CEP126 (Centrosomal Protein 126), also known as KIAA1377, is a 126 kDa protein critical for microtubule organization, centrosome function, and primary cilium formation . The CEP126 antibody is a research tool designed to detect and study this protein’s localization and function in cellular processes. It is primarily used in immunofluorescence (IF), Western blotting (WB), and immunohistochemistry (IHC) to visualize CEP126 in centrosomes, pericentriolar satellites, and cilium bases .
CEP126 antibodies enable studies of:
Centrosome dynamics: Tracking CEP126 localization during interphase, mitosis, and cytokinesis .
Microtubule organization: Assessing radial microtubule array disruption in CEP126-depleted cells .
Primary cilium formation: Detecting defects in cilium assembly after CEP126 knockdown .
Protein interactions: Investigating binding partners (e.g., p150Glued of the dynein-dynactin complex) .
CEP126 regulates microtubule anchoring and pericentriolar satellite transport. Its depletion causes:
Disorganized microtubules: Loss of radial MT arrays, leading to spindle defects in mitosis .
Satellite dispersion: PCM1 and pericentrin lose centrosomal localization, impairing MT organization .
Cilium defects: Reduced ciliation in serum-starved cells (e.g., hTERT-RPE-1) .
CEP126 interacts with p150Glued, a dynein-dynactin subunit, suggesting involvement in satellite transport . Truncation mutants lacking the C-terminal domain disrupt MT organization, highlighting its structural role .
Localization variability: Endogenous CEP126’s pericentriolar satellite localization is hard to detect in fixed cells, necessitating live imaging or overexpression .
Antibody specificity: Ensure validation for target epitope (e.g., AA 559–670) to avoid off-target binding .
Experimental conditions: Optimize dilution (1:20–1:200 for IHC) and buffer compatibility (e.g., PBS with glycerol) .
CEP126 is a centrosomal protein that localizes to the centrosome, pericentriolar satellites, and the base of the primary cilium. It plays crucial roles in:
Regulation of microtubule organization at the centrosome
Pericentriolar satellite transport to the centrosome
Primary cilium formation
Mitotic spindle organization
Cytokinesis
Research has demonstrated that CEP126 depletion results in dispersion of pericentriolar satellites and disruption of the radial organization of microtubules. Additionally, it causes disorganization of the mitotic spindle and impairs primary cilium formation in hTERT-RPE-1 and IMCD3 cells .
CEP126 antibodies can be detected through multiple techniques:
| Method | Applications | Typical Dilutions |
|---|---|---|
| Western Blot | Protein expression | 1:1000-1:5000 |
| Immunofluorescence | Protein localization | 1:100-1:500 |
| Immunohistochemistry | Tissue localization | 1:20-1:200 |
| ELISA | Quantitative detection | 1:2000-1:10000 |
For optimal results, researchers should validate antibodies for their specific application and cell type. When using immunofluorescence, co-staining with centrosomal markers like γ-tubulin is recommended for accurate identification of centrosomal localization .
When selecting a CEP126 antibody, consider:
Specificity: Confirm the antibody specifically recognizes CEP126 with minimal cross-reactivity
Host species: Consider compatibility with other antibodies for co-localization studies
Application validation: Ensure the antibody is validated for your specific application (WB, IF, IHC, ELISA)
Epitope recognition: Choose antibodies recognizing functionally relevant domains based on your research questions
Clonality: Polyclonal antibodies may recognize multiple epitopes, while monoclonal antibodies offer higher specificity
For critical experiments, validation through siRNA knockdown controls is recommended to confirm specificity and sensitivity of the selected antibody .
To visualize CEP126 localization:
Fix cells with 4% paraformaldehyde (10 minutes) or ice-cold methanol (5 minutes) depending on epitope accessibility
Permeabilize with 0.2% Triton X-100 if using paraformaldehyde fixation
Block with 3-5% BSA or normal serum from the secondary antibody host species
Incubate with anti-CEP126 antibody (typically 1:100-1:500 dilution)
Counterstain with markers for:
Centrosome (γ-tubulin)
Cilia (acetylated tubulin)
Pericentriolar satellites (PCM1)
Use confocal microscopy for best resolution of centrosomal and ciliary structures
Live imaging can also be performed with transfection of fluorescently tagged CEP126, though caution should be taken as overexpression may affect normal protein function .
To investigate CEP126 functional domains:
Expression of deletion mutants: Create constructs lacking specific domains (e.g., C-terminal domain, centrosome localization domain)
The 1-967 truncation mutant (lacking C-terminal domain) localizes to the centrosome but severely impairs microtubule organization
The 520-655 region contains the putative centrosome-localization domain
Domain-specific antibodies: Use antibodies recognizing specific domains to study localization patterns
Point mutations: Introduce specific mutations in functional domains to study their effects
Rescue experiments: Deplete endogenous CEP126 using siRNA and express RNAi-resistant wildtype or mutant CEP126 to determine which domains are essential for function
Research has shown that expression of the 1-967 CEP126 truncation mutant caused >80% reduction in ciliated cells, demonstrating the importance of the C-terminal domain in cilium formation .
To study CEP126's role in cilium formation:
Cell models: Use cilium-forming cell lines like hTERT-RPE-1 or IMCD3
Cilium induction protocol:
Grow cells to 70-80% confluence
Serum-starve for 24-48 hours to induce cilium formation
Fix and stain with anti-acetylated tubulin (cilium marker) and anti-CEP126
Quantification metrics:
Percentage of ciliated cells
Cilium length
CEP126 localization at cilium base
Perturbation approaches:
siRNA knockdown (two different siRNAs to control for off-target effects)
Expression of truncation mutants (particularly the 1-967 mutant)
CRISPR/Cas9 knockout (for long-term studies)
Rescue experiments:
Express RNAi-resistant wildtype CEP126 in knockdown cells
Studies have demonstrated that CEP126 depletion reduced the percentage of ciliated cells by >80%, and similar results were observed with expression of the 1-967 truncation mutant, highlighting CEP126's essential role in cilium formation .
To investigate CEP126 protein interactions:
Co-immunoprecipitation:
Transfect cells with tagged CEP126 (e.g., Flag-tagged)
Lyse cells in buffer containing 50 mM Tris-HCl pH 7.5, 150 mM NaCl, 1% NP-40, and protease inhibitors
Immunoprecipitate with anti-tag antibody
Analyze by Western blot for interacting proteins
Proximity ligation assay (PLA):
Allows detection of protein interactions in situ
Requires antibodies from different species against CEP126 and potential interactors
Yeast two-hybrid screening:
Use CEP126 domains as bait to identify novel interactors
Mass spectrometry:
Immunoprecipitate CEP126 and perform mass spectrometry analysis
Compare results from different cell types or conditions
CEP126 has been shown to interact with p150Glued, a subunit of the dynein-dynactin complex, which is involved in the transport of pericentriolar satellites. Both full-length CEP126 and the 1-967 truncation mutant interact with p150Glued .
For quantitative assessment of CEP126:
Western blot quantification:
Use β-actin or GAPDH as loading controls
Perform densitometry analysis with software like ImageJ
Run multiple biological replicates for statistical significance
ELISA:
Commercial CEP126 ELISA kits are available with detection ranges of 7.813-500 ng/mL
Minimum detection limit: approximately 4.688-7.813 ng/mL
Sample types: serum, plasma, tissue homogenates
RT-qPCR:
Design primers spanning exon-exon junctions
Normalize to housekeeping genes
Validate knockdown efficiency at the mRNA level
Immunofluorescence quantification:
Measure fluorescence intensity at the centrosome
Use automated image analysis software for unbiased quantification
Compare to control conditions with consistent imaging parameters
When using ELISA for CEP126 detection, sample dilution (at least 1:2 with Sample Dilution Buffer) is recommended for optimal results .
Challenges and solutions for studying CEP126's role in microtubule organization:
Dynamic nature of microtubules:
Use live imaging with EB1-GFP to track growing microtubule plus ends
Perform microtubule regrowth assays after nocodazole treatment
Distinguishing direct vs. indirect effects:
Compare acute (siRNA) vs. chronic (CRISPR) depletion methods
Use rescue experiments with domain mutants
Visualization of microtubule networks:
Use COS7 cells, which have well-defined astral microtubule arrays
Employ super-resolution microscopy techniques
Quantification challenges:
Develop automated image analysis methods to quantify:
Microtubule radial organization
Centrosomal microtubule density
Microtubule dynamics parameters
Distinguishing CEP126 functions:
Use carefully timed expression of mutants (e.g., 20-hour expression of the 1-967 truncation mutant)
Combine with cell cycle synchronization methods
Research has shown that in CEP126-depleted cells, microtubules do not radiate from the centrosome, with most microtubules randomly oriented throughout the cytoplasm. Less than 20% of CEP126 knockdown cells showed a focused microtubule array compared to more than 80% of control cells .
Comparison of detection methods:
| Method | Advantages | Limitations | Best Applications |
|---|---|---|---|
| Antibody-based (IF, IHC, WB) | - Detects endogenous protein - Provides localization information - Compatible with fixed samples | - Depends on antibody quality - May have cross-reactivity - Limited quantification | - Protein localization - Expression studies - Tissue distribution |
| Fluorescent protein tagging | - Live imaging compatible - High specificity - Enables dynamics studies | - Overexpression artifacts - Tag may affect function - Requires transfection | - Protein dynamics - Live-cell tracking - Structure-function studies |
| RNA-based (qPCR, RNA-seq) | - Highly quantitative - Genome-wide context - No antibody required | - mRNA ≠ protein levels - No localization data - No post-translational info | - Expression profiling - Transcriptional regulation - Knockdown validation |
| Mass spectrometry | - Unbiased detection - Identifies modifications - Interaction studies | - Complex sample prep - Limited spatial info - Requires specialized equipment | - Proteomics - Post-translational modifications - Protein interactions |
For the most comprehensive understanding of CEP126 biology, combining multiple approaches is recommended. For example, validating antibody specificity by comparing with tagged protein expression and confirming knockdown by both qPCR and Western blot .
To study CEP126's role in pericentriolar satellite function:
Co-localization studies:
Co-stain for CEP126 and pericentriolar satellite markers (PCM1, pericentrin)
Use dual-color live imaging with tagged proteins
Apply super-resolution microscopy techniques
Functional assays:
Measure pericentriolar satellite motility using live imaging
Assess satellite distribution after CEP126 depletion
Quantify pericentrin and PCM1 dispersion
Microtubule dependency:
Treat with nocodazole to disrupt microtubules
Examine satellite redistribution in CEP126 normal vs. depleted cells
Motor protein involvement:
Investigate the interaction between CEP126 and p150Glued
Use dynein inhibitors to compare with CEP126 depletion effects
Perform co-immunoprecipitation of CEP126 with motor proteins
Research has shown that in CEP126-depleted cells, PCM1-positive pericentriolar satellites become dispersed throughout the cytoplasm rather than clustering around the centrosome. This phenotype resembles what occurs when dynein-dynactin function is disrupted, consistent with CEP126's interaction with p150Glued .
To study CEP126's role in cell cycle:
Cell synchronization approaches:
Double thymidine block for G1/S synchronization
Nocodazole block and release for M-phase studies
Serum starvation/stimulation for G0/G1 transition
Cell cycle markers:
Co-stain CEP126 with:
Cyclin B1 (G2/M)
PCNA (S phase)
Phospho-histone H3 (mitosis)
Live cell cycle reporters:
FUCCI system to track cell cycle progression
Combine with CEP126-FP fusions
Functional assays:
Flow cytometry analysis of cell cycle distribution after CEP126 depletion
Time-lapse imaging of mitotic progression
Spindle assembly checkpoint activation assessment
Mitotic defect analysis:
Quantify spindle abnormalities in CEP126-depleted cells
Measure mitotic duration
Assess cytokinesis completion rates
Research has demonstrated that CEP126 localizes to the centrosome throughout the cell cycle and to the midbody during cytokinesis. CEP126 depletion induces disorganization of the mitotic spindle, suggesting it plays important roles in mitotic progression .
Common issues and solutions:
High background staining:
Increase blocking time/concentration (5% BSA, 1 hour)
Reduce primary antibody concentration
Include 0.1-0.3% Triton X-100 in antibody dilution buffers
Use more stringent washing (0.1% Tween-20 in PBS, 3×10 minutes)
No centrosomal signal:
Try different fixation methods (PFA vs. methanol)
Optimize antigen retrieval (if applicable)
Confirm antibody recognizes native protein conformation
Verify epitope accessibility in your experimental conditions
Variable staining intensity:
Standardize cell culture conditions
Control for cell cycle stage (synchronized populations)
Optimize antibody concentration using titration experiments
Use consistent imaging parameters
Non-centrosomal staining:
Validate antibody specificity using siRNA knockdown
Use multiple antibodies targeting different epitopes
Include appropriate negative controls
Cell type variation:
Optimize protocols for each cell line
Consider expression levels in different cell types
Adjust antibody concentration accordingly
Researchers have reported that while CEP126 clearly localizes to the centrosome, the pericentriolar satellite localization is more readily visualized using overexpressed tagged protein rather than by immunolabeling of endogenous CEP126 in fixed cells .
When facing contradictory results:
Methodological differences:
Compare experimental approaches (transient vs. stable knockdown)
Evaluate knockdown efficiency across studies
Consider cell-type specific differences
Examine timing of analyses (acute vs. chronic effects)
Antibody considerations:
Verify antibodies recognize the same epitopes
Confirm specificity in your experimental system
Use multiple antibodies when possible
Functional redundancy:
Consider compensation by related proteins
Investigate potential adaptation in long-term knockdowns
Perform double knockdowns of functionally related proteins
Cell cycle effects:
Synchronize cells to eliminate cell-cycle variability
Analyze phenotypes in specific cell cycle stages
Technical validation:
Include proper controls (positive, negative, and technical)
Use complementary approaches (fixed and live imaging)
Quantify results with appropriate statistical analysis
The complex nature of centrosome biology often results in seemingly contradictory findings. For example, different truncation mutants of CEP126 may show varying effects on microtubule organization depending on the domains affected and expression levels .
For robust quantification:
Automated image analysis workflows:
Use CellProfiler or ImageJ/Fiji for unbiased quantification
Develop macros for batch processing of images
Implement machine learning approaches for complex phenotypes
Quantification metrics for common phenotypes:
| Phenotype | Recommended Metrics | Analysis Approach |
|---|---|---|
| Satellite dispersion | Distance from centrosome | Radial distribution analysis |
| MT organization | Angle variance from centrosome | Radial line scan analysis |
| Cilium formation | % ciliated cells, cilium length | Automated detection and measurement |
| Protein localization | Intensity at centrosome vs. cytoplasm | Ratio analysis with defined ROIs |
| Spindle defects | Spindle angle, pole distance | 3D measurement tools |
Statistical considerations:
Use appropriate statistical tests (t-test, ANOVA)
Account for multiple comparisons
Report effect sizes along with p-values
Include biological replicates (n≥3)
Data presentation:
Include representative images alongside quantification
Use consistent scaling and color schemes
Provide clear legends and methodology descriptions
For robust analysis of microtubule organization defects, researchers have quantified the percentage of cells with focused microtubule arrays, finding that while >80% of control cells showed focused arrays, <20% of CEP126 knockdown cells maintained this organization .
Single-cell methodologies offer several advantages:
Single-cell RNA sequencing:
Reveal cell-to-cell variation in CEP126 expression
Identify co-regulated gene networks
Discover cell-type specific expression patterns
Track changes across cell cycle or differentiation
Single-cell protein analysis:
Mass cytometry (CyTOF) for multi-parameter protein analysis
Single-cell Western blotting for protein heterogeneity
Correlate CEP126 levels with other centrosomal proteins
Live single-cell imaging:
Track dynamic changes in CEP126 localization
Measure protein turnover rates using photobleaching
Correlate phenotypes with expression levels
Examine cell-to-cell variability in response to perturbations
CRISPR screens at single-cell resolution:
Identify genetic interactions with CEP126
Discover context-dependent functions
Link genotype to phenotype at single-cell level
These approaches could reveal how CEP126 expression heterogeneity impacts centrosome function and cilium formation across different cell populations or within developing tissues .
Advanced approaches for antibody validation:
CRISPR knockout validation:
Generate CEP126 knockout cell lines as ultimate negative controls
Perform Western blot and immunofluorescence to confirm antibody specificity
Create epitope-deleted cell lines for domain-specific antibodies
Orthogonal antibody testing:
Compare antibodies recognizing different epitopes
Correlate results from different detection methods
Use tagged proteins as reference standards
Mass spectrometry validation:
Identify all proteins pulled down by CEP126 antibodies
Quantify on-target vs. off-target binding
Map precise epitopes recognized by the antibody
High-throughput antibody screening:
Test multiple antibodies against protein arrays
Evaluate cross-reactivity profiles systematically
Develop antibody specificity scores
In situ antibody validation:
Proximity ligation assays with multiple antibody pairs
Pre-absorption controls with recombinant antigens
Competitive binding assays
For CEP126 antibodies, researchers should be particularly vigilant about cross-reactivity with other centrosomal proteins, as the centrosome contains hundreds of proteins in a small volume .
High-throughput screening strategies:
Genetic screens:
CRISPR/Cas9 knockout libraries targeting the genome
siRNA/shRNA libraries for transient knockdown
cDNA overexpression libraries to identify suppressors
Screening readouts:
High-content imaging for cilium formation
Automated microtubule organization analysis
Reporter assays for centrosome-dependent processes
Chemical screens:
Small molecule libraries targeting kinases, phosphatases
Compounds affecting microtubule dynamics
Targeted degraders (PROTACs) for acute protein depletion
Biosensor approaches:
FRET sensors for protein-protein interactions
Split-GFP complementation for proximity detection
Activity-based probes for functional readouts
Data integration:
Combine genetic and chemical screening data
Apply machine learning for pattern recognition
Integrate with public datasets for broader context
High-throughput approaches could be particularly valuable for identifying factors that regulate CEP126's interactions with the dynein-dynactin complex or proteins involved in pericentriolar satellite transport .
Potential disease connections for CEP126:
Primary ciliopathies:
Given CEP126's role in cilium formation, mutations could contribute to:
Polycystic kidney disease
Bardet-Biedl syndrome
Joubert syndrome
Primary ciliary dyskinesia
Centrosome amplification in cancer:
CEP126 dysregulation might affect:
Centrosome duplication control
Mitotic spindle organization
Genomic stability maintenance
Microcephaly and neurodevelopmental disorders:
Proper centrosome function is critical for neurogenesis
CEP126 mutations could affect neural progenitor division
Brain development might be particularly sensitive to CEP126 dysfunction
Experimental approaches to investigate disease connections:
Patient-derived cell studies
CRISPR engineering of disease-associated mutations
Animal models of CEP126 dysfunction
Tissue-specific knockouts to assess organ-specific effects
The essential role of CEP126 in cilium formation suggests it could be involved in ciliopathies, though specific disease associations have not yet been definitively established in the literature .
Cutting-edge imaging approaches:
Super-resolution microscopy:
STED, STORM, or PALM imaging for nanoscale localization
Resolve subdomains within centrosomes (~200-500 nm)
Map precise CEP126 localization relative to centrioles
Live-cell super-resolution:
Track dynamic movement of CEP126
Observe pericentriolar satellite transport in real-time
Correlate with microtubule dynamics
Correlative light and electron microscopy (CLEM):
Combine fluorescence with ultrastructural details
Link CEP126 localization to specific centrosomal structures
Examine cilium base architecture
Light-sheet microscopy:
Reduced phototoxicity for long-term imaging
Track CEP126 dynamics through entire cell cycle
Observe cilium formation in real-time
Expansion microscopy:
Physical expansion of specimens for improved resolution
Visualize protein arrangements within centrosome
Compatible with standard confocal microscopy
Advanced imaging could reveal how CEP126 participates in the transport of pericentriolar satellites and interacts with the dynein-dynactin complex, which is challenging to observe with conventional microscopy .