ANP32D (acidic leucine-rich nuclear phosphoprotein 32 family member D) shares 89.3% amino acid sequence identity with ANP32A but exhibits divergent biological roles . While ANP32A functions as a tumor suppressor, ANP32D is tumorigenic, particularly in prostate cancer, and lacks the tumor-suppressive 25-amino-acid region present in ANP32A . Its intronless gene structure suggests it may act as a pseudogene expressed in pathological contexts .
Key Features of ANP32D:
Domains: Leucine-rich repeats (LRR) at the N-terminus; acidic C-terminal region .
Expression: Preferentially upregulated in prostate cancer and other malignancies .
ANP32D is implicated in tumorigenesis:
Prostate Cancer: Overexpressed in malignant prostate tissue compared to benign samples, correlating with oncogenic activity .
Therapeutic Target: Lacks tumor-suppressive regions found in ANP32A, making it a candidate for targeted cancer therapies .
Transcriptional Regulation: ANP32D modulates chromatin architecture through interactions with histone tails and transcription factors, akin to other ANP32 family members .
Apoptosis: Unlike ANP32A/B, which regulate caspases, ANP32D’s role in apoptosis remains less defined but may involve PP2A phosphatase inhibition .
Mechanistic Studies: Clarify ANP32D’s interactions with PP2A and caspases to elucidate its tumorigenic pathways .
Diagnostic Potential: Validate ANP32D as a biomarker for prostate cancer using IHC and serum assays .
Therapeutic Development: Explore small-molecule inhibitors targeting ANP32D’s LRR domain to disrupt oncogenic signaling .
ANP32D (acidic leucine-rich nuclear phosphoprotein 32 family member D) is a 14 kDa protein belonging to the acidic nuclear phosphoprotein 32 family. Unlike its family member PP32 (ANP32A), which functions as a tumor suppressor, ANP32D demonstrates tumorigenic properties. The critical distinction lies in the absence of a specific 25 amino acid region in ANP32D that is present in PP32 and responsible for its tumor suppression function . ANP32D is also known as PP32R2 (Phosphoprotein 32-related protein 2) or tumorigenic protein pp32r2.
While ANP32A and ANP32B are essential for influenza virus replication and interact with viral polymerase complexes, current research does not indicate ANP32D plays a similar role in viral replication . ANP32D's functional divergence from other family members makes it a distinct target for cancer research applications.
ANP32D antibodies such as the PACO47302 are validated for multiple research applications:
Application | Recommended Dilution | Notes |
---|---|---|
ELISA | 1:2000-1:10000 | For protein quantification |
Immunohistochemistry (IHC) | 1:20-1:200 | For tissue section analysis |
Immunofluorescence (IF) | 1:50-1:200 | For cellular localization studies |
Western Blot | Validated | For protein detection and quantification |
These applications enable researchers to investigate ANP32D expression patterns, subcellular localization, and protein interactions in various experimental contexts . When designing experiments, researchers should consider optimizing the antibody dilution within the recommended range based on their specific sample type and detection method.
For optimal antibody performance and longevity, ANP32D antibodies should be stored in their recommended buffer conditions. For example, the PACO47302 antibody is stored in a preservative solution containing 0.03% Proclin 300, 50% glycerol, and 0.01M PBS at pH 7.4 .
Methodological approach to antibody storage:
Store antibody aliquots at -20°C for long-term storage
Avoid repeated freeze-thaw cycles (create single-use aliquots)
For short-term use (1-2 weeks), store at 4°C
Protect from light, particularly fluorophore-conjugated variants
Centrifuge briefly before opening to collect solution at the bottom of the tube
Avoid introducing contaminants into the stock solution
When diluting the antibody, use freshly prepared buffer solutions, preferably containing a carrier protein (like BSA) to prevent nonspecific adsorption to container surfaces.
ANP32D expression has been documented in several human tissues and cancer samples. Immunohistochemical analysis with ANP32D antibodies has successfully detected the protein in:
Various cancer cell models
Unlike ANP32A and ANP32B that are widely expressed across multiple tissue types, ANP32D demonstrates a more restricted expression pattern. This makes it a potentially specific marker for certain cancer types. When investigating a new tissue or cell type, researchers should include appropriate positive and negative controls to validate ANP32D expression.
To investigate the contrasting functions of ANP32D (tumorigenic) versus ANP32A (tumor suppressor), researchers can employ a multi-faceted approach using ANP32D antibodies:
Differential expression analysis: Use ANP32D and ANP32A antibodies in matched normal/tumor tissue arrays to quantify relative expression patterns. Correlate expression with tumor grade, stage, and patient outcomes.
Co-immunoprecipitation studies: Employ ANP32D antibodies to pull down protein complexes and analyze interaction partners that differ from ANP32A complexes. This can reveal unique signaling pathways and molecular mechanisms.
Dual immunofluorescence: Perform simultaneous labeling of ANP32D and ANP32A to examine potential mutual exclusivity or co-expression patterns at the single-cell level.
Functional domain analysis: Utilize truncated ANP32D constructs with and without the 25 amino acid region absent in wild-type ANP32D (but present in ANP32A) to dissect functional differences. ANP32D antibodies can verify expression and localization of these constructs.
Phosphorylation studies: Examine post-translational modifications that may distinguish ANP32D's tumorigenic activities using phospho-specific antibodies alongside total ANP32D antibodies.
The critical methodological consideration is to include proper controls that account for potential antibody cross-reactivity between family members due to their sequence homology .
For cancer research applications, ANP32D antibodies can be employed with the following optimized protocols:
Deparaffinize sections and perform antigen retrieval (citrate buffer pH 6.0, 95°C, 20 minutes)
Block endogenous peroxidase (3% H₂O₂, 10 minutes)
Protein block (5% normal goat serum, 1 hour)
Incubate with ANP32D primary antibody (1:100 dilution, overnight at 4°C)
Apply HRP-conjugated secondary antibody (1:500, 1 hour at room temperature)
Develop with DAB and counterstain with hematoxylin
Score expression in relation to tumor/normal boundaries and correlate with pathological features
Fix cells (4% paraformaldehyde, 15 minutes)
Permeabilize (0.1% Triton X-100, 10 minutes)
Block (3% BSA, 1 hour)
Incubate with ANP32D antibody (1:100 dilution, overnight at 4°C)
Apply fluorophore-conjugated secondary antibody (1:500, Alexa Fluor 488, 1 hour)
Counterstain nuclei with DAPI
Image on confocal microscope for precise subcellular localization
For gastric cancer and HeLa cells, these protocols have been validated to produce specific ANP32D labeling . The key methodological consideration is to include matched isotype controls and antigen pre-absorption controls to confirm staining specificity.
Distinguishing between highly homologous ANP32 family members requires careful methodological approaches:
Antibody selection: Choose ANP32D antibodies raised against unique epitopes not conserved in ANP32A/B/E. Antibodies targeting the C-terminal region are optimal since this region shows greatest sequence divergence among family members.
Western blot differentiation: ANP32D has a molecular weight of approximately 14 kDa, distinguishing it from ANP32A/B (~30 kDa). Use gradient gels (4-20%) for optimal separation.
qRT-PCR validation: Design primers targeting unique regions of ANP32D mRNA to confirm antibody specificity at the transcript level. This cross-validation approach ensures the detected protein is genuinely ANP32D.
Knockout/knockdown controls: Generate CRISPR/siRNA models with selective depletion of individual ANP32 family members to confirm antibody specificity.
Mass spectrometry confirmation: For critical experiments, immunoprecipitate using ANP32D antibodies and confirm protein identity by mass spectrometry peptide analysis.
The sequence homology between ANP32 proteins necessitates rigorous validation, particularly when examining tissues that express multiple family members simultaneously .
Several methodological challenges exist when working with ANP32D antibodies:
Cross-reactivity with other ANP32 family members: Due to sequence homology, antibodies may recognize multiple family proteins. Mitigation strategy: Validate antibody specificity using recombinant proteins of all ANP32 family members in parallel Western blots.
Nonspecific binding in high-background tissues: Some tissues (e.g., liver) naturally exhibit high background. Mitigation strategy: Optimize blocking conditions using tissue-specific blockers and extend blocking time to 2 hours.
Epitope masking due to protein-protein interactions: ANP32D's interactions may obscure antibody binding sites. Mitigation strategy: Test multiple antibodies targeting different epitopes.
Fixation-dependent epitope changes: Some epitopes may be sensitive to fixation methods. Mitigation strategy: Compare different fixation protocols (paraformaldehyde, methanol, acetone) to determine optimal epitope preservation.
Low signal strength due to low expression levels: ANP32D may be expressed at low levels in some tissues. Mitigation strategy: Implement signal amplification methods like tyramide signal amplification or polymer-based detection systems.
Batch-to-batch variability in polyclonal antibodies: Polyclonal antibodies like PACO47302 may show batch variations. Mitigation strategy: Validate each new lot against a reference sample and maintain consistent lot usage throughout a study.
A systematic validation approach using multiple detection methods will ensure robust and reproducible results when working with ANP32D antibodies .
ANP32D is involved in transcriptional regulation and apoptotic pathways. Researchers can use ANP32D antibodies to investigate these functions through several methodological approaches:
Chromatin Immunoprecipitation (ChIP):
Use ANP32D antibodies to precipitate chromatin fragments
Sequence associated DNA (ChIP-seq) to identify genomic binding sites
Analyze binding motifs to determine transcriptional networks regulated by ANP32D
Compare with ANP32A/B binding patterns to identify unique targets
Proximity Ligation Assay (PLA):
Detect in situ interactions between ANP32D and transcription factors or apoptotic regulators
Quantify interaction frequency under different cellular stresses
Compare interaction patterns in normal versus cancer cells
Subcellular Fractionation and Western Blot:
Separate nuclear, cytoplasmic, and chromatin-bound fractions
Probe with ANP32D antibodies to track redistribution during apoptosis
Monitor post-translational modifications that regulate ANP32D function
TUNEL Assay with ANP32D Co-staining:
Perform TUNEL staining to identify apoptotic cells
Co-stain with ANP32D antibodies to correlate expression with apoptotic status
Quantify relative expression in apoptotic versus non-apoptotic cells
Live-Cell Imaging with Tagged ANP32D and Antibody Validation:
Express fluorescently-tagged ANP32D in living cells
Induce apoptosis and track protein redistribution
Validate observations with fixed-cell antibody staining
Unlike ANP32A and ANP32B that have established roles in influenza virus replication , ANP32D's functions appear more centered on cellular growth control and apoptosis regulation, making these approaches particularly relevant for cancer biology investigations.
ANP32D's tumorigenic properties contrast with the tumor suppressor functions of other family members, positioning it as a potential therapeutic target. Methodological approaches using ANP32D antibodies can facilitate translational research:
Biomarker development:
Quantify ANP32D expression in tumor tissue microarrays using standardized IHC protocols
Correlate expression with clinical outcomes to establish prognostic value
Develop scoring systems for potential clinical implementation
Drug discovery screening:
Use ANP32D antibodies in high-content screening assays to identify compounds that modulate its expression or localization
Develop ELISA-based assays to quantify ANP32D-target protein interactions for inhibitor screening
Validate hits using functional assays in cancer cell models
Therapeutic antibody development:
Perform epitope mapping to identify functionally critical regions of ANP32D
Generate and test neutralizing antibodies that inhibit ANP32D's tumorigenic functions
Evaluate antibody-drug conjugates targeting ANP32D-expressing cancer cells
The absence of the 25 amino acid tumor suppressor region in ANP32D provides a specific target for therapeutic development. Researchers should focus on this unique structural feature when designing interventions.
Advanced methodologies for investigating ANP32D's interactome include:
BioID proximity labeling:
Generate ANP32D-BioID fusion proteins
Identify proximal proteins through biotinylation
Confirm interactions with co-immunoprecipitation using ANP32D antibodies
Compare interactomes between normal and cancer cellular contexts
CRISPR-based screening:
Perform genome-wide CRISPR screens in ANP32D-dependent cancer models
Identify synthetic lethal interactions
Validate hits using ANP32D antibodies to assess expression and localization changes
Quantitative interactomics:
Use SILAC or TMT labeling with ANP32D immunoprecipitation
Quantify differential interactions under various cellular conditions
Create dynamic interaction networks that change during cancer progression
Single-molecule imaging:
Employ super-resolution microscopy with ANP32D antibodies
Track individual ANP32D molecules and their interaction partners
Quantify interaction kinetics and spatial organization
Cryo-EM structural analysis:
These approaches can reveal how ANP32D's interactions differ from those of ANP32A and ANP32B, which have established roles in processes like influenza virus replication .