Gene Name: CDC23
Protein Aliases: APC8, CUT23, ANAPC8
Molecular Weight: Reported as 64 kDa (Cell Signaling Technology) and 68 kDa (Boster Bio) , reflecting potential isoform differences or experimental conditions
Structure: Contains tetratricopeptide repeat (TPR) domains critical for APC/C assembly
Conservation: Evolutionarily conserved from plants (e.g., Arabidopsis thaliana) to mammals
Cell Cycle Regulation:
APC8 facilitates APC/C-mediated ubiquitination of securin and cyclin B, enabling metaphase-to-anaphase transition .
Disease Associations:
Western Blot: Detects endogenous APC8 at 64-68 kDa in HeLa (human) , K562 (human) , and plant tissues .
Immunoprecipitation: Validated for APC/C complex isolation .
Cross-Reactivity:
| Subunit | Role in APC/C Assembly | Interaction Partners |
|---|---|---|
| APC8/CDC23 | Scaffold for TPR subcomplex | APC3 (CDC27), APC6 (CDC16) |
| APC10 | Substrate recognition | Coactivators (CDC20, CDH1) |
Critical Residues:
Cell Cycle Dependence: APC8 levels remain constant, but APC/C activity peaks during mitosis .
Post-Translational Modifications: Phosphorylation at Ser/Thr residues modulates coactivator binding .
Therapeutic Targeting: APC8 inhibition could suppress cell proliferation in APC8-overexpressing cancers .
Meiotic Regulation: Arabidopsis APC8 studies suggest conserved roles in gametogenesis .
APC/C Dynamics: Single-molecule imaging of APC8-containing complexes to resolve ubiquitination mechanisms .
APC8 (also known as CDC23 or ANAPC8) is a highly conserved component of the anaphase-promoting complex/cyclosome (APC/C), which functions as a cell cycle-regulated E3 ubiquitin ligase. This complex plays a critical role in controlling progression through mitosis and the G1 phase of the cell cycle by targeting specific proteins for degradation. The APC/C is responsible for degrading anaphase inhibitors, mitotic cyclins, and spindle-associated proteins, ensuring that mitotic events occur in the proper sequence . Due to its central role in cell cycle regulation, APC8 is an important target for research in cancer biology, developmental processes, and fundamental cell biology.
APC8 antibodies are primarily used in Western blotting (WB) and enzyme-linked immunosorbent assays (ELISA) . They can be utilized to detect and quantify APC8 protein expression, analyze cell cycle-dependent changes in APC/C components, evaluate APC8 localization through immunofluorescence, and investigate protein-protein interactions via co-immunoprecipitation. These applications enable researchers to study APC8's role in cell cycle progression, mitotic regulation, and potential involvement in disease mechanisms.
The expected molecular weight of APC8 protein in Western blot analysis is approximately 68 kDa . This information is crucial for correctly identifying the protein band of interest and distinguishing it from non-specific binding or other cross-reactive proteins. When conducting Western blot experiments, researchers should be aware that apparent molecular weights can vary slightly depending on the cell type, post-translational modifications, and the specific gel system used.
For optimal Western blotting results with APC8 antibodies, consider the following protocol refinements:
Sample preparation: Lyse cells in a buffer containing protease inhibitors to prevent protein degradation.
Protein loading: Load 20-50 μg of total protein per lane for cell lysates.
Gel percentage: Use 8-10% SDS-PAGE gels for optimal separation around the 68 kDa range.
Transfer conditions: Transfer proteins to PVDF or nitrocellulose membranes at 100V for 1 hour or 30V overnight.
Blocking: Block membranes with 5% non-fat milk or BSA in TBS-T for 1 hour at room temperature.
Primary antibody incubation: Dilute APC8 antibody at 1:500-1:1000 in blocking buffer and incubate overnight at 4°C .
Washing: Wash 3-5 times with TBS-T, 5 minutes each.
Secondary antibody: Use appropriate HRP-conjugated anti-rabbit IgG at 1:5000-1:10000 dilution.
Detection: Use enhanced chemiluminescence (ECL) for visualization.
Validation has been performed on RAW264.7 cells, confirming the specificity of anti-APC8 antibody for Western blot applications .
When working with APC8 antibodies, implement the following controls to ensure result validity:
Positive control: Include a cell line known to express APC8 (e.g., RAW264.7 cells have been validated) .
Negative control: Consider using:
Loading control: Use housekeeping proteins (β-actin, GAPDH, tubulin) to normalize protein loading
Molecular weight marker: Include to confirm the target band appears at the expected size (68 kDa)
Peptide competition assay: Pre-incubate the antibody with the immunizing peptide (human APC8 amino acids 220-269) to verify specificity
These controls help distinguish specific signals from artifacts, ensuring experimental reliability and reproducibility.
While the available APC8 antibodies are primarily validated for Western blot and ELISA applications , researchers interested in using them for flow cytometry should consider the following approach:
Antibody selection: Choose an APC8 antibody that recognizes the native conformation of the protein or an extracellular epitope if examining surface expression.
Cell preparation:
Titration: Perform antibody titration to determine optimal concentration and minimize background
Controls:
Unstained cells
Isotype controls (rabbit IgG)
FMO (fluorescence minus one) controls
Positive and negative cell populations if available
Panel design: When incorporating into multicolor panels, consider:
Validation: Confirm flow cytometry results with orthogonal methods like Western blotting
To investigate cell cycle-dependent changes in the APC/C complex using APC8 antibodies:
Synchronization strategies:
Double thymidine block (G1/S boundary)
Nocodazole treatment (mitotic arrest)
Serum starvation/release (G0/G1 transition)
Time course analysis:
Co-immunoprecipitation:
Use anti-APC8 antibody to pull down the entire APC/C complex
Analyze co-precipitated proteins by Western blot or mass spectrometry
Examine changes in complex composition across cell cycle phases
Chromatin association:
Perform subcellular fractionation at different cell cycle phases
Analyze APC8 distribution between cytoplasmic and chromatin-bound fractions
Correlate with APC/C activity markers (substrate degradation)
Post-translational modifications:
Use phospho-specific antibodies after immunoprecipitation with anti-APC8
Apply 2D gel electrophoresis to separate phosphorylated forms
Consider mass spectrometry to identify specific modifications
This methodology permits dynamic assessment of APC/C composition, localization, and activity throughout the cell cycle, illuminating regulatory mechanisms of this essential complex in normal and pathological states.
Validating antibody specificity is crucial for reliable experimental outcomes. For APC8 antibodies, consider these rigorous validation approaches:
Genetic validation:
Peptide competition:
Cross-species reactivity:
Detection of recombinant protein:
Express tagged APC8 (His, GST, or FLAG-tagged)
Confirm detection by both anti-APC8 and anti-tag antibodies
Multiple antibody concordance:
Mass spectrometry:
Immunoprecipitate with anti-APC8 antibody
Analyze pulled-down proteins by mass spectrometry
Confirm presence of APC8 and known interacting partners
To investigate APC8's role in cancer progression using available antibodies, consider this comprehensive experimental approach:
Expression analysis across cancer types:
Create tissue microarrays of tumor and matched normal samples
Perform immunohistochemistry with anti-APC8 antibody
Quantify expression differences and correlate with clinical parameters
Functional studies in cancer cell lines:
Modulate APC8 expression (overexpression, knockdown, knockout)
Analyze effects on:
Cell proliferation (growth curves, colony formation)
Cell cycle distribution (propidium iodide staining, flow cytometry)
Mitotic progression (time-lapse imaging)
Chromosome segregation (metaphase spreads)
Genomic stability (micronuclei formation)
Substrate degradation kinetics:
Synchronize cells at different cell cycle phases
Perform Western blots for APC8 and key substrates (cyclins, securin)
Compare substrate half-lives between normal and cancer cells
APC/C complex integrity:
In vivo models:
Generate xenografts with APC8-modulated cancer cells
Analyze tumor growth, invasion, and metastatic potential
Use IHC with anti-APC8 to analyze expression in tumor sections
This multifaceted approach allows for comprehensive assessment of APC8's potential roles in cancer initiation, progression, and therapeutic response.
When working with APC8 antibodies, researchers may encounter these common issues and their corresponding solutions:
In particularly challenging cases, consider using the validated applications (Western blot, ELISA) rather than adapting the antibody to non-validated applications without thorough optimization.
When analyzing variations in APC8 detection patterns, consider these interpretation frameworks:
Cell type-specific variations:
Different cell types may express APC8 at varying levels based on proliferation rates and cellular functions
Certain cell types may express splice variants or isoforms that alter detection patterns
Post-translational modifications may differ between cell types, affecting antibody recognition
Cell cycle-dependent variations:
APC/C components show subtle regulation throughout the cell cycle
Sample timing relative to cell cycle phase may affect detection patterns
Compare with established cell cycle markers to contextualize observations
Experimental condition influences:
Stress conditions (hypoxia, nutrient deprivation) may alter APC8 expression or localization
Treatment with cell cycle inhibitors can affect APC/C complex composition
Growth factors or hormones may indirectly regulate APC8 through upstream pathways
Analytical framework:
Always normalize to appropriate loading controls for quantitative comparisons
Consider relative changes rather than absolute signal intensities
Validate observations with orthogonal detection methods
Correlate Western blot results with functional outcomes (substrate degradation kinetics)
Remember that APC8 functions as part of a multi-subunit complex, so variations may reflect changes in the entire APC/C rather than APC8 alone. When possible, analyze multiple APC/C components to gain a more comprehensive understanding of complex regulation.
Quantitative Western blot analysis:
Densitometry normalization to loading controls (β-actin, GAPDH)
Multiple biological replicates (minimum n=3)
Present data as mean ± standard deviation or standard error
Apply appropriate statistical tests:
Two groups: Student's t-test (parametric) or Mann-Whitney U test (non-parametric)
Multiple groups: ANOVA with post-hoc tests (Tukey, Bonferroni)
Time course: Repeated measures ANOVA
Correlation analyses:
Pearson correlation (parametric) or Spearman rank correlation (non-parametric) for:
APC8 expression versus clinical parameters
APC8 levels versus substrate degradation rates
Correlation with other APC/C components
Survival analyses for clinical studies:
Stratify patients by APC8 expression levels (high/low based on median)
Generate Kaplan-Meier survival curves
Apply log-rank test for significance
Consider multivariate Cox regression to control for confounding variables
Experimental design considerations:
Perform power calculations to determine appropriate sample sizes
Define statistical significance threshold (typically p<0.05)
Consider multiple testing corrections for large-scale analyses (Bonferroni, FDR)
Report both statistical significance and effect sizes
Utilize appropriate graphical representations (box plots, scatter plots with error bars)
These statistical approaches help distinguish biologically meaningful findings from experimental noise, particularly important when dealing with the subtle regulatory changes often observed in APC/C component expression.
APC8 antibodies can be powerful tools for exploring protein-protein interactions within the APC/C complex using these advanced methodologies:
Co-immunoprecipitation (Co-IP):
Proximity ligation assay (PLA):
Combine APC8 antibody with antibodies against suspected interacting partners
Visualize interactions as fluorescent spots when proteins are within 40 nm
Quantify interaction frequency and subcellular localization
Particularly useful for transient or context-dependent interactions
Bimolecular fluorescence complementation (BiFC):
Tag APC8 and potential partners with complementary fluorescent protein fragments
Visualize reconstituted fluorescence when proteins interact
Analyze interaction dynamics in living cells
Validate observed interactions with co-IP using anti-APC8 antibody
Crosslinking mass spectrometry:
Perform chemical crosslinking of protein complexes
Immunoprecipitate with anti-APC8 antibody
Analyze crosslinked peptides by mass spectrometry
Map structural organization of APC/C subunits
FRET/FLIM analysis:
Label APC8 and interacting partners with appropriate fluorophore pairs
Measure energy transfer as indication of protein proximity
Analyze interaction dynamics in real-time
Correlate with cell cycle phases or treatments
These techniques provide complementary information about APC8's interaction network, illuminating both stable core interactions and dynamic regulatory associations that govern APC/C function.
Emerging research suggests APC8 may have functions beyond canonical cell cycle regulation. Future research directions include:
Developmental roles:
Investigate APC8's function in tissue-specific differentiation programs
Explore phenotypes of conditional APC8 knockouts in model organisms
Examine potential parallels to Drosophila studies showing developmental delay and pupal lethality
Use APC8 antibodies to track expression patterns during development
Neurological functions:
Explore APC8's role in post-mitotic neurons
Investigate connections to neurodegeneration or neurodevelopmental disorders
Analyze APC8 localization at neuronal synapses using immunofluorescence
Examine regulation of neuronal protein turnover by APC/C
Stress response pathways:
Investigate APC8 regulation under cellular stress conditions
Explore potential non-canonical substrates targeted during stress
Analyze post-translational modifications of APC8 during stress response
Examine APC8-dependent regulation of stress response factors
Metabolic regulation:
Study potential connections between APC/C activity and metabolic pathways
Investigate APC8's role in regulating metabolic enzymes or signaling proteins
Analyze potential metabolic phenotypes in APC8-depleted cells
Explore connections to nutrient sensing pathways
Therapeutic targeting:
Develop small molecule inhibitors specifically targeting APC8-substrate interactions
Explore synthetic lethality approaches in cancer therapy
Investigate APC8 as a biomarker for response to cell cycle-targeting drugs
Use APC8 antibodies to monitor drug effects on complex integrity
These research directions could significantly expand our understanding of APC8 biology beyond its established mitotic functions, potentially revealing novel therapeutic targets or diagnostic approaches.
When utilizing APC8 antibodies in research, scientists should remember these essential considerations:
These considerations help ensure that research using APC8 antibodies produces reliable, reproducible, and biologically meaningful results that advance our understanding of this important cell cycle regulator.
To remain at the forefront of APC8 antibody technology and applications, researchers should implement these strategies:
Literature monitoring:
Set up automated alerts for new publications regarding APC8 or APC/C
Regularly review high-impact journals in cell biology, cancer research, and proteomics
Pay particular attention to methods sections describing novel applications
Follow key research groups working on cell cycle regulation and APC/C biology
Scientific community engagement:
Attend relevant conferences focusing on cell cycle, mitosis, or protein degradation
Participate in specialized workshops on antibody validation techniques
Join research interest groups or online forums discussing APC/C biology
Engage with core facilities specializing in proteomics or antibody applications
Resource utilization:
Regularly check antibody validation initiatives and databases
Consult repositories of validation data from large-scale projects
Explore antibodypedia.com for user-contributed data on antibody performance
Review manufacturer websites for updated application notes and citations
Collaborative approaches:
Establish collaborations with groups specializing in antibody development
Participate in multi-lab validation studies
Share reagents and protocols through material transfer agreements
Contribute to community standards for antibody validation
Technology integration:
Stay informed about emerging antibody technologies (nanobodies, recombinant antibodies)
Explore complementary approaches (CRISPR, optogenetics) for functional validation
Consider how new imaging or proteomics technologies can enhance antibody applications
Evaluate computational approaches for predicting antibody performance