  7. CDYL functions as a REST corepressor, bridging REST and G9a to repress transcription. PMID: 19061646
Database Links

HGNC: 1811

OMIM: 603778

KEGG: hsa:9425

STRING: 9606.ENSP00000380718

UniGene: Hs.269092

Subcellular Location
[Isoform 2]: Nucleus. Chromosome.
Tissue Specificity
Expressed in the hippocampus with reduced expression in epileptic tissue compared to normal adjacent tissue (at protein level). Ubiquitous. Expressed at moderate levels in all tissues examined. Isoform 2: Most abundantly expressed isoform.

Q&A

What is CDYL and what are its key biological functions?

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:

  • Acting as a partner of the inactive X chromosome

  • Functioning as a transcriptional co-repressor by recruiting histone deacetylases (HDACs)

  • Participating in multimeric repressive chromatin complexes to preserve epigenetic landscapes

  • Contributing to X chromosome inactivation in females

  • 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.

What are the different isoforms of CDYL and how do they differ functionally?

Research indicates three main CDYL isoforms (variants) with distinct functions:

IsoformDescriptionKey FeaturesFunction
Cdyla (CDYL1)Full-length isoformContains both chromodomain and CoA-pocket domainPrimary transcriptional co-repressor
CdylbVariantModified domain structureMay have distinct binding properties
CdylcVariantModified domain structureMay have distinct binding properties

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.

What are the validated applications for CDYL antibodies in research?

CDYL antibodies have been validated for multiple experimental applications according to the available data:

ApplicationValidation StatusKey Considerations
Western Blotting (WB)Validated Typical dilutions: 1:500-1:2000
Immunoprecipitation (IP)Validated Useful for protein-protein interaction studies
Immunofluorescence (IF)Validated Shows nuclear localization
Chromatin Immunoprecipitation (ChIP)Validated Essential for studying CDYL binding to chromatin
ChIP-seqValidated For genome-wide analysis of CDYL binding sites
Immunohistochemistry-Paraffin (IHC-P)Validated For tissue distribution studies

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 .

How can I optimize ChIP experiments using CDYL antibodies?

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:

    • Input chromatin control

    • IgG negative control

    • Positive control targeting known CDYL binding sites such as the Kcnb1 intron region (+897 to +1143 bp)

  • 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.

How does CDYL interact with histone-modifying enzymes and what methods can detect these interactions?

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):

    • CDYL has been shown to interact with HDAC1 and HDAC2 (but not HDAC3) through its CoA-pocket domain .

    • FLAG-tagged CDYL constructs can be immunoprecipitated using anti-FLAG antibodies, followed by western blotting for HDACs .

  • GST pull-down assays:

    • The CoA-pocket domain of CDYL specifically interacts with HDAC1, while the chromodomain does not .

    • GST fusion proteins containing either the chromodomain or CoA-pocket domain can be used for in vitro binding assays with labeled HDAC1 .

  • Immunofluorescence co-localization:

    • Co-expression of CDYL with HDAC1/2 induces the recruitment of these HDACs into CDYL-specific nuclear spots .

  • In vivo validation:

    • For physiological relevance, endogenous interactions can be confirmed in tissues with high CDYL expression, such as spermatid cells .

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 .

What experimental approaches can determine if CDYL is involved in specific gene silencing mechanisms?

To investigate CDYL's role in gene silencing, researchers can employ several complementary approaches:

  • ChIP-seq analysis:

    • Identify genome-wide binding sites of CDYL, as demonstrated in studies of DRG neurons where CDYL was found to bind to genes including Kcnb1, Npas4, and Nsf .

    • Compare binding profiles with histone modifications (H3K27me3, H3K9me2/3) to identify correlations .

  • Knockout/knockdown validation:

    • Generate CDYL knockout or knockdown models (e.g., Cdyl cKO mice) to assess changes in target gene expression .

    • Quantify mRNA levels of potential target genes in wild-type vs. knockout conditions using qRT-PCR .

  • Histone modification analysis:

    • Perform ChIP-qPCR at CDYL binding sites to assess changes in histone modifications (H3K27me3, H3K9me2, H3K9me3, H3K27ac) in the presence or absence of CDYL .

    • A decrease in H3K27me3 levels was observed at the Kcnb1 gene in Cdyl cKO mice, supporting CDYL's role in establishing this repressive mark .

  • Functional assays:

    • Assess the functional consequences of CDYL-mediated gene silencing using appropriate cellular or animal models.

    • For example, CDYL deficiency was shown to brake neuronal excitability and nociception through its effects on Kv2.1 channel expression .

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 .

How can CDYL function be pharmacologically modulated and how might I assess the efficacy of inhibitors?

Recent advances have led to the development of specific CDYL antagonists that can be used to probe CDYL function:

  • Available inhibitors:

    • UNC6261 is a potent CDYL antagonist that binds to the CDYL chromodomain with an IC50 of 81 ± 16 nM and a Kd of 139 ± 3.3 nM .

    • UNC7394 serves as a structurally similar negative control compound with no measurable binding to CDYL .

  • Selectivity profile:

    • UNC6261 shows 13-fold selectivity for CDYL over the closely related protein MPP8, and more than 45-fold selectivity over members of the HP1 and Polycomb family of chromodomains .

  • 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.

What are common technical challenges when using CDYL antibodies, and how can these be addressed?

Researchers frequently encounter several technical issues when working with CDYL antibodies:

  • Nuclear protein extraction efficiency:

    • CDYL is primarily nuclear and tightly associated with chromatin

    • The CoA-pocket domain alone may remain tightly associated with insoluble nuclear materials after extraction

    • Solution: Use specialized nuclear extraction buffers containing DNase or higher salt concentrations

  • Specificity across CDYL isoforms:

    • Multiple CDYL isoforms exist (Cdyla, Cdylb, Cdylc)

    • Solution: Verify antibody epitopes to determine which isoforms are recognized; use western blotting to confirm detection of expected bands (approximately 62 kDa for the main isoform)

  • Cross-reactivity with related proteins:

    • CDYL has a related family member CDYL2

    • Solution: Validate antibody specificity using knockout/knockdown controls or peptide competition assays

  • Fixation sensitivity in immunofluorescence:

    • Some epitopes may be sensitive to particular fixation methods

    • Solution: Compare different fixation protocols (paraformaldehyde, methanol, or digitonin permeabilization as described in )

  • Batch-to-batch variability:

    • Traditional polyclonal antibodies may show lot-to-lot variations

    • Solution: Consider using recombinant monoclonal antibodies like CDYL (E7S8T) Rabbit mAb for superior lot-to-lot consistency

How can I validate CDYL antibody specificity for my experimental system?

Thorough validation of CDYL antibodies is critical for reliable experimental results:

  • Western blot validation:

    • Verify the detection of bands at expected molecular weights (approximately 62 kDa)

    • Test in multiple cell lines known to express CDYL (e.g., 293T, A431, HeLa, HepG2)

    • Include positive controls such as U-251MG cells, which have been validated for CDYL expression

  • 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:

    • Compare results using antibodies targeting different epitopes of CDYL

    • Consider testing both polyclonal (e.g., ab5188 ) and monoclonal (e.g., E7S8T ) antibodies

  • Application-specific validation:

    • For ChIP applications, verify enrichment at known CDYL binding sites such as the Kcnb1 intron region

    • For IF, confirm expected nuclear localization and colocalization with appropriate markers

Proper validation ensures experimental reproducibility and reliability when working with CDYL antibodies.

How should experimental controls be designed when studying CDYL in different cellular contexts?

Appropriate experimental controls are essential when investigating CDYL function across different biological systems:

  • Cell type-specific expression controls:

    • CDYL shows differential expression across cell types (e.g., highly expressed in spermatids)

    • Include RNA and protein expression analysis to establish baseline CDYL levels in your system

  • Subcellular localization controls:

    • CDYL primarily localizes to the nucleus, often in specific nuclear spots

    • Include nuclear markers and heterochromatin markers to validate expected localization patterns

  • Genetic modification controls:

    • When using CDYL knockout or knockdown models, include rescue experiments with wild-type CDYL

    • For domain-specific functions, use rescue with specific CDYL mutants (e.g., CoA-binding mutants N441A and K463A that lose CoA binding but retain repressor activity)

  • 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:

    • For Cdyl conditional knockout mice (cKO), carefully establish appropriate littermate controls

    • Consider tissue-specific knockout strategies when studying specialized functions (e.g., DRG-specific deletion for pain studies)

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 .

What are the emerging roles of CDYL in disease states and potential therapeutic applications?

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:

    • CDYL is required for neuronal migration during brain development by repressing RHOA expression

    • CDYL contributes to inhibition of intrinsic neuronal excitability by repressing SCN8A expression

    • Potential research direction: Further characterization of CDYL's role in neurodevelopmental disorders

  • Reproductive biology:

    • CDYL shows hydro-lyase activity, mediating the conversion of crotonyl-CoA to beta-hydroxybutyryl-CoA, thereby negatively regulating histone crotonylation, which is important during spermatogenesis

    • Future research might explore CDYL's role in male fertility disorders

These emerging roles highlight the importance of developing specific modulators of CDYL function for both research tools and potential therapeutic applications.

How do CDYL and CDYL2 differ functionally, and what methods can distinguish between them in experimental settings?

CDYL and CDYL2 are related chromodomain proteins with distinct but overlapping functions:

  • Structural and functional differences:

    • Both contain chromodomains that bind methylated histones, but may have different binding preferences

    • CDYL2 interacts with histone methyltransferases and deacetylases to establish repressive chromatin environments

    • Different tissue distribution patterns may suggest specialized functions

  • Experimental methods to distinguish between CDYL and CDYL2:

    ApproachMethodologyNotes
    Antibody specificityUse validated antibodies specific to each proteinCDYL: ab5188 , E7S8T ; CDYL2: ab183854
    Western blottingCDYL runs at ~62 kDa; CDYL2 at ~57 kDaAlways include molecular weight markers
    qRT-PCRDesign primers specific to unique regionsValidate primer specificity with overexpression controls
    ChIP-seqMap genome-wide binding profilesMay reveal distinct chromatin-binding preferences
    Knockout modelsGenerate specific knockout cell lines for eachCRISPR-Cas9 targeting of unique exons
  • Inhibitor selectivity:

    • UNC6261 shows binding to CDYL2 with an IC50 of 81 ± 16 nM and to CDYL with a Kd of 139 ± 3.3 nM

    • Further development of selective inhibitors may help distinguish functions

  • 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.

What cutting-edge methodologies are emerging for studying CDYL's role in chromatin regulation and gene silencing?

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:

    • Development of PROTAC (PROteolysis TArgeting Chimera) molecules targeting CDYL could allow temporal control of protein degradation

    • The recently developed CDYL antagonist UNC6261 provides a foundation for creating additional chemical probes

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.

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.