ADNP antibodies are immunoreagents designed to detect ADNP, a 1,102-amino-acid protein (~124 kDa theoretical molecular weight) with roles in neuroprotection, chromatin regulation, and microtubule dynamics . These antibodies enable researchers to study ADNP's expression, localization, and interactions in cellular and tissue contexts.
Recent studies have standardized protocols for evaluating ADNP antibody performance using knockout (KO) cell lines and isogenic controls . Key findings include:
Discrepancies in observed molecular weight (e.g., 150 kDa vs. 124 kDa) are attributed to post-translational modifications or SDS-PAGE anomalies .
N-terminal antibodies (e.g., Aviva OASG00229) show stronger specificity in blocking peptide assays .
ADNP binds 10,642 genomic sites in embryonic stem cells (ESCs), primarily at intergenic regions and promoters. Targets include genes regulating metabolism and cell signaling (e.g., GTPase binding) .
ADNP interacts with microtubule-associated proteins (e.g., EB1/EB3) to stabilize neuronal structures .
The NAP motif (aa 354–361) is essential for neuroprotection, with peptide analogs showing therapeutic potential in autism and Alzheimer’s models .
Helsmoortel-Van der Aa syndrome: Mutant ADNP proteins show reduced stability, detectable via C-terminal antibodies .
Peripheral ADNP levels serve as biomarkers for Alzheimer’s and schizophrenia .
Antibody Selection: Prioritize antibodies validated in KO models (e.g., AF6737, 17987-1-AP) .
Protocols: Use standardized workflows from Protocol Exchange for reproducibility .
Controls: Include KO lysates and blocking peptides to confirm specificity .
A robust validation approach for ADNP antibodies requires multiple complementary techniques. Blocking peptide competition assays are essential for distinguishing between specific and non-specific signals. In this method, the antibody is pre-incubated with its corresponding immunizing peptide before application to samples, resulting in the disappearance of specific bands if the antibody is truly detecting ADNP. This technique has successfully validated N-terminal ADNP antibodies, resulting in unique band signals around 150 kDa, above the theoretical molecular weight of 124 kDa .
Additionally, comparison between parental and CRISPR/Cas9-generated homozygous ADNP knockout cell lines provides definitive validation. The complete disappearance of the 150 kDa band in knockout samples confirms antibody specificity for intact ADNP . For researchers without access to knockout lines, siRNA knockdown can be employed as an alternative approach, though the partial reduction in signal provides less definitive validation than complete knockout models.
Selecting the appropriate ADNP antibody depends on your experimental application and the specific domain of interest. For Western blot applications detecting full-length ADNP, antibodies targeting either the N-terminal or C-terminal domains have successfully identified ADNP at approximately 150 kDa . The Aviva Systems N-terminal antibody has been extensively validated using blocking peptide competition assays and shows reliable detection across various sample types .
For immunoprecipitation studies, N-terminal immunoprecipitation-competent antibodies have demonstrated effectiveness in enriching both wild-type and truncated mutant ADNP in overexpression systems . When studying ADNP mutations that affect the C-terminus, N-terminal antibodies are preferred to ensure detection of truncated proteins. For immunofluorescence applications, standardized validation protocols comparing readouts between knockout cell lines and isogenic parental controls have identified high-performing antibodies .
Several commercial antibodies from manufacturers including Aviva Systems, R&D Systems, and Abcam have been systematically characterized across multiple applications, providing researchers with reliable options based on their specific experimental needs .
The inconsistent molecular weight detection of ADNP (ranging from 60 to 175 kDa) across independent studies stems from multiple factors. The theoretical molecular weight of human ADNP is 124 kDa, but most validated antibodies detect the protein at approximately 150 kDa . This discrepancy likely results from:
Post-translational modifications affecting protein mobility in SDS-PAGE
Use of different gel systems and running conditions across laboratories
Variation in antibody specificity and epitope recognition
Sample preparation methods affecting protein conformation and stability
Cross-reactivity with homologous proteins or splice variants
Researchers should be aware that recombinant tagged versions of ADNP will show correspondingly higher molecular weights. For example, GFP-tagged ADNP appears at approximately 175 kDa (adding the 25 kDa GFP tag to the 150 kDa observed for native ADNP) . Consistent observation of ADNP at 150 kDa across multiple validated antibodies targeting different epitopes suggests this represents the true migration pattern of the full-length protein.
Distinguishing non-specific bands from genuine ADNP signals requires a multi-faceted approach:
Blocking peptide competition assays: Pre-incubation of the antibody with its immunizing peptide results in disappearance of specific ADNP bands while non-specific signals remain. This approach has been successfully used with N-terminal ADNP antibodies (Aviva Systems) to identify the specific 150 kDa ADNP band among multiple non-specific signals .
Use of knockout controls: Homozygous CRISPR/Cas9-generated ADNP knockout cell lines provide the gold standard control. Only signals that disappear in knockout samples represent genuine ADNP detection . When knockout controls are unavailable, siRNA knockdown samples can provide partial validation.
Multiple antibody approach: Using antibodies targeting different epitopes (N-terminal and C-terminal) can confirm specificity when the same band is detected by both antibodies. Detection with different C-terminal antibodies has confirmed ADNP signals at an observed molecular weight of 150 kDa .
Overexpression controls: Including samples overexpressing tagged ADNP can help identify the appropriate molecular weight region, accounting for the added tag size.
Researchers should be aware that many commercially available ADNP antibodies produce non-specific bands without unique ADNP signals, necessitating thorough validation before experimental use .
Detecting native ADNP protein requires carefully optimized experimental conditions:
The variability in ADNP detection across studies underscores the importance of these optimized conditions for reliable results.
Mutant ADNP proteins are particularly challenging to detect in patient-derived samples due to several factors:
Protein instability: Truncated mutant ADNP proteins may be unstable in native contexts, undergoing rapid degradation. Western blotting of patient-derived hiPSCs, immortalized lymphoblastoid cell lines, and post-mortem patient brain material has failed to detect native mutant ADNP protein despite confirmed mRNA expression .
Epitope loss: Most HVDAS-causing mutations occur in the last exon of ADNP, resulting in truncated proteins lacking C-terminal epitopes. This necessitates the use of N-terminal antibodies for detection attempts.
Low abundance: Mutant proteins may be expressed at levels below the detection threshold of standard Western blot techniques.
Antibody limitations: Current antibodies may have insufficient sensitivity to detect low-abundance mutant proteins in complex patient samples.
Interestingly, epitope-tagged mutant ADNP constructs can be detected when overexpressed in experimental systems, suggesting that addition of an epitope tag (such as GFPSpark or Flag-tag) may stabilize the mutant protein . Alternative approaches such as enrichment through immunoprecipitation with N-terminal antibodies have been attempted but failed to detect native mutant protein in patient-derived samples despite successfully enriching overexpressed truncated mutants .
For successful immunofluorescence studies using ADNP antibodies, researchers should follow these methodological guidelines:
Fixation: 4% paraformaldehyde for 15-20 minutes at room temperature preserves ADNP epitopes while maintaining cellular architecture. Alternative fixatives such as methanol may be tested for specific antibodies if paraformaldehyde yields suboptimal results.
Permeabilization: 0.1% Triton X-100 for 10 minutes allows antibody access to the predominantly nuclear ADNP. For delicate samples, 0.05% saponin may provide gentler permeabilization.
Blocking: 5% normal serum from the species in which the secondary antibody was raised (typically goat or donkey) with 1% BSA in PBS for 1 hour at room temperature reduces non-specific binding.
Antibody selection: Several commercial antibodies have been characterized specifically for immunofluorescence applications using standardized validation protocols comparing readouts between knockout cell lines and isogenic parental controls . This systematic approach identifies antibodies that provide specific nuclear staining patterns consistent with ADNP's known subcellular localization.
Controls: Include negative controls (primary antibody omission, isotype controls, and ideally ADNP knockout cells) and positive controls (cell lines known to express high levels of ADNP).
Co-staining: ADNP co-staining with nuclear markers (DAPI) and known interaction partners (such as components of the SWI/SNF chromatin remodeling complex) can provide additional validation of specificity.
Imaging parameters: Confocal microscopy with appropriate laser settings to avoid bleed-through when performing multi-color imaging. Z-stack acquisition may be necessary to fully capture the nuclear distribution of ADNP.
For research focused on ADNP's role in chromatin remodeling, detecting its co-localization with other SWI/SNF components requires careful optimization of fixation and permeabilization protocols to preserve nuclear architecture.
Optimizing ADNP immunoprecipitation for protein interaction studies requires careful consideration of several methodological aspects:
Antibody selection: N-terminal immunoprecipitation-competent ADNP antibodies have successfully enriched both wild-type and truncated mutant ADNP in overexpression systems . The choice between N-terminal and C-terminal antibodies depends on the specific protein interactions being investigated and whether C-terminal truncating mutations are present.
Lysis conditions: For detecting chromatin-associated interactions, nuclear extraction protocols with moderate salt concentrations (150-300 mM NaCl) balance extraction efficiency with preservation of protein complexes. For cytosolic interactions, milder lysis buffers (e.g., 25 mM Tris-HCl pH 7.4, 150 mM NaCl, 1% NP-40, 1 mM EDTA with protease inhibitors) are recommended.
Cross-linking: For transient or weak interactions, in situ cross-linking with 1% formaldehyde prior to lysis can stabilize protein complexes. This approach has been valuable for confirming ADNP as a component of the SWI/SNF chromatin remodeling complex .
IP protocol optimization:
Pre-clearing lysates with protein A/G beads reduces non-specific binding
Overnight antibody incubation at 4°C maximizes antigen capture
Including 5% glycerol and 0.1% NP-40 in wash buffers can preserve complex integrity
Graduated salt washes (150 mM to 300 mM NaCl) can help distinguish between stable and transient interactors
Detection methods: Mass spectrometry following immunoprecipitation provides unbiased identification of interaction partners, as demonstrated in the discovery of ADNP as a component of the SWI/SNF chromatin remodeling complex . Western blotting with antibodies against suspected interaction partners (such as SIRT1 or EB1/EB3 microtubule-end binding proteins) can confirm specific interactions .
For capturing interactions between ADNP and microtubule-associated proteins, protocols should include stabilization of microtubules with taxol prior to gentle lysis to preserve these cytoskeletal interactions.
When studying ADNP protein in disease models, particularly in Helsmoortel-Van der Aa syndrome (HVDAS) research, the following controls are essential:
Genetic confirmation: Sequencing verification of ADNP mutations in patient-derived samples or engineered cell lines is critical, as different mutations may affect protein stability and function differently.
mRNA expression controls: RT-PCR or RNA-seq verification of mutant ADNP mRNA expression is necessary, especially given that most HVDAS-causing mutations escape nonsense-mediated decay .
Antibody validation controls:
Blocking peptide competition assays to confirm antibody specificity
Isogenic wild-type controls alongside mutant samples
Multiple antibodies targeting different epitopes to confirm detection
Positive controls using tagged ADNP constructs with the same mutation
Cell type considerations: ADNP expression and the effects of mutations may vary across cell types. Comparing multiple relevant cell types (e.g., neural progenitors, mature neurons, and non-neural cells) provides a more comprehensive understanding of mutation effects.
Developmental timing: For developmental disorders like HVDAS, examining ADNP expression across different developmental stages is crucial, as age-dependent changes in ADNP expression have been observed in mouse models .
Functional readouts: Beyond protein detection, functional assays measuring chromatin remodeling activity, transcriptional regulation, or microtubule stability provide important insights into the consequences of ADNP mutations.
The challenges in detecting native mutant ADNP protein in patient samples despite confirmed mRNA expression suggest potential post-transcriptional regulation mechanisms that should be investigated. Alternative approaches may be necessary, such as using epitope-tagged constructs that can stabilize mutant proteins .
Post-translational modifications (PTMs) can significantly impact ADNP antibody recognition, presenting both challenges and opportunities for researchers:
Impact on apparent molecular weight: The discrepancy between ADNP's theoretical molecular weight (124 kDa) and its observed migration pattern (approximately 150 kDa) likely reflects extensive post-translational modifications . These modifications alter protein migration in SDS-PAGE without necessarily affecting antibody binding.
Epitope masking: Some PTMs may directly mask antibody epitopes, reducing or preventing antibody binding. Phosphorylation, particularly near antibody recognition sites, can alter local protein conformation and interfere with antibody access.
Methodological considerations:
Phosphatase treatment of lysates prior to Western blotting can determine if phosphorylation affects antibody recognition
Sample preparation with deglycosylating enzymes may reveal if glycosylation contributes to the higher-than-expected molecular weight
Including deacetylase inhibitors in lysis buffers may preserve acetylation states relevant to antibody binding
PTM-specific antibodies: For advanced research on ADNP regulation, development of PTM-specific antibodies (e.g., phospho-ADNP, acetylated-ADNP) could provide valuable insights into activity-dependent regulation of this neuroprotective protein.
Functional significance: ADNP interacts with SIRT1, a NAD⁺-dependent deacetylase enzyme, suggesting acetylation may regulate ADNP function . Similarly, interactions with microtubule-end binding proteins (EB1/EB3) may be regulated by phosphorylation events.
Researchers studying ADNP in different physiological contexts should consider how condition-specific PTMs might affect antibody detection, potentially necessitating different antibodies or sample preparation methods for optimal detection across experimental conditions.
Studying truncated ADNP mutants in Helsmoortel-Van der Aa syndrome requires specialized methodological approaches to overcome detection challenges:
Epitope tagging strategies: N-terminal tagging of ADNP with GFPSpark or Flag-tag has successfully enabled detection of mutant forms after introduction of patient mutations in expression systems . These tags appear to stabilize the otherwise unstable truncated proteins.
Site-directed mutagenesis: Introduction of patient-specific mutations into wild-type ADNP expression vectors via site-directed mutagenesis allows controlled studies of each mutation's effects on protein stability and function.
Inducible expression systems: Doxycycline-inducible expression systems can control expression levels of mutant proteins, potentially preventing toxicity or compensatory mechanisms that might obscure phenotypes.
Patient-derived models:
hiPSCs from patients provide an endogenous context for studying mutations
Immortalized lymphoblastoid cell lines offer accessible patient-derived material
Post-mortem brain tissue, while limited in availability, provides insights into in vivo effects
CRISPR/Cas9 knock-in models: Generation of isogenic cell lines differing only in the ADNP mutation provides the most controlled experimental system for studying mutation-specific effects.
RNA analysis: Given the challenges in protein detection, comprehensive RNA analysis (including RNA-seq and targeted RT-PCR) can provide insights into expression levels and potential alternative splicing induced by mutations.
Alternative detection methods: Proximity ligation assays may detect mutant proteins at levels below Western blot detection thresholds, particularly when examining interactions with known binding partners.
Despite these approaches, researchers should note that Western blotting of patient-derived samples has consistently failed to detect native mutant ADNP protein, even with N-terminal antibodies that should recognize truncated forms . This suggests fundamental instability of mutant proteins in their native context and raises important questions about disease mechanisms.
To resolve contradictory findings regarding ADNP molecular weight across different studies (ranging from 60 to 175 kDa), the following experimental design approach is recommended:
Multi-antibody validation panel: Assemble a comprehensive panel of commercially available ADNP antibodies targeting different epitopes (N-terminal, central region, C-terminal) and systematically test them under identical conditions.
Standardized sample preparation:
Use multiple cell types with confirmed ADNP expression
Apply identical lysis conditions across all samples
Include both reducing and non-reducing conditions to assess potential disulfide bonding effects
Process samples immediately to minimize degradation
Cross-laboratory standardization:
Run identical samples on different gel systems (gradient gels vs. fixed percentage)
Use multiple molecular weight markers from different manufacturers
Standardize transfer conditions and blocking reagents
Definitive controls:
Include CRISPR/Cas9-generated ADNP knockout cells alongside parental controls
Run recombinant ADNP protein (both tagged and untagged versions)
Include samples with ADNP siRNA knockdown
Test blocking peptide competition for each antibody
Advanced analytical techniques:
Mass spectrometry analysis of immunoprecipitated ADNP to confirm molecular weight
2D gel electrophoresis to separate potential ADNP isoforms or modified forms
Native gel electrophoresis to detect possible complexes involving ADNP
Protocol documentation:
Create a comprehensive database of experimental conditions used across studies
Develop a standardized reporting format for ADNP detection methods
This systematic approach would enable identification of factors contributing to variability while establishing consensus on the true molecular weight of ADNP. Current evidence from validated antibodies consistently identifies ADNP at approximately 150 kDa, despite its theoretical molecular weight of 124 kDa , suggesting this represents the actual migration pattern of the full-length protein.
Current challenges in detecting native mutant ADNP proteins might be addressed through several innovative approaches:
Single-molecule imaging techniques: Methods such as stochastic optical reconstruction microscopy (STORM) or photoactivated localization microscopy (PALM) using fluorescently tagged antibodies may detect low-abundance mutant proteins below the threshold of traditional Western blotting.
Proximity-based detection methods: Techniques like proximity ligation assay (PLA) or split-GFP complementation can amplify detection signals when ADNP interacts with known binding partners, potentially revealing mutant proteins at levels too low for direct detection.
Mass spectrometry innovations:
Targeted multiple reaction monitoring (MRM) mass spectrometry can detect specific peptides from ADNP with extremely high sensitivity
Parallel reaction monitoring (PRM) could identify mutant-specific peptide fragments
SWATH-MS (Sequential Window Acquisition of all Theoretical Mass Spectra) might detect mutant peptides in complex samples without prior enrichment
Nanobody development: Engineering smaller antibody fragments (nanobodies) with enhanced epitope access might improve detection of unstable or partially degraded mutant proteins.
RNA-protein tethering strategies: MS2 or λN peptide tagging of ADNP mRNA combined with fluorescent protein tagging could enable visualization of newly synthesized mutant proteins before degradation.
Degradation pathway inhibitors: Systematic testing of proteasome, autophagy, and other degradation pathway inhibitors during detection attempts might temporarily stabilize mutant proteins sufficiently for detection.
In situ protein analysis: Emerging spatial transcriptomics and proteomics methods might detect mutant proteins within their cellular context without disrupting potentially stabilizing interactions.
These approaches could provide crucial insights into whether mutant ADNP proteins are transiently expressed and rapidly degraded or whether they form stable complexes with other proteins that have eluded detection by current methods.
Developing more specific and sensitive antibodies for ADNP detection requires strategic approaches to antibody engineering and validation:
Strategic epitope selection:
Target highly unique sequences within ADNP with minimal homology to other proteins
Select epitopes in structured regions that maintain conformation after denaturation
Develop antibodies recognizing junction sites between domains for enhanced specificity
Generate epitope-specific antibodies recognizing mutant-specific neoepitopes created by frameshifts
Advanced immunization strategies:
Use multiple peptide immunogens from different ADNP regions
Employ DNA immunization with full-length ADNP to generate diverse antibody responses
Implement negative selection strategies to remove cross-reactive antibody-producing B cells
Antibody engineering techniques:
Affinity maturation through directed evolution or computational design
Fragment antigen-binding (Fab) or single-chain variable fragment (scFv) optimization
Development of bispecific antibodies requiring dual epitope binding for signal generation
Rigorous validation protocols:
Establish comprehensive validation pipelines using CRISPR/Cas9 knockout controls
Implement blocking peptide competition assays for all candidates
Test specificity across multiple sample types and experimental conditions
Compare detection limits against existing antibodies using dilution series
Application-specific optimization:
Develop distinct antibodies optimized for Western blot, immunoprecipitation, and immunofluorescence applications
Test fixation compatibility for immunohistochemistry applications
Assess performance in challenging sample types such as post-mortem tissue
Recent collaborative initiatives seeking to address antibody reproducibility issues have begun characterizing commercially available ADNP antibodies using standardized experimental protocols . These efforts provide valuable resources for researchers while establishing benchmarks for next-generation antibody development.
The observed instability of mutant ADNP proteins has profound implications for therapeutic development strategies in ADNP-related disorders:
Mechanism reconsideration: The failure to detect native mutant ADNP protein despite confirmed mRNA expression suggests that HVDAS may not function through a dominant-negative or gain-of-function mechanism as previously hypothesized, but rather through haploinsufficiency . This fundamentally shifts therapeutic targeting strategies from mutant protein modification to wild-type compensation.
Protein stabilization approaches:
Small molecules that bind and stabilize mutant ADNP might restore partial function
Chaperone modulators could prevent premature degradation of mutant proteins
Proteasome inhibitors might temporarily increase mutant protein levels for functional recovery assessment
Gene therapy considerations:
AAV-mediated delivery of wild-type ADNP cDNA might compensate for haploinsufficiency
CRISPR/Cas9-mediated correction of mutations in patient cells could restore normal protein expression
Antisense oligonucleotides might promote exon skipping to preserve reading frame in nonsense mutations
Peptide-based therapies: NAP (davunetide), an eight-amino-acid peptide derived from ADNP, has shown neuroprotective effects and might compensate for reduced ADNP function. Optimized delivery systems for this and similar peptides represent promising therapeutic avenues .
Biomarker development challenges: The difficulty in detecting mutant ADNP protein complicates development of protein-based biomarkers for treatment response monitoring. Alternative biomarkers reflecting downstream pathway activity may be more practical.
Regulatory pathway activation: Identifying and targeting downstream pathways affected by ADNP deficiency (such as WNT-beta-catenin signaling, which ADNP positively modulates) might circumvent the need to address the unstable mutant protein directly .
Understanding the precise fate of mutant ADNP proteins—whether they are rapidly degraded after synthesis or form transient intermediates with partial function—will be crucial for optimizing therapeutic approaches and assessing potential side effects of stabilization strategies.