WDR73 Antibody, Biotin conjugated is a polyclonal antibody raised against a synthetic peptide corresponding to amino acids 44–73 of human WDR73. It is covalently linked to biotin, enabling its use in detection systems employing streptavidin-horseradish peroxidase (HRP) or streptavidin-fluorophore conjugates .
The antibody has been validated across multiple platforms, with specificity confirmed through knockdown and overexpression studies. For example:
Knockdown Validation: WDR73-depleted podocytes and fibroblasts showed loss of signal in Western blotting .
Overexpression Validation: HEK-293T cells expressing recombinant WDR73 exhibited strong immunoreactivity .
This antibody has been instrumental in studying WDR73’s role in Galloway-Mowat syndrome, a disorder characterized by microcephaly, nephrotic syndrome, and intellectual disability. Key findings include:
WDR73 localizes to spindle poles and astral microtubules during mitosis .
Loss of WDR73 disrupts microtubule networks and causes abnormal nuclear morphology in fibroblasts .
Sensitivity: Detects endogenous WDR73 in human tissues at concentrations as low as 0.1 µg/mL in ELISA .
Cross-reactivity: No reported cross-reactivity with other WD40-repeat proteins (e.g., WDR5, WDR62) .
| Application | Recommended Dilution | Buffer Conditions | Source |
|---|---|---|---|
| Western Blot | 1:500–1:1000 | Tris-HCl, 0.1% Tween-20 | |
| Immunofluorescence | 1:50 | PBS with 1% BSA | |
| Flow Cytometry | 1:100 | Permeabilization with 0.1% Triton |
WDR73 is a WD40-repeat-containing protein that plays a crucial role in regulating microtubule organization and dynamics. Its significance stems from its involvement in multiple cellular processes and its association with Galloway-Mowat syndrome (GAMOS), a rare disorder characterized by the co-occurrence of neurological defects and renal-glomerular disease. WDR73 is expressed in human cerebral cortex, hippocampus, and kidney cells, making it relevant for both neurological and renal research contexts . Loss-of-function mutations in WDR73 lead to defects in cell survival and microtubule organization, which explains the dual neurologic and renal phenotype observed in affected individuals .
Biotin-conjugated WDR73 antibodies have biotin molecules covalently attached to the antibody structure, which provides significant advantages for detection systems. This conjugation allows for interaction with streptavidin or avidin-coupled detection reagents, enabling signal amplification through the exceptionally strong biotin-avidin binding. Unlike unconjugated antibodies that require a secondary antibody for detection, biotin-conjugated antibodies can be directly detected using streptavidin-coupled reporters (fluorophores, enzymes, etc.), resulting in stronger signals, reduced background, and simplified experimental protocols. This is particularly valuable when studying proteins like WDR73 that may have relatively low expression levels in certain cell types .
When optimizing ELISA protocols with biotin-conjugated WDR73 antibodies, several parameters require careful adjustment. Start with a concentration titration (typically 0.1-10 μg/ml) to determine the optimal antibody concentration that provides the best signal-to-noise ratio. Use a high-quality streptavidin-HRP conjugate at an appropriate dilution (typically 1:1000-1:5000). Blocking solutions should contain BSA or casein rather than materials containing endogenous biotin (like milk). Include proper controls: a biotin-conjugated isotype control antibody and a known positive sample. For detecting endogenous WDR73, which has specific subcellular localization patterns, cell lysate preparation methodology is critical - ensure complete extraction of the cytosolic and nuclear fractions using appropriate buffers .
Sample preparation techniques vary based on tissue type due to WDR73's differential expression and subcellular localization. For brain tissue, where WDR73 is expressed in the cerebral cortex and hippocampus, fixation with 4% paraformaldehyde followed by permeabilization is recommended . For kidney samples, additional considerations include using specialized extraction buffers containing protease inhibitors to preserve WDR73's integrity. For cultured cells such as fibroblasts or podocytes, fixation with either cold 100% methanol or 4% paraformaldehyde is effective, with methanol being preferred when studying microtubule associations . For all tissue types, antigen retrieval may be necessary if using fixed tissues, and blocking with NH₄Cl after PFA fixation helps reduce background fluorescence .
Verifying antibody specificity is crucial for reliable results when working with WDR73. First, perform western blot analysis using recombinant WDR73 protein and lysates from tissues known to express WDR73 (brain, kidney) to confirm single band detection at the expected molecular weight (approximately 40-42 kDa). Compare staining patterns with published localization data showing WDR73's diffuse cytosolic pattern during interphase and spindle pole localization during mitosis . Implement knockout/knockdown controls by using WDR73-depleted cells generated via shRNA as negative controls . For biotin-conjugated antibodies specifically, include appropriate controls for endogenous biotin and potential streptavidin binding to ensure signals are specific to WDR73 rather than resulting from non-specific interactions.
To study WDR73's role in microtubule dynamics, biotin-conjugated WDR73 antibodies can be used in sophisticated co-localization experiments with microtubule markers. Implement dual immunofluorescence protocols using the biotin-conjugated WDR73 antibody detected with streptavidin-fluorophore conjugates alongside antibodies against α-, β-, and γ-tubulin, with which WDR73 is known to interact . For dynamic studies, design microtubule depolymerization and repolymerization assays using nocodazole treatment (30 μM for 4 hours) followed by washout and fixation at various time points (0, 5, and 60 minutes) . Quantify co-localization using advanced image analysis software and correlate WDR73 localization changes with microtubule network alterations. This approach allows visualization of WDR73's redistribution during microtubule reorganization and provides insights into its functional role in maintaining cytoskeletal integrity.
Investigating WDR73's interactions with Integrator complex components (particularly INTS9 and INTS11) requires specialized applications of biotin-conjugated antibodies . Design co-immunoprecipitation experiments using streptavidin beads to pull down biotin-conjugated WDR73 antibody complexes, followed by western blotting for INTS9 and INTS11. For in situ visualization, perform proximity ligation assays combining biotin-conjugated WDR73 antibodies with antibodies against Integrator components. Implement chromatin immunoprecipitation (ChIP) assays to investigate WDR73's role in transcriptional regulation alongside Integrator components, particularly following epidermal growth factor stimulation, which has been shown to trigger WDR73-dependent transcriptional responses . These approaches help elucidate WDR73's functional integration with RNA processing machinery and its impact on gene expression regulation.
For studying pathological mechanisms in Galloway-Mowat syndrome, biotin-conjugated WDR73 antibodies offer several sophisticated research applications. Compare WDR73 expression and localization patterns between normal tissues and patient-derived samples (if available) or genetically modified cellular models carrying WDR73 mutations (p.Phe296Leufs26 or p.Arg256Profs18) . Design experiments to analyze downstream effects of WDR73 dysfunction on UsnRNA processing and gene expression patterns of cell cycle regulatory proteins, which are altered upon WDR73 suppression . Implement rescue experiments in WDR73-depleted podocytes to evaluate functional recovery and identify critical domains for WDR73 function . These approaches provide insights into the molecular mechanisms underlying the nephrocerebellar pathology in Galloway-Mowat syndrome and potential therapeutic targets.
Common issues with biotin-conjugated WDR73 antibodies include high background signals, weak specific signals, and inconsistent results. High background often results from endogenous biotin in samples; mitigate this by using avidin/biotin blocking kits before applying the antibody and avoiding biotin-containing blocking reagents like milk. Weak signals may result from suboptimal antibody concentration or inadequate sample preparation; optimize by testing concentration ranges and ensuring proper extraction of WDR73 from cellular compartments, particularly during mitosis when it relocates to spindle poles . Inconsistent results might stem from WDR73's cell-cycle-dependent localization; synchronize cells when possible or carefully document cell cycle stages during analysis. Additionally, WDR73's interaction with heat shock proteins (HSP-70 and HSP-90) may affect epitope accessibility; consider mild denaturation or epitope retrieval methods to improve detection .
Validating WDR73 antibody results across different cell types requires a systematic approach due to potential variations in WDR73 expression and function. Implement parallel experiments in multiple relevant cell types, particularly podocytes and neuronal cells, which are affected in Galloway-Mowat syndrome . For each cell type, confirm antibody specificity using siRNA/shRNA knockdown controls to demonstrate signal reduction proportional to WDR73 depletion . Compare results with published WDR73 localization patterns specific to each cell type and cell cycle stage. Employ multiple detection methods (e.g., western blot, immunofluorescence, and ELISA) to cross-validate findings. If exploring novel cell types, first establish baseline WDR73 expression levels via qPCR before antibody-based studies. This multi-faceted validation approach ensures reliable cross-cell type comparisons and minimizes cell-type-specific artifacts.
When evaluating biotin-conjugated WDR73 antibody performance, several quality control metrics should be systematically assessed. First, determine antibody specificity through western blotting against recombinant WDR73 and cellular lysates, looking for a single band at the expected molecular weight. Calculate signal-to-noise ratios in immunostaining applications, with values >10:1 indicating good performance. Assess batch-to-batch consistency by comparing staining patterns and signal intensities across multiple antibody lots. Evaluate epitope accessibility by comparing results across different fixation and permeabilization protocols. Test for potential cross-reactivity with other WD40 domain-containing proteins through competitive binding assays. For biotin conjugation specifically, determine the biotin-to-antibody ratio (typically 3-5 biotin molecules per antibody is optimal) and verify that conjugation hasn't compromised antigen recognition. Document antibody performance characteristics for reproducibility and reliable interpretation of experimental outcomes.
WDR73 antibodies can be instrumental in investigating UsnRNA processing pathways through several sophisticated approaches. Design RNA immunoprecipitation (RIP) assays using biotin-conjugated WDR73 antibodies to pull down WDR73-associated RNA complexes, followed by RT-PCR or sequencing to identify bound UsnRNAs. Implement CLIP-seq (cross-linking immunoprecipitation followed by sequencing) to map WDR73 binding sites on RNA with nucleotide resolution. Use the antibodies in combination with pulse-chase labeling of newly synthesized RNA to track WDR73's involvement in UsnRNA maturation kinetics. For functional studies, compare UsnRNA processing efficiency between normal and WDR73-depleted cells using northern blotting or RT-PCR with primers specific for precursor and mature UsnRNA forms . These approaches collectively illuminate WDR73's mechanistic contribution to RNA metabolism, which is dysregulated in conditions like Galloway-Mowat syndrome.
For studying WDR73's role in cell cycle regulation, several specialized experimental designs using WDR73 antibodies are appropriate. Implement cell synchronization protocols followed by immunofluorescence at different cell cycle stages to track WDR73's dynamic relocalization from cytoplasm during interphase to spindle poles during mitosis . Design live-cell imaging experiments using cell-permeable fluorescent tags coupled to Fab fragments of WDR73 antibodies to monitor real-time dynamics during cell division. Perform chromatin immunoprecipitation followed by sequencing (ChIP-seq) to identify cell cycle-regulated genes under WDR73 influence, particularly those encoding cell cycle regulatory proteins whose expression is altered upon WDR73 suppression . Compare cell cycle progression parameters (using flow cytometry with propidium iodide staining) between normal and WDR73-depleted cells to quantify cell cycle abnormalities. These approaches collectively elucidate WDR73's multifaceted roles in maintaining proper cell cycle progression.
To explore WDR73's interactions with heat shock proteins (HSP-70 and HSP-90) and cellular stress responses, researchers can implement several advanced approaches using biotin-conjugated WDR73 antibodies. Design co-immunoprecipitation experiments under different cellular stress conditions (heat shock, oxidative stress, ER stress) to assess dynamic changes in WDR73-HSP interactions. Perform proximity ligation assays to visualize and quantify these interactions in situ at single-molecule resolution. Implement FRET (Förster Resonance Energy Transfer) analysis using labeled antibodies to measure interaction distances and binding dynamics. Compare stress response gene expression profiles between wildtype and WDR73-mutant cells (particularly those carrying p.Phe296Leufs26 or p.Arg256Profs18 mutations, which show increased interaction with HSP-70/HSP-90) . These methods help elucidate how WDR73-HSP interactions influence protein folding, stability, and cellular adaptation to stress, potentially explaining the cellular vulnerability observed in Galloway-Mowat syndrome.