ANAPC1, also known as mitotic checkpoint regulator (MCPR), is a highly conserved component of the anaphase promoting complex/cyclosome (APC/C). This cell cycle-regulated E3 ubiquitin ligase controls progression through mitosis and the G1 phase of the cell cycle . The APC/C complex, which includes ANAPC1, is responsible for degrading anaphase inhibitors, mitotic cyclins, and spindle-associated proteins to ensure proper sequential progression of mitotic events . Individual APC/C components, including ANAPC1, are expressed at relatively consistent levels across most tissues and cell lines, suggesting they primarily function as part of this complex rather than independently .
ANAPC1 antibodies have been validated for multiple experimental applications, with different antibodies showing varying effectiveness across techniques. The most common applications include:
When designing experiments, researchers should select antibodies specifically validated for their intended application to ensure optimal results .
Most commercially available ANAPC1 antibodies are polyclonal, including those referenced in the search results. Polyclonal antibodies recognize multiple epitopes on the ANAPC1 protein, providing advantages for certain applications:
Detection sensitivity is typically higher as multiple epitopes are recognized
More robust against minor protein conformational changes
Often more effective for applications like immunoprecipitation
The polyclonal antibodies in the search results were developed using different methodological approaches:
When selecting an ANAPC1 antibody, researchers should evaluate whether their experimental question requires detection of specific post-translational modifications, particular protein domains, or total ANAPC1 protein levels .
Based on validation data in the search results, the following samples serve as effective positive controls for ANAPC1 antibody testing:
Including appropriate positive controls is essential for confirming antibody specificity and optimizing experimental conditions .
Recent comprehensive analysis of 2,031 samples revealed significant upregulation of ANAPC1 mRNA in lung squamous cell carcinoma (LUSC) tissues (SMD = 1.97, 95% CI [1.26–2.67]) . This finding was further confirmed at the protein level through immunohistochemical analysis . The clinical significance of this overexpression is substantial:
Functionally, ANAPC1 knockdown inhibits cell proliferation, while overexpression reduces immune cell infiltration and immunotherapy effectiveness . These findings suggest ANAPC1 antibodies are valuable tools for cancer research, particularly in:
Prognostic biomarker development
Therapeutic target validation
Patient stratification studies
Mechanistic investigations of cell cycle dysregulation in cancer
Phosphorylation may play a crucial regulatory role in ANAPC1 function. When using phospho-specific antibodies such as those targeting pSer377 , researchers should consider:
Sample preparation protocols:
Rapid sample collection and processing is essential
Include phosphatase inhibitors in all buffers
Avoid freeze-thaw cycles that may alter phosphorylation states
Validation requirements:
Confirm specificity using phosphatase-treated negative controls
Verify phospho-specificity with competing phospho-peptides
Test cross-reactivity with non-phosphorylated ANAPC1 protein
Experimental design:
Consider cell cycle synchronization to capture dynamic phosphorylation events
Evaluate phosphorylation under different cellular stresses or treatments
Correlate phosphorylation with functional outcomes
Storage considerations:
ANAPC1 is a large protein with a calculated molecular weight of 217 kDa (1944 amino acids), though it typically appears at 200-210 kDa on Western blots . This size creates technical challenges requiring specific optimization:
Sample preparation:
Use strong lysis buffers (RIPA or stronger) with protease inhibitors
Extend denaturation time (10+ minutes at 95°C)
Sonicate samples to shear DNA and improve protein extraction
Gel electrophoresis:
Use low percentage acrylamide gels (6-8%) or gradient gels
Run at lower voltage for longer time to improve separation
Include high molecular weight markers
Transfer optimization:
Extend transfer time (overnight at low voltage is often optimal)
Reduce methanol concentration in transfer buffer
Consider semi-dry transfer systems designed for large proteins
Detection protocols:
Troubleshooting approach:
Optimizing IHC protocols for ANAPC1 detection requires attention to several technical aspects:
Antigen retrieval methods:
Antibody dilution optimization:
Signal amplification considerations:
Standard avidin-biotin complex methods work well for most applications
Tyramide signal amplification may improve detection of low abundance targets
Polymer detection systems can reduce background in challenging tissues
Tissue-specific considerations:
Control selection:
Understanding ANAPC1's role in the APC/C complex requires sophisticated experimental approaches:
Co-immunoprecipitation studies:
Use ANAPC1 antibodies to pull down the intact complex
Western blot for other APC/C components to confirm association
Compare complex composition across cell cycle phases
Proximity-based interaction assays:
Proximity ligation assay (PLA) to visualize interactions in situ
FRET/BRET approaches for real-time interaction dynamics
BioID or APEX2 proximity labeling to identify novel interactions
Functional assays:
Substrate ubiquitination assays with reconstituted complexes
Cell cycle progression analysis after ANAPC1 depletion/mutation
Live cell imaging with fluorescently tagged ANAPC1 and substrates
Structural approaches:
Cryo-EM analysis of complete APC/C complex
Domain mapping through deletion/mutation analysis
Cross-linking mass spectrometry to identify interaction interfaces
Cell cycle synchronization methods:
Double thymidine block for G1/S boundary
Nocodazole treatment for M phase
Analyze ANAPC1 expression, localization and modification across phases
The highly conserved nature of ANAPC1 across species suggests evolutionary importance in APC/C function , making comparative studies across model organisms potentially informative.
Recent research has identified interactions between ANAPC1 and microRNAs in disease contexts . To investigate these interactions, researchers can employ several methodological approaches:
Expression correlation analysis:
Quantify ANAPC1 and candidate miRNAs across patient samples
Perform correlation analysis to identify significant associations
Stratify by disease subtype, stage, or outcome
Direct binding assessment:
Luciferase reporter assays with ANAPC1 3'UTR constructs
miRNA mimic/inhibitor transfection followed by ANAPC1 quantification
RNA immunoprecipitation to capture ANAPC1 mRNA-miRNA complexes
Functional validation:
CRISPR-mediated mutation of miRNA binding sites in ANAPC1
Rescue experiments with miRNA-resistant ANAPC1 constructs
Phenotypic assays following miRNA or ANAPC1 modulation
Clinical relevance assessment:
Patient sample analysis stratified by ANAPC1/miRNA expression patterns
Survival analysis based on ANAPC1/miRNA signatures
Development of multivariate prediction models incorporating both factors
Therapeutic targeting strategies:
miRNA delivery approaches to modulate ANAPC1 expression
Small molecule screens to identify compounds disrupting miRNA-ANAPC1 interactions
Combination approaches targeting both ANAPC1 and associated miRNAs
Emerging microfluidics-enabled antibody discovery platforms can be adapted for ANAPC1 antibody development or screening:
Single-cell compartmentalization strategy:
Detection optimization:
Validation approach:
Secondary screening for cross-reactivity with related proteins
Epitope binning to identify antibodies targeting different regions
Functional assays to identify antibodies that modulate ANAPC1 activity
Technical considerations:
Application-specific optimization:
For phospho-specific antibodies, use phosphorylated peptides as targets
For domain-specific antibodies, use truncated protein constructs
For neutralizing antibodies, implement functional screening component
This microfluidics approach can dramatically accelerate ANAPC1 antibody development, reducing traditional timeframes from months to weeks .
When faced with contradictory results regarding ANAPC1's role in different cancer contexts, researchers should consider:
Systematic validation approach:
Use multiple antibodies targeting different ANAPC1 epitopes
Employ both RNA and protein detection methods
Include appropriate positive and negative controls
Technical variation analysis:
Standardize sample preparation, fixation, and processing
Compare antibody performance across different batches
Implement quantitative analysis methods with normalization
Biological context evaluation:
Assess cell type-specific ANAPC1 functions
Consider tumor heterogeneity in sample analysis
Evaluate microenvironment influences on ANAPC1 expression/function
Advanced experimental design:
Implement in vivo models with tissue-specific ANAPC1 modulation
Use patient-derived xenografts to maintain tumor heterogeneity
Employ single-cell approaches to resolve population heterogeneity
Integrated data analysis:
Correlate ANAPC1 expression with comprehensive molecular profiling
Perform multivariate analysis incorporating clinical parameters
Apply machine learning approaches to identify contextual patterns
The recent finding of ANAPC1 overexpression in lung squamous cell carcinoma with prognostic implications provides a starting point for systematic investigations across cancer types.
Based on recent findings identifying tenovin-1, carboxyatractyloside, and phycocyanobilin as potential antitumor agents targeting ANAPC1 , researchers can implement a systematic testing approach:
Compound binding validation:
Perform molecular docking simulations to predict binding sites
Validate binding using thermal shift assays or surface plasmon resonance
Conduct competition assays with known ANAPC1 interactors
Functional screening cascade:
Primary screen: cell viability in ANAPC1-overexpressing cancer lines
Secondary screen: impact on APC/C complex formation and activity
Tertiary screen: cell cycle progression and mitotic regulation
Mechanism of action studies:
Assess effects on ANAPC1 protein levels and stability
Evaluate impact on ANAPC1 subcellular localization
Determine effects on ANAPC1 post-translational modifications
Predictive biomarker development:
Correlate drug sensitivity with ANAPC1 expression levels
Identify genetic or molecular features predicting response
Develop companion diagnostic approaches using ANAPC1 antibodies
In vivo validation approach:
Test efficacy in xenograft models with varying ANAPC1 expression
Evaluate pharmacokinetics and biodistribution
Implement combination studies with standard-of-care therapies
This systematic approach leverages the recent finding that ANAPC1 knockdown inhibits cancer cell proliferation , suggesting therapeutic potential in targeting this protein.
Recent research indicates ANAPC1 overexpression reduces immune cell infiltration and immunotherapy effectiveness . To further investigate this immune regulatory role:
Immune profiling in ANAPC1-modulated systems:
Flow cytometry to quantify immune cell populations
Spatial transcriptomics to assess immune cell distribution
Cytokine profiling to identify altered signaling pathways
Mechanistic investigation:
Co-culture systems with ANAPC1-modified cancer cells and immune cells
Evaluation of antigen presentation machinery in ANAPC1-high/low cells
Assessment of immunomodulatory molecule expression (PD-L1, etc.)
Clinical correlation studies:
IHC analysis of ANAPC1 expression and immune infiltrates in patient samples
Correlation of ANAPC1 levels with immunotherapy response metrics
Development of composite biomarkers incorporating ANAPC1 and immune signatures
Therapeutic targeting approaches:
Combination strategies targeting ANAPC1 and immune checkpoints
Evaluation of immunotherapy sensitization through ANAPC1 inhibition
Development of ANAPC1-targeting antibody-drug conjugates
Translational validation:
Prospective analysis of ANAPC1 expression in immunotherapy trials
Preclinical models testing ANAPC1-based patient selection strategies
Assessment of dynamic ANAPC1 changes during immunotherapy
Understanding ANAPC1's immunomodulatory functions could significantly impact patient selection strategies for cancer immunotherapies and lead to novel combination approaches.