CEP164 is a 1,460-amino acid protein localized to the mother centriole, where it regulates:
Primary cilia formation: Mediates vesicular docking to the mother centriole, a prerequisite for ciliogenesis .
Cell cycle progression: Essential for G2/M checkpoint control and chromosome segregation .
DNA damage response (DDR): Interacts with ATR/ATM kinases to maintain genomic stability .
Mutations in CEP164 are linked to nephronophthisis (NPHP15) and Senior-Løken syndrome, disorders characterized by renal and retinal degeneration .
Commercial and research-grade CEP164 antibodies are validated for diverse applications, including Western blot (WB), immunofluorescence (IF), and immunohistochemistry (IHC). Key examples include:
Ciliogenesis: Depletion of CEP164 via siRNA abolishes primary cilium formation by blocking vesicle docking at the mother centriole .
Cell Cycle Defects: CEP164-null cells exhibit impaired S-phase progression and increased apoptosis .
Disease Modeling: Antibodies aid in detecting CEP164 loss in renal epithelial cells, modeling nephronophthisis .
Immunofluorescence: CEP164 antibodies label distal appendages of the mother centriole, distinguishing them from subdistal appendage markers like ninein .
Western Blot: Detects a ~200 kDa band in human cell lysates, validated by siRNA knockdown .
Renal Ciliopathies: CEP164 dysfunction disrupts renal tubule integrity, promoting fibrosis via epithelial-to-mesenchymal transition (EMT) .
Cancer: Overexpression correlates with centrosome amplification, a hallmark of genomic instability in tumors .
Validation: Antibody specificity confirmed via siRNA knockdown and rescue experiments .
Storage: Stable at -80°C in PBS or Tris-glycine buffer; avoid freeze-thaw cycles .
Limitations: Cross-reactivity with rodent CEP164 varies by product .
Recent genome-editing studies show that CEP164-null cells fail to form cilia but retain normal proliferation, underscoring its cilia-specific role . Additionally, CEP164’s WW domain is critical for interacting with ciliogenesis regulators like NudCL2 .
CEP164 is a 164 kDa centrosomal protein comprising 1,460 amino acid residues that localizes specifically to the distal appendages of the mother centriole. It plays a critical role in primary cilium formation, as demonstrated by siRNA experiments showing that only 3.6% of Cep164-depleted cells formed primary cilia compared to 95% of control cells . CEP164 functions in microtubule organization and maintenance for primary cilia formation and is involved in G2/M checkpoint regulation and nuclear divisions . Its expression is typically limited to the mother centriole, making it a valuable marker for distinguishing between mother and daughter centrioles in research applications .
CEP164 is particularly important in research because:
It serves as a specific marker for mature centrioles
It plays essential roles in ciliogenesis
Dysfunction has been implicated in ciliopathies including Bardet-Biedl syndrome and Meckel-Gruber syndrome
It forms distinctive bar- or ring-like structures at the base of primary cilia that can be visualized through immunofluorescence
Different CEP164 antibodies target distinct regions of the protein and demonstrate varying efficacy across applications:
For immunofluorescence applications, antibodies targeting the N-terminal fragment (1-298) have been successfully used to identify CEP164's localization to the mother centriole . For Western blot applications, these antibodies typically detect a band of approximately 200 kDa (despite the predicted 164 kDa size), possibly due to the protein's relatively acidic isoelectric point (5.32) .
When selecting an antibody, consider:
The specific application (WB, IF/ICC, IHC, IP)
Required species reactivity
Whether a conjugated antibody would simplify your protocol
The region of CEP164 that should be targeted based on your research question
Optimizing Western blot protocols for CEP164 requires attention to several key factors:
Sample preparation: For centrosomal proteins like CEP164, enrichment techniques may improve detection. Consider using centrosome purification protocols or specific lysis buffers that preserve large protein complexes .
Gel selection: Due to CEP164's high molecular weight (~200 kDa observed size), use low percentage (6-8%) polyacrylamide gels or gradient gels to ensure proper resolution .
Transfer conditions: For large proteins like CEP164:
Extend transfer time (overnight at low voltage)
Use PVDF membranes rather than nitrocellulose
Add SDS (0.1%) to transfer buffer to improve large protein migration
Antibody dilution: Most CEP164 antibodies work optimally at dilutions between 1:200-1:1000 for Western blot applications . Initial optimization should test multiple dilutions.
Detection system: Enhanced chemiluminescence systems with extended exposure times may be necessary for optimal detection.
Controls: Include positive controls such as HEK-293 cell lysate, which has been validated for CEP164 detection .
Expected results: Anticipate a band at approximately 200 kDa, which is higher than the calculated 164 kDa, due to the protein's acidic properties .
If non-specific bands appear, further optimization of blocking conditions (5% BSA instead of milk) and more stringent washing steps may improve results.
CEP164 represents an excellent marker for distinguishing mother from daughter centrioles because it specifically localizes to the distal appendages of mature mother centrioles. When implementing this approach:
Immunofluorescence protocol optimization:
Expected staining pattern:
Experimental verification:
In studies of primary cilia formation, researchers observed that CEP164 "always associated with the one centriole that was located at the base of the PC, in agreement with the fact that only the mature centriole is able to initiate PC formation"
The ring-like structure represents the distal appendages of the mother centriole, which are absent in daughter centrioles
Additional markers for confirmation:
This approach enables precise identification of centriole maturation status in studies of centrosome duplication, cell cycle progression, and ciliogenesis.
Thorough validation of a new CEP164 antibody requires multiple controls to ensure specificity and reliability:
Positive cellular controls:
Negative controls:
Specificity controls:
Application-specific controls:
For IF: Compare with previously characterized CEP164 staining patterns (ringlike structure at mother centriole)
For WB: Compare with exogenously expressed tagged CEP164 (myc-tagged CEP164 has been shown to exhibit similar migration patterns to endogenous protein)
For IHC: Include isotype control antibodies
Cross-reactivity assessment:
If studying non-human samples, validate specificity in the relevant species
Sequence alignment analysis can predict potential cross-reactivity
Documentation of these validation steps provides essential quality assurance for subsequent experiments and publications.
Troubleshooting CEP164 immunofluorescence staining involves systematic evaluation of multiple factors:
Fixation optimization:
Test different fixation methods (paraformaldehyde vs. methanol)
Methanol fixation (5-10 minutes at -20°C) often works better for centrosomal proteins
For some epitopes, a combined approach of brief paraformaldehyde fixation followed by methanol may improve results
Permeabilization considerations:
Ensure adequate permeabilization (0.1-0.5% Triton X-100)
Extend permeabilization time if nuclear or centrosomal staining is weak
For difficult epitopes, try alternative detergents like saponin
Antigen retrieval methods:
Antibody dilution optimization:
Signal amplification strategies:
Consider using a biotin-streptavidin system for signal enhancement
Tyramide signal amplification can dramatically increase sensitivity
Background reduction:
Increase blocking time and concentration (5% BSA or 10% normal serum)
Add 0.1-0.3% Triton X-100 to antibody dilution buffers
Include 0.1% Tween-20 in wash buffers and increase washing steps
Microscopy considerations:
CEP164 localizes to a small structure; high-resolution imaging is essential
Consider deconvolution or super-resolution techniques for optimal visualization
Z-stack imaging helps ensure capture of the centriole plane
If non-specific nuclear staining occurs, pre-absorption of the antibody with nuclear extracts can sometimes improve specificity.
CEP164 antibodies provide valuable tools for investigating ciliopathies, as CEP164 dysfunction has been implicated in disorders such as Bardet-Biedl syndrome and Meckel-Gruber syndrome . Implementing these antibodies effectively requires:
Patient sample analysis approaches:
Immunohistochemistry of patient biopsies to assess CEP164 localization abnormalities
Quantification of CEP164 expression levels in patient-derived cells
Comparative analysis of centriole structure and ciliogenesis capacity using CEP164 as a marker
Examination of CEP164 post-translational modifications that might be altered in disease states
Disease modeling experimental design:
Use CEP164 antibodies to validate disease models (patient-derived cells, CRISPR-edited cell lines, animal models)
Quantitative assessment of:
Percentage of cells with CEP164-positive centrioles
Intensity and morphology of CEP164 staining at distal appendages
Correlation between CEP164 localization and primary cilia formation
Pathway analysis methodologies:
Co-immunoprecipitation with CEP164 antibodies to identify altered protein interactions in disease states
Combined CEP164 immunostaining with markers of ciliary trafficking or signaling pathways
Correlative analysis of CEP164 localization with ciliopathy phenotypes
Therapeutic screening applications:
Using CEP164 immunostaining as a readout for high-content screening of compounds that might restore normal centriole function
Monitoring CEP164 localization and function during gene therapy approaches
Implementation considerations:
Different tissue types may require optimized protocols
Control samples from healthy individuals must be processed identically
Quantitative image analysis using standardized parameters enhances reproducibility
This approach enables researchers to determine whether ciliopathy phenotypes correlate with specific defects in CEP164 localization, expression, or function, potentially revealing novel therapeutic targets.
Investigating CEP164 phosphorylation states requires specialized approaches:
Phospho-specific antibody development and validation:
While general CEP164 antibodies detect the protein regardless of phosphorylation state, phospho-specific antibodies may need to be custom-developed
Validation should include:
Western blot comparison before and after phosphatase treatment
Mutation of putative phosphorylation sites (alanine substitution)
Mass spectrometry confirmation of specificity
Cell synchronization protocols:
For G1/S boundary: Double thymidine block
For G2/M: Nocodazole treatment
For mitotic stages: Release from nocodazole with time-course analysis
Confirmation of synchronization using flow cytometry or specific cell cycle markers
Kinase inhibition experiments:
Treatment with specific kinase inhibitors to identify regulatory kinases
Analysis of CEP164 phosphorylation status following inhibitor treatment
Correlation with functional outcomes (centriole maturation, ciliogenesis capability)
Combining techniques for comprehensive analysis:
Immunoprecipitation with general CEP164 antibodies followed by phospho-specific Western blotting
Immunofluorescence using phospho-specific antibodies to determine localization changes
Phos-tag gel electrophoresis to separate differentially phosphorylated forms
Analysis framework:
Quantitative Western blot analysis normalizing phospho-signal to total CEP164
Time-course experiments throughout cell cycle with multiple timepoints
Correlation of phosphorylation changes with functional outcomes
This methodology allows researchers to connect CEP164 phosphorylation events with specific cell cycle transitions and centrosomal functions, revealing regulatory mechanisms governing centriole maturation and primary cilium formation.
Super-resolution microscopy significantly enhances CEP164 visualization, revealing details impossible to resolve with conventional microscopy:
Technique selection considerations:
Structured Illumination Microscopy (SIM): Provides ~100 nm resolution, revealing the ring-like arrangement of CEP164 at distal appendages
Stimulated Emission Depletion (STED): Achieves ~20-50 nm resolution, allowing visualization of individual distal appendage substructures
Single Molecule Localization Microscopy (STORM/PALM): Offers ~10-20 nm resolution, enabling precise positioning of CEP164 relative to other proteins
Sample preparation optimizations:
Minimize sample thickness: Grow cells on high-precision coverslips
Reduce background: Use highly specific primary antibodies and minimize non-specific binding
Implement specialized fixation: Glutaraldehyde addition (0.1-0.2%) can help preserve ultrastructure
Select appropriate fluorophores: Choose bright, photostable dyes compatible with super-resolution techniques
Multi-color experimental design:
Carefully select fluorophore combinations to avoid bleed-through
Include reference markers such as γ-tubulin (centriole barrel) and acetylated tubulin (ciliary axoneme)
Consider sequential staining approaches for difficult combinations
Data analysis approaches:
3D reconstruction to visualize the complete distal appendage structure
Quantitative analysis of CEP164 distribution patterns
Nearest neighbor analysis to determine spatial relationships with other proteins
Temporal analysis in live-cell experiments (if applicable)
Expected insights:
Precise mapping of CEP164 within the ninefold symmetrical arrangement of distal appendages
Detailed visualization of structural changes during centriole maturation
Nanoscale distribution changes in response to cellular signals or in disease models
These approaches have revealed that CEP164 forms distinct "arms" at the distal appendages rather than a continuous ring, information that was not distinguishable using conventional microscopy techniques.
Recent research has connected CEP164 to DNA damage response pathways. An optimal experimental design to investigate this connection includes:
Integrated analysis approach:
Create stable cell lines with inducible CEP164 knockdown or overexpression
Establish complementary systems with CEP164 mutants lacking specific domains
Develop reporter cell lines to simultaneously monitor CEP164 status and DNA damage markers
Damage induction protocols:
Compare different DNA damaging agents (ionizing radiation, UV, chemical agents)
Apply dose-response analysis to identify threshold effects
Use micro-irradiation to induce localized damage for real-time recruitment studies
Multi-parameter assessment:
Immunofluorescence co-localization studies of CEP164 with DNA damage markers (γH2AX, 53BP1)
Western blot analysis of CEP164 modifications following DNA damage
Chromatin immunoprecipitation to assess CEP164 association with damaged DNA regions
Flow cytometry analysis of cell cycle progression and checkpoint activation
Functional outcome measurements:
Comet assay to assess DNA break repair kinetics
Survival assays to determine functional consequences of CEP164 manipulation
Cell cycle analysis to identify specific checkpoint dependencies
Assessment of genomic stability markers (micronuclei formation, chromosomal aberrations)
Mechanistic dissection:
Proximity labeling approaches to identify damage-specific interaction partners
Domain deletion analysis to map regions required for damage response
Phospho-proteomic analysis to identify damage-induced modifications
RNA-seq to assess transcriptional consequences of CEP164 dysfunction during damage response
This comprehensive approach enables researchers to distinguish CEP164's centrosomal functions from potential direct roles in DNA damage signaling or repair pathways, clarifying this protein's multifunctional nature.
Several cutting-edge technologies are enhancing the research applications of CEP164 antibodies:
Single-cell protein analysis:
Mass cytometry (CyTOF) incorporating CEP164 antibodies allows simultaneous analysis of centrosomal status and multiple cellular parameters at single-cell resolution
Microfluidic approaches for temporal analysis of CEP164 dynamics in individual cells
Proximity labeling approaches:
BioID or APEX2 fusions with CEP164 enable identification of proximal proteins in living cells
Spatial proteomics revealing the CEP164 "interactome" at different cell cycle stages
TurboID variants for rapid labeling of transient interactions
Live-cell imaging innovations:
Development of intrabodies or nanobodies against CEP164 for live-cell applications
Split-fluorescent protein complementation assays to visualize CEP164 interactions in real-time
Lattice light-sheet microscopy for extended, high-resolution imaging of CEP164 dynamics
Spatial transcriptomics integration:
Combining CEP164 immunostaining with spatial transcriptomics to correlate centrosome status with local gene expression patterns
RNA-protein co-detection methods to identify RNAs associated with CEP164-positive structures
These emerging methods are expanding our understanding of CEP164 beyond static localization to dynamic functional relationships in diverse cellular contexts, offering new insights into centrosome biology and ciliopathies.