WDR73 is a WD40-repeat domain protein that plays crucial roles in microtubule functions and spindle poles during mitotic cell division. Its significance stems from its association with Galloway-Mowat syndrome (GAMOS), a rare genetic disorder characterized by the co-occurrence of neurological symptoms (including microcephaly, cerebral atrophy, and neural migration defects) and glomerular-renal disease. WDR73 mutations have been identified as causative factors in this syndrome, making it an important target for understanding disease mechanisms in both neurological and renal contexts .
The FITC-conjugated WDR73 antibody is primarily utilized for:
Flow cytometry (FACS) for analyzing WDR73 expression in cell populations
Immunofluorescence (IF) for visualizing subcellular localization
ELISA assays for quantitative protein detection
Fluorescence-based Western blotting
This conjugated antibody eliminates the need for secondary antibody incubation in fluorescence-based applications, particularly beneficial for multicolor immunofluorescence studies .
Most commercially available WDR73 antibodies, including FITC-conjugated versions, demonstrate specificity for human WDR73. According to product specifications, these antibodies typically recognize epitopes in the N-terminal region, particularly amino acids 44-73, and are validated through Western blotting against human samples. Specificity is typically confirmed through overexpression studies and shRNA knockdown validation .
| Antibody Type | Epitope Region | Host | Validated Applications | Reactivity |
|---|---|---|---|---|
| Polyclonal | N-terminal (AA 44-73) | Rabbit | WB, IHC, IF, FACS, ELISA | Human |
| Monoclonal | Various epitopes | Mouse/Rabbit | WB, IHC, IF, FACS | Human |
For optimal immunofluorescence results with FITC-conjugated WDR73 antibody:
Fixation method selection: Both paraformaldehyde (PFA) and methanol fixation protocols have been successfully utilized, but their effectiveness varies depending on cell type and subcellular localization interests.
For cytoskeletal association studies: 4% paraformaldehyde (20 minutes at room temperature) preserves cytoskeletal structures better
For nuclear/chromatin studies: Ice-cold 100% methanol (5 minutes) provides superior nuclear antigen accessibility
Permeabilization optimization: After PFA fixation, use 0.5% Triton X-100 in PBS for 15 minutes to ensure adequate antibody penetration, particularly for nuclear antigens
Blocking parameters: 5% bovine serum albumin in PBS for 1 hour at room temperature effectively minimizes non-specific binding
WDR73 antibody dilution range: Optimal dilutions typically fall between 1:50-1:200 for immunofluorescence applications .
A robust validation protocol requires multiple controls:
Negative controls:
WDR73 knockout/knockdown cells (CRISPR/Cas9 or shRNA approaches)
Isotype control antibodies (rabbit IgG-FITC conjugated)
Primary antibody omission control
Positive controls:
Cells overexpressing recombinant WDR73 (documented in validation studies)
Tissues with known WDR73 expression (cerebral cortex, hippocampus, kidney glomeruli)
Specificity validation:
For optimal flow cytometry results:
Sample preparation:
For cell suspensions: 1×10⁶ cells per sample
Fixation: 4% paraformaldehyde (10-15 minutes, room temperature)
Permeabilization: If needed for intracellular staining, use 0.1% saponin or commercial permeabilization buffer
Antibody incubation:
Dilution range: 1:10-1:50 (higher concentration than for IF)
Incubation time: 30-60 minutes at 4°C in the dark
Washing: 3× with PBS containing 1% FBS
Controls and gating strategy:
Unstained cells for autofluorescence baseline
Isotype-FITC control for non-specific binding
Begin gating on leukocytes when working with blood samples
Additional surface markers may be required to identify specific cell populations
Storage precautions: Do not freeze FITC-conjugated antibodies; store at 4°C protected from light .
WDR73 has been shown to interact with INTS9 and INTS11 components of the Integrator complex. To investigate these interactions:
Co-localization studies:
Use multi-color immunofluorescence with FITC-conjugated WDR73 antibody and differently conjugated antibodies against INTS9, INTS11, or other Integrator components
Analyze nuclear co-localization using confocal microscopy with Z-stack acquisition
Quantify co-localization using Pearson's or Mander's correlation coefficients
Cell synchronization strategies:
Since WDR73-Integrator interactions may be cell cycle-dependent, synchronize cells using double-thymidine block protocols
Analyze different cell cycle phases separately
Proximity ligation assay (PLA) adaptation:
To study WDR73's microtubule regulatory functions:
Live-cell imaging:
Microinjection of FITC-conjugated WDR73 antibody into live cells
Time-lapse microscopy during mitosis to track spindle pole localization
Co-localization with tubulin:
Dual immunofluorescence with FITC-WDR73 antibody and α/β/γ-tubulin antibodies
Quantitative analysis of co-localization at mitotic structures
Functional perturbation studies:
Combine with tubulin-targeting drugs (nocodazole, taxol) to disrupt microtubule dynamics
Analyze changes in WDR73 localization patterns
Correlate with cell cycle progression abnormalities
WDR73 mutant expression effects:
Recent research has implicated WDR73 in focal adhesion regulation, particularly in podocytes. To investigate this:
Podocyte-specific staining protocols:
For immortalized podocyte cell lines: Use FITC-WDR73 antibody (1:50-1:100) with phalloidin counterstaining to visualize actin filaments
For kidney tissue sections: Combine with podocyte markers (synaptopodin, nephrin) to identify podocytes specifically
PIP4K2C-WDR73 interaction analysis:
Dual staining with FITC-WDR73 antibody and PIP4K2C antibodies
Analyze effects of WDR73 depletion on PIP4K2C expression and stability
Quantify PIP2 levels in relation to WDR73 expression
Focal adhesion quantification:
Several technical challenges may arise:
Photobleaching:
Problem: FITC is relatively susceptible to photobleaching during prolonged imaging
Solution: Use anti-fade mounting media containing DABCO or propyl gallate; minimize exposure during image acquisition; consider using alternative more photostable fluorophores for long-term imaging studies
Background fluorescence:
Problem: High autofluorescence in certain tissues (especially kidney) in the FITC channel
Solution: Implement additional blocking steps with normal serum (5-10%); use Sudan Black B (0.1-0.3%) post-staining to reduce autofluorescence; consider spectral unmixing during image acquisition
pH sensitivity:
Problem: FITC fluorescence is pH-dependent and decreases in acidic environments
Solution: Maintain buffers at pH 7.2-8.0; avoid acidic fixatives; use pH-stable mountants
Signal amplification challenges:
When facing discrepancies in WDR73 localization patterns:
Epitope mapping analysis:
Different antibodies may recognize distinct epitopes, potentially masked in certain conformations or protein complexes
Compare antibodies targeting N-terminal versus C-terminal regions
Validation hierarchy:
Prioritize localization data obtained from antibodies validated by multiple methods (knockout controls, overexpression, peptide competition)
Consider complementary approaches (GFP-tagged WDR73 expression) to confirm localization
Cell-type and context dependency:
WDR73 localization may genuinely differ between cell types and physiological states
FITC-conjugated WDR73 antibody has shown both nuclear and cytoplasmic distribution, which aligns with its multiple reported functions
Standardize experimental conditions including cell cycle stage, confluency, and fixation protocols .
To ensure detection specificity:
Signal pattern analysis:
Specific WDR73 staining should show:
Enrichment at mitotic spindles during cell division
Nuclear and/or cytoplasmic localization patterns consistent with published data
Co-localization with known interaction partners (tubulin, INTS9/11)
Non-specific patterns often appear as:
Uniform cytoplasmic staining without subcellular enrichment
Excessive membrane or nucleolar signals
Persistence in knockout/knockdown controls
Quantitative validation approaches:
Establish signal-to-noise ratios across multiple experiments
Implement intensity thresholding based on negative controls
Perform peptide competition assays at increasing peptide concentrations
Multi-method confirmation:
Current research applications include:
Patient-derived cell analysis:
FITC-WDR73 antibody enables visualization of mutant protein localization and abundance in patient fibroblasts and induced pluripotent stem cell (iPSC)-derived podocytes and neurons
Flow cytometric quantification of expression levels across different patient mutations
Animal model validation:
Immunofluorescence analysis of conditional Wdr73 knockout mouse tissues
Correlation of WDR73 expression patterns with podocyte foot process injury and albuminuria development
Mechanistic pathway investigations:
Innovative methodological approaches include:
Super-resolution microscopy applications:
STED and STORM microscopy for nanoscale localization of WDR73 at microtubule structures
Single-molecule tracking of WDR73 dynamics during cell division
Multi-omics integration strategies:
Fluorescence-activated cell sorting (FACS) with FITC-WDR73 antibody followed by transcriptomics/proteomics
Combining with proximity labeling approaches (BioID, APEX) to identify context-specific interaction partners
Functional screening platforms:
WDR73 research is illuminating:
Integrator complex regulation:
WDR73's interaction with INTS9/11 suggests it functions in UsnRNA processing
FITC-WDR73 antibody enables visualization of dynamic interactions with Integrator components during transcriptional responses
Post-mitotic cell maintenance mechanisms:
Both podocytes and neurons are terminally differentiated cells affected by WDR73 mutations
WDR73 studies reveal common pathways critical for maintaining these specialized cell types
Microtubule-associated protein networks:
WDR73 interacts with α-, β-, and γ-tubulin, HSP-70, HSP-90, and other proteins
FITC-conjugated antibody allows visualization of these interaction networks in relevant cellular contexts
Phosphoinositide signaling in cell adhesion: