CHD4 monoclonal antibody is a highly specific laboratory reagent designed to detect chromodomain helicase DNA-binding protein 4 (CHD4), a critical component of the nucleosome remodeling and deacetylase (NuRD) complex. This antibody enables researchers to study CHD4's roles in chromatin remodeling, transcriptional regulation, and DNA repair across experimental models .
Radioresistance in HPV-negative HNSCC: High CHD4 expression correlates with poor survival in HPV-negative head and neck squamous cell carcinoma. Knockdown via CRISPR/Cas9 enhances radiosensitivity, suggesting CHD4 as a therapeutic target .
Super-Enhancer Regulation: CHD4 maintains chromatin accessibility at super-enhancers in fusion-positive rhabdomyosarcoma, enabling oncogene expression (e.g., PAX3-FOXO1) .
B Cell Development: CHD4 is essential for pre-B cell maturation but dispensable for naive B cell homeostasis. Its loss impairs class-switch recombination by reducing AID recruitment to the Igh locus .
NuRD Complex Function: CHD4 collaborates with HDAC1/2 to deacetylate histones, repress transcription, and regulate DNA damage repair .
Vendor | Clone | Host | Applications | Catalog Number |
---|---|---|---|---|
Cell Signaling Tech | D4B7 | Rabbit | WB, IF, ChIP | #12011 |
Proteintech | 66222-1-Ig | Mouse | WB, IHC, IF/ICC | 66222-1-Ig |
EMD Millipore | 3F2/4 | Mouse | WB, IHC, IF | MABE455 |
Assay Genie | ARC59887 | Rabbit | IHC, ELISA | CAB23537 |
CHD4 is implicated in tumor survival across >90% of cancer cell lines screened (CRISPR/RNAi datasets) . Its dual role in chromatin remodeling and DNA repair positions it as a potential target for:
CHD4 (Chromodomain-helicase-DNA-binding protein 4) is an ATP-dependent helicase that binds and distorts nucleosomal DNA, acting as a key component of the histone deacetylase NuRD complex involved in chromatin remodeling . It plays critical roles in transcriptional repression, DNA damage repair, and neurogenesis. CHD4's significance stems from its fundamental role in epigenetic regulation and its dysregulation in various pathological conditions, particularly cancer . Research into CHD4 provides insights into basic mechanisms of gene expression control, DNA repair pathways, and potential therapeutic targets for diseases associated with chromatin dysregulation .
CHD4 monoclonal antibodies can be reliably employed in multiple research techniques including:
Immunohistochemistry (IHC-P) with formalin-fixed paraffin-embedded tissues
Chromatin immunoprecipitation (ChIP) for studying CHD4-DNA interactions
Immunoprecipitation for protein complex isolation and characterization
Each application requires specific optimization of antibody dilution, with recommended IHC-P dilutions typically in the range of 1:50-1:200 .
For optimal CHD4 detection in immunohistochemistry:
Start with standard formalin fixation (10% neutral buffered formalin for 24-48 hours)
Implement heat-mediated antigen retrieval using sodium citrate buffer (pH 6.0)
Test multiple antibody dilutions (starting with the manufacturer's recommended range of 1:50-1:200)
Include both positive controls (tissues known to express CHD4, such as breast carcinoma) and negative controls (primary antibody omission)
If signal is weak, extend the primary antibody incubation time (overnight at 4°C)
For specialized applications, compare different fixatives (paraformaldehyde, methanol) to determine optimal epitope preservation
Remember that CHD4 has dual localization in the nucleus and cytoplasm, with additional specific localization to centrosomes and microtubule organizing centers .
To investigate CHD4's function in DNA damage response:
Baseline characterization:
DNA damage induction:
Time-course experiments:
Examine CHD4 recruitment to damage sites at multiple timepoints (5min, 30min, 1h, 6h, 24h)
Co-stain with γH2AX to mark DNA damage sites
Document CHD4 redistribution using the monoclonal antibody in immunofluorescence
Functional assessment:
Mechanism exploration:
Include appropriate controls and statistical analysis for each experimental approach.
For optimal CHD4 ChIP experiments:
Crosslinking optimization:
Test multiple formaldehyde concentrations (1-2%) and times (5-15 minutes)
For CHD4's interaction with nucleosomal DNA, dual crosslinking with ethylene glycol bis(succinimidyl succinate) (EGS) followed by formaldehyde can improve efficiency
Sonication parameters:
Optimize sonication to achieve chromatin fragments of 200-500bp
Verify fragment size by agarose gel electrophoresis before proceeding
Antibody selection and validation:
CHD4-specific considerations:
Increase salt concentration in wash buffers to reduce background
Add detergents (0.1% SDS, 1% Triton X-100) to improve specificity
For studying CHD4 at active chromatin sites, consider using sequential ChIP with histone modification antibodies
Data analysis:
Design primers for both positive control regions (known CHD4 binding sites) and negative control regions
Normalize to input and IgG controls
For genome-wide analysis, consider ChIP-seq to identify global binding patterns
Remember that CHD4's association with chromatin can be dynamic and context-dependent, requiring careful experimental timing and cellular conditions.
To investigate CHD4-NuRD complex interactions:
Co-immunoprecipitation (Co-IP) strategy:
Buffer optimization:
Use gentle lysis conditions to preserve protein-protein interactions (e.g., 150mM NaCl, 0.5% NP-40)
Test multiple detergent concentrations to maintain complex integrity
Include protease and phosphatase inhibitors to prevent degradation
Size exclusion chromatography:
Fractionate nuclear extracts to isolate intact NuRD complex
Analyze fractions by Western blot using the CHD4 monoclonal antibody
Compare fractionation patterns under different cellular conditions
Proximity ligation assay (PLA):
Utilize CHD4 monoclonal antibody in combination with antibodies against other NuRD components
This allows visualization of protein interactions in situ with subcellular resolution
Quantify interaction signals under different experimental conditions
CRISPR-based methods:
This multi-faceted approach provides comprehensive insights into how CHD4 functions within the NuRD complex in different cellular contexts.
To evaluate CHD4 as a cancer prognostic marker:
This comprehensive approach provides robust assessment of CHD4's prognostic significance across cancer types.
To study CHD4's involvement in cancer radioresistance:
Expression correlation:
Functional modulation:
Radiation response assessment:
Mechanistic investigations:
Analyze CHD4 recruitment to DNA damage sites using the monoclonal antibody in immunofluorescence
Assess impact on PARP-dependent pathways, as CHD4 is recruited to PARP-dependent sites of DNA damage
Investigate potential synergistic effects with PARP inhibitors
Examine cell cycle regulation, particularly the G1/S transition where CHD4 controls p53 deacetylation
In vivo validation:
Develop xenograft models with CHD4 knockdown versus control cells
Apply fractionated radiotherapy and measure tumor growth delay
Analyze tumor samples by IHC-P using the CHD4 monoclonal antibody
This systematic approach has revealed that CHD4 knockdown significantly increases radiosensitivity, particularly at clinically relevant dose fractions of 2 and 4 Gy .
When facing contradictory CHD4 expression data across cancer subtypes:
Technical validation:
Verify antibody specificity through positive and negative controls
Compare results from multiple antibody clones targeting different CHD4 epitopes
Cross-validate with orthogonal methods (Western blot, qRT-PCR, proteomics)
Contextual analysis:
Subcellular localization assessment:
Isoform-specific analysis:
Determine if different cancer types express alternative CHD4 isoforms
Design experiments to distinguish between isoforms (isoform-specific antibodies, RT-PCR)
Consider post-translational modifications that might affect antibody recognition
Functional context:
Statistical rigor:
Ensure appropriate statistical power for subgroup analyses
Apply multiple comparison corrections when analyzing across cancer types
Consider meta-analysis approaches when comparing independent datasets
This comprehensive approach helps resolve apparent contradictions and provides deeper insights into the context-dependent roles of CHD4 in different cancer settings.
To validate CHD4 monoclonal antibody specificity:
Genetic validation:
Expression system verification:
Express tagged CHD4 (His, FLAG, GFP) in a model system
Perform parallel detection with both the CHD4 monoclonal antibody and an antibody against the tag
Confirm signal co-localization or co-detection
Application-specific controls:
Cross-reactivity assessment:
Orthogonal method comparison:
Compare results with alternative antibody clones targeting different epitopes
Correlate protein detection with mRNA expression data
Verify findings with mass spectrometry when possible
This rigorous validation approach ensures reliable and reproducible results across experimental contexts.
Common technical challenges with CHD4 antibodies and their solutions:
High molecular weight protein detection problems:
Nuclear protein extraction difficulties:
Issue: Incomplete extraction of chromatin-bound CHD4
Solution: Use specialized nuclear extraction buffers with high salt (300-400mM NaCl), include nuclease treatment, and extend extraction time
Weak signal in fixed tissues:
Background in immunofluorescence:
Issue: Non-specific binding in IF applications
Solution: Increase blocking time/concentration, use alternative blocking agents (BSA, serum, commercial blockers), optimize antibody dilution, and include additional wash steps
Inconsistent ChIP results:
Issue: Variable chromatin immunoprecipitation efficiency
Solution: Optimize crosslinking conditions, ensure consistent sonication, pre-clear lysates, and use protein A/G beads pre-blocked with BSA
Batch-to-batch variability:
Issue: Different antibody lots showing variable performance
Solution: Test each new lot against a reference sample, maintain consistent positive controls, and consider purchasing larger amounts of a single lot for long-term studies
Species cross-reactivity limitations:
Issue: Antibody not working in claimed species
Solution: Verify sequence homology in the epitope region, test multiple antibody concentrations, and consider species-specific antibodies for critical experiments
These troubleshooting approaches address the most common technical challenges encountered when working with CHD4 monoclonal antibodies.
Essential controls for studying CHD4 recruitment to DNA damage sites:
Experimental controls:
Untreated control: Baseline CHD4 distribution before damage induction
Time course controls: Multiple timepoints to capture dynamic recruitment (5min, 30min, 1h, 4h, 24h)
Dose response: Multiple radiation doses (2Gy, 4Gy, 8Gy) to assess dose-dependent effects
Positive damage marker: Co-staining with γH2AX to confirm damage site formation
Recovery control: Allow repair time to demonstrate CHD4 dissociation from resolved damage sites
Technical controls:
Antibody specificity control: CHD4 knockdown cells to verify signal specificity
Secondary antibody control: Samples with secondary antibody only to assess background
Isotype control: Non-specific IgG of the same isotype to evaluate non-specific binding
Blocking peptide control: Pre-incubation with immunizing peptide to confirm epitope specificity
Biological mechanism controls:
PARP inhibition: Apply PARP inhibitors to block CHD4 recruitment to damage sites, as CHD4 binds to poly(ADP-ribosyl)ated proteins
ATM/ATR inhibition: Use kinase inhibitors to assess dependency on these damage signaling pathways
ZMYND8 depletion: Knockdown ZMYND8 to block CHD4 localization to acetylated damaged chromatin
Cell cycle synchronization: Compare recruitment in different cell cycle phases
Validation approaches:
Orthogonal damage induction: Compare IR with chemical damage agents (e.g., etoposide, bleomycin)
Live-cell imaging: Use fluorescently tagged CHD4 to monitor real-time recruitment kinetics
Biochemical fractionation: Isolate chromatin fractions before and after damage to quantify CHD4 recruitment
Functional consequence controls:
This comprehensive control framework ensures robust and interpretable data on CHD4's role in the DNA damage response and radioresistance.
To investigate CHD4-p53 regulatory interactions:
Baseline characterization:
Quantify CHD4 and p53 expression levels across the cell cycle using synchronized cells
Perform co-immunoprecipitation with CHD4 monoclonal antibody to assess physical interaction with p53
Map interaction domains through truncation mutants
Cell cycle analysis:
p53 acetylation status:
Assess p53 acetylation levels in CHD4-depleted versus control cells
Use acetylation-specific antibodies (K382, K120) for Western blot analysis
Compare acetylation dynamics after DNA damage with and without CHD4
Chromatin regulation:
Perform sequential ChIP (CHD4 followed by p53 or vice versa) at p53 target gene promoters
Analyze histone modifications (H3K27ac, H3K4me3, H3K9me3) at these loci
Map NuRD complex recruitment to p53-regulated genes
Rescue experiments:
Complement CHD4 knockdown with wild-type or mutant CHD4 constructs
Test HDAC inhibitors to determine if the effects are dependent on deacetylase activity
Perform simultaneous p53 knockdown to assess dependency of phenotypes on p53
Stress response:
This experimental framework will elucidate how CHD4 controls cell cycle progression through p53 deacetylation and identify potential intervention points for cancer therapy.
Advanced techniques for studying CHD4 chromatin dynamics in live cells:
Fluorescent protein fusion strategies:
Generate endogenous CHD4-GFP/RFP knock-in using CRISPR/Cas9
Create stable cell lines with doxycycline-inducible fluorescent CHD4
Design constructs with minimal linkers to prevent interference with function
Validate fusion protein functionality through rescue experiments
Live-cell imaging approaches:
Implement high-speed confocal microscopy for rapid dynamics
Use spinning disk confocal for reduced phototoxicity in long-term imaging
Apply lattice light-sheet microscopy for high spatiotemporal resolution
Employ TIRF microscopy to analyze near-membrane CHD4 populations
Advanced dynamic analysis techniques:
Fluorescence Recovery After Photobleaching (FRAP) to measure CHD4 residence time on chromatin
Fluorescence Loss In Photobleaching (FLIP) to assess exchange rates between nuclear compartments
Single-particle tracking to monitor individual CHD4 molecules
Pair correlation analysis to detect transient interactions
Multi-color imaging strategies:
Co-express fluorescently tagged histones to visualize chromatin
Include DNA damage markers (53BP1, γH2AX) to study recruitment to damage sites
Label NuRD complex partners to analyze co-recruitment dynamics
Use optogenetic tools to induce local DNA damage and monitor real-time recruitment
Quantitative analysis methods:
Implement automated tracking algorithms for CHD4 movement
Apply mathematical modeling to extract kinetic parameters
Develop computational approaches to correlate CHD4 dynamics with chromatin states
Use machine learning for pattern recognition in dynamic behaviors
Complementary biochemical validation:
This multi-faceted approach provides unprecedented insights into the spatiotemporal dynamics of CHD4 chromatin interactions in living cells.
Utilizing CHD4 monoclonal antibodies to evaluate CHD4 as a cancer therapeutic target:
Target validation approaches:
Profile CHD4 expression across cancer types and normal tissues using tissue microarrays
Correlate expression with clinical outcomes to identify cancer types most likely to benefit from CHD4 targeting
Perform synthetic lethality screens to identify genetic contexts where CHD4 inhibition is most effective
Functional assessment:
Use the antibody to monitor CHD4 levels after genetic or pharmacological inhibition
Create inducible CHD4 knockdown systems and measure effects on cancer cell viability and proliferation
Assess changes in DNA damage repair capacity and radiosensitivity
Evaluate combination effects with standard therapies (radiation, chemotherapy)
Mechanism exploration:
Perform ChIP-seq before and after CHD4 depletion to identify critical target genes
Use RNA-seq to determine transcriptional consequences of CHD4 inhibition
Analyze changes in chromatin accessibility (ATAC-seq) and histone modifications
Study effects on cancer-relevant pathways (DNA repair, cell cycle, apoptosis)
Drug development applications:
Develop cell-based screening assays using the antibody to monitor CHD4 inhibition
Implement high-content imaging to assess CHD4 displacement from chromatin
Create proximity-based assays (BRET, FRET) with labeled antibodies for drug screening
Use the antibody for target engagement studies of CHD4 inhibitors
Therapeutic window assessment:
Compare CHD4 dependency between cancer and normal cells
Identify cancer-specific vulnerabilities created by CHD4 inhibition
Study synthetic lethal interactions with common cancer mutations
Translational research:
Develop companion diagnostic approaches using the CHD4 antibody
Identify biomarkers that predict response to CHD4 targeting
Create patient-derived xenograft models to test CHD4 inhibition strategies
Evidence suggests CHD4 inhibition could be particularly effective in radioresistant HPV-negative head and neck squamous cell carcinoma, where high CHD4 expression correlates with poor survival .
Critical factors for selecting CHD4 monoclonal antibody clones:
Epitope location and characteristics:
Identify which protein domain the antibody recognizes (chromodomain, helicase domain, C-terminal region)
Determine if the epitope is accessible in your application (some epitopes may be masked in native conformations)
Consider whether post-translational modifications might affect epitope recognition
Application-specific validation:
Clone characteristics:
Evaluate isotype (IgG1, IgG2a, etc.) which may affect secondary antibody selection
Consider host species (mouse, rabbit) in relation to your experimental system
Assess purification method (protein A/G, affinity) which affects specificity and background
Species cross-reactivity needs:
Format requirements:
Performance metrics:
Compare sensitivity (detection limit) between different clones
Assess specificity (background, cross-reactivity) from validation data
Review literature citations using specific clones for your application
Special considerations:
Creating a decision matrix with these criteria helps systematically select the optimal CHD4 antibody clone for specific research needs.
Protocol adaptations for differentiating chromatin-bound and soluble CHD4:
Biochemical fractionation approach:
Soluble CHD4 extraction:
Lyse cells in low-salt buffer (10mM HEPES pH 7.9, 10mM KCl, 1.5mM MgCl₂, 0.34M sucrose, 10% glycerol, 1mM DTT, protease inhibitors)
Add 0.1% Triton X-100 and incubate on ice for 5 minutes
Centrifuge at 1,300 × g for 4 minutes at 4°C
Collect supernatant as cytoplasmic/nucleoplasmic fraction
Perform immunoprecipitation with CHD4 monoclonal antibody
Chromatin-bound CHD4 extraction:
Resuspend nuclear pellet in no-salt buffer (3mM EDTA, 0.2mM EGTA, 1mM DTT, protease inhibitors)
Incubate on ice for 30 minutes with gentle mixing
Centrifuge at 1,700 × g for 4 minutes at 4°C
Discard supernatant (nucleoplasmic proteins)
Extract chromatin-bound proteins with high-salt buffer (50mM Tris-HCl pH 7.4, 300-400mM NaCl, 1% NP-40, 0.5% sodium deoxycholate, protease inhibitors)
Sonicate briefly to release chromatin-bound proteins
Clarify by centrifugation and perform immunoprecipitation
Sequential immunoprecipitation strategy:
Extract soluble proteins first with mild detergent
Perform first IP with CHD4 antibody
Subsequently extract chromatin-bound proteins from the residual pellet
Conduct second IP with the same antibody
Compare protein partners between fractions by mass spectrometry
In situ chromatin binding analysis:
Perform detergent extraction of unfixed cells on coverslips to remove soluble proteins
Fix remaining chromatin-bound proteins
Immunostain with CHD4 monoclonal antibody
Compare with standard fixation protocol to assess proportion of chromatin-bound CHD4
Quantitative assessment:
Use Western blotting to quantify distribution between fractions
Include controls for fraction purity (GAPDH for cytoplasm, histone H3 for chromatin)
Analyze CHD4 modifications specific to each pool
Adaptation for specific conditions:
This fractionation approach provides crucial insights into the dynamic regulation of CHD4 between soluble and chromatin-bound states under different cellular conditions.
Specialized approaches for studying CHD4 in stem cells and primary neurons:
Stem cell-specific considerations:
Technical adaptations:
Optimize fixation to maintain stemness markers (shorter fixation times)
Use gentle permeabilization to preserve nuclear architecture
Implement low-detergent extraction for chromatin studies
Adjust antibody concentrations for potentially different expression levels
Experimental design:
Correlate CHD4 expression/localization with pluripotency markers (Oct4, Nanog, Sox2)
Track CHD4 dynamics during differentiation using time-course analysis
Perform CHD4 ChIP-seq to identify stem cell-specific binding sites
Create stem cell-specific CHD4 knockdown/knockout using inducible systems
Functional assessments:
Evaluate impact of CHD4 modulation on self-renewal versus differentiation
Analyze changes in lineage-specific gene expression after CHD4 depletion
Compare CHD4-NuRD complex composition between stem cells and differentiated cells
Primary neuron adaptations:
Technical modifications:
Use neuronal-optimized fixation protocols (4% PFA, 10-15 minutes)
Implement antigen retrieval methods specifically validated for neural tissues
Extend antibody incubation times (overnight at 4°C) for better penetration
Use detergent-free mounting media to preserve neuronal morphology
Neuronal-specific approaches:
Perform subcellular localization studies (soma vs. dendrites vs. axons)
Correlate CHD4 expression with neuronal maturation markers
Investigate CHD4 in different neuronal subtypes (glutamatergic, GABAergic)
Examine CHD4 response to neuronal activity or depolarization
Functional investigations:
Common specialized techniques:
Low-input ChIP protocols for limited cell numbers
Single-cell approaches to address heterogeneity
Ex vivo slice cultures for maintaining tissue architecture
Live imaging in organoid systems
These tailored methodological approaches account for the unique characteristics of stem cells and neurons while investigating CHD4's specialized functions in these cell types.
Single-cell approaches for investigating CHD4 in heterogeneous cancers:
Single-cell protein analysis techniques:
Implement mass cytometry (CyTOF) with CHD4 monoclonal antibodies to quantify expression in thousands of individual cells
Develop multiplex immunofluorescence panels including CHD4 and cancer subtype markers
Apply imaging mass cytometry for spatial context of CHD4 expression in tumor sections
Utilize microfluidic antibody capture for single-cell Western blotting
Integrated multi-omics approaches:
Combine single-cell CHD4 protein detection with scRNA-seq using CITE-seq
Correlate CHD4 protein levels with chromatin accessibility using scATAC-seq
Implement scCUT&Tag to map CHD4 binding sites in individual cells
Relate CHD4 levels to DNA damage repair capacity at single-cell resolution
Tumor heterogeneity characterization:
Identify distinct CHD4-high and CHD4-low subpopulations within tumors
Correlate CHD4 expression with cancer stem cell markers
Map CHD4 expression across tumor spatial gradients (center vs. periphery)
Track how CHD4-expressing subclones evolve during treatment
Functional readouts at single-cell level:
Clinical applications:
Technical considerations:
Optimize antibody concentration and signal amplification for low abundance detection
Implement machine learning algorithms for unbiased cell clustering
Develop computational approaches to integrate CHD4 data across multiple platforms
Address fixation and permeabilization challenges for nuclear protein detection
This single-cell perspective would transform our understanding of CHD4's role in tumor heterogeneity and therapy resistance mechanisms.
Advanced approaches for studying CHD4 post-translational modifications:
Identification of modification sites:
Perform mass spectrometry analysis of immunoprecipitated CHD4 to map all modifications
Create modification-specific antibodies for key sites (phosphorylation, acetylation, etc.)
Use proximity labeling (BioID, APEX) coupled with CHD4 to identify modification enzymes
Apply crosslinking mass spectrometry to map interactions between modified domains
Functional significance assessment:
Generate site-specific mutants (phospho-mimetic, phospho-dead) through CRISPR knock-in
Create inducible expression systems for modified versus unmodified CHD4
Perform domain-swap experiments to isolate modification-dependent regions
Use temporal control of modifications through optogenetic or chemical-genetic approaches
Regulatory mechanism exploration:
Chromatin association analysis:
Perform ChIP-seq with modification-specific antibodies
Compare genomic localization of differently modified CHD4 populations
Analyze how modifications affect CHD4 residence time on chromatin using FRAP
Study impact on nucleosome remodeling activity with in vitro assays
Complex assembly regulation:
Therapeutic targeting strategies:
Screen for compounds that modulate specific CHD4 modifications
Test combination approaches targeting both CHD4 and its modifying enzymes
Explore synthetic lethality between CHD4 modifications and cancer mutations
Develop modification-specific inhibitors to disrupt key CHD4 functions
This comprehensive approach would provide unprecedented insights into how post-translational modifications regulate CHD4's diverse cellular functions, potentially revealing new therapeutic opportunities.
Emerging technologies for therapeutic targeting of CHD4 in radioresistant cancers:
Advanced targeting modalities:
Develop proteolysis-targeting chimeras (PROTACs) to induce CHD4 degradation
Create allosteric inhibitors targeting CHD4 ATPase activity
Design conformation-specific antibody-based therapeutics
Implement RNA-based approaches (ASOs, siRNA) with cancer-specific delivery
Structure-guided drug development:
Utilize cryo-EM structures of CHD4-nucleosome complexes for rational drug design
Apply fragment-based screening to identify binding pockets
Develop ATP-competitive inhibitors for the helicase domain
Create domain-specific inhibitors targeting chromodomain-histone interactions
Functional screening platforms:
Implement CRISPR screens to identify synthetic lethal interactions with CHD4 inhibition
Use high-content imaging to assess effects on DNA damage response
Develop patient-derived organoids to test CHD4-targeting strategies
Create reporter systems to monitor CHD4-dependent transcriptional repression
Precision medicine approaches:
Combination therapy strategies:
Delivery innovations: