FBXO30 antibodies are polyclonal or monoclonal reagents optimized for detecting FBXO30 in experimental workflows such as Western blot (WB), immunocytochemistry (ICC), and ELISA. These antibodies enable researchers to investigate FBXO30's roles in:
FBXO30 is a tumor suppressor and E3 ligase with critical regulatory roles:
| Parameter | Detail |
|---|---|
| Reactivity | Human |
| Applications | WB (1:2,000–1:10,000), ELISA |
| Observed MW | 90 kDa (vs. predicted 82 kDa) |
| Validated Samples | HeLa cells, A549 cells |
| Storage | -20°C in PBS with 50% glycerol |
| Immunogen | FBXO30 fusion protein (Ag27791) |
| Parameter | Detail |
|---|---|
| Reactivity | Human |
| Applications | ICC/IF (1–4 µg/mL) |
| Immunogen | Recombinant peptide (AA sequence provided) |
| Specificity | Validated via protein array with 384 non-target controls |
| Parameter | Detail |
|---|---|
| Reactivity | Human |
| Applications | WB (1:500 dilution) |
| Observed Bands | 37, 45, 60, 77, 82, 86 kDa (suggests splice variants or degradation) |
| Blocking Buffer | PBS with 0.02% sodium azide |
ccRCC Studies: FBXO30 expression is significantly reduced in ccRCC tissues compared to normal adjacent tissues . Low FBXO30 correlates with poor survival (TCGA data) .
Mechanistic Insights: FBXO30 degrades HIF-1α under normoxia, disrupting hypoxia-driven oncogenesis .
Eg5 Interaction: FBXO30 targets Eg5 for ubiquitination, ensuring mitotic spindle integrity. Fbxo30 knockout mice exhibit defective mammopoiesis and centrosome abnormalities .
RARγ Degradation: FBXO30-mediated RARγ ubiquitination modulates BMP signaling, critical for neural progenitor cell differentiation .
Cross-Reactivity: Most antibodies are validated for human samples only .
Band Discrepancies: Observed molecular weights (e.g., 90 kDa for Proteintech’s antibody) may differ from predicted values due to post-translational modifications .
Buffer Compatibility: Glycerol-based storage buffers (e.g., 40–50%) ensure long-term stability .
FBXO30’s role in degrading oncoproteins (e.g., HIF-1α) positions it as a potential therapeutic target. Antibodies against FBXO30 are instrumental in:
For effective immunostaining of FBXO30 in tissue sections, a multistep approach is recommended. Begin with deparaffinization and rehydration using xylene and ethanol gradients. Antigen retrieval is crucial and should be performed using 10 mM sodium citrate buffer (pH 6.0) at 95-100°C for 20-30 minutes. Permeabilize sections with 0.3% Triton X-100 in 10 mM Tris-HCl buffer for 30 minutes, followed by blocking with 2% bovine serum albumin (BSA) for 60 minutes. Incubate with primary FBXO30 antibody diluted in 10 mM Tris-HCl buffer containing 2% BSA at 4°C overnight, followed by appropriate secondary antibody incubation for 2-4 hours at room temperature. Counterstain nuclei with DAPI and mount using an anti-fade mounting medium .
Antibody validation requires a multi-pronged approach. First, perform Western blotting using both recombinant FBXO30 protein and tissue/cell lysates known to express FBXO30, checking for a single band at the expected molecular weight (~30-35 kDa). Second, include a positive control (cells overexpressing FBXO30) and a negative control (FBXO30 knockout tissues or cells with FBXO30 silenced by shRNA). Third, perform immunoprecipitation followed by mass spectrometry to confirm that the antibody pulls down authentic FBXO30. Fourth, use immunofluorescence with blocking peptides to demonstrate specificity. Finally, parallel testing with at least two distinct antibodies targeting different epitopes of FBXO30 should yield similar results. Analysis of FBXO30 knockout mice tissues, which should show absence of staining, provides the gold standard for antibody validation .
When investigating FBXO30-substrate interactions, several critical controls are necessary. First, include negative controls using structurally similar F-box proteins (such as other FBXO family members) to confirm specificity. Second, perform reciprocal co-immunoprecipitation experiments (pull down with anti-FBXO30 and probe for the substrate, then pull down with anti-substrate and probe for FBXO30) as demonstrated with FBXO30 and EG5 interaction. Third, include proteasome inhibitor controls (e.g., MG132) to stabilize transient interactions. Fourth, utilize truncation mutants of both FBXO30 and the substrate to map interaction domains, as shown with the C-terminal region of EG5 (AA812-1052) being the interaction site with FBXO30. Fifth, confirm the functional consequences of the interaction through ubiquitination assays with recombinant E1, E2, FBXO30, substrate, and ubiquitin, followed by detection with anti-polyubiquitin antibodies .
For effective subcellular localization of FBXO30, a differential centrifugation approach is recommended. Begin with gentle cell lysis using a hypotonic buffer containing protease inhibitors. Centrifuge at 1,000g for 10 minutes to separate nuclei (pellet). Further centrifuge the supernatant at 10,000g for 15 minutes to pellet mitochondria, followed by 100,000g for 1 hour to separate microsomes (pellet) from cytosol (supernatant). For more detailed fractionation, use density gradient centrifugation with sucrose or Percoll. Confirm fraction purity using markers: lamin A/C for nuclei, cytochrome c for mitochondria, calnexin for ER, and GAPDH for cytosol. For FBXO30 specifically, analyze fractions by Western blotting, with particular attention to nuclear and cytoplasmic fractions, as F-box proteins often shuttle between these compartments depending on cell cycle stage and substrate availability .
Distinguishing between FBXO30's distinct ubiquitination targets requires carefully designed experiments that isolate each pathway. For RARγ pathway analysis, use cell systems where EG5 is either silenced or mutated at FBXO30 binding sites (C-terminal region AA812-1052). Conversely, for EG5 pathway studies, use systems where RARγ is inactivated. Temporal analysis is crucial, as FBXO30-RARγ interaction predominates during neural development, while FBXO30-EG5 interaction is critical during cell cycle progression and mammopoiesis.
Create the following experimental matrix to isolate each pathway:
| Experimental Condition | FBXO30 Status | Pathway Focus | Key Readouts |
|---|---|---|---|
| RA treatment + RARγ present, EG5 silenced | Wild-type | RARγ pathway | BMP signaling markers |
| No RA + RARγ silenced, EG5 present | Wild-type | EG5 pathway | Mitotic spindle formation |
| RA treatment + both present | Wild-type | Dual pathway | Competition analysis |
| RA treatment + both present | Knockout/inhibited | Rescue experiments | Pathway-specific phenotypes |
For biochemical distinction, use in vitro ubiquitination assays with site-specific mutants. The E3 ligase activity of FBXO30 toward RARγ results in suppression of BMP signaling, while its activity toward EG5 affects centrosome homeostasis and mitotic spindle formation. These distinct downstream effects serve as reliable readouts for pathway-specific activity .
Analysis of FBXO30 expression in neural tube defect (NTD) tissues requires specialized methodological approaches. Fresh tissue procurement is critical, with rapid fixation (within 30 minutes post-collection) to preserve protein integrity. For human NTD samples, ethical considerations and appropriate consent protocols must be established. Parallel analysis of retinol levels is essential, as high retinol conditions affect FBXO30 levels.
For comprehensive analysis, implement this multi-modal approach:
Quantitative immunohistochemistry using validated FBXO30 antibodies with automated image analysis to ensure unbiased quantification
Laser capture microdissection to isolate specific cell populations from the neural tube for protein and RNA extraction
Western blotting with gradient gels to detect both full-length FBXO30 and potential isoforms or degradation products
qRT-PCR with multiple reference genes validated for stability in neural tissues
Analysis of BMP target gene expression (downstream readout of FBXO30-RARγ pathway)
When comparing NTD and control tissues, account for variables including gestational age, sample preservation time, and retinol levels. A scoring system incorporating both FBXO30 levels and downstream targets (BMP pathway genes) provides the most reliable assessment of FBXO30 functional status in NTD tissues .
To investigate FBXO30's role in mitotic spindle defects in cancer cells, a comprehensive experimental design should include:
First, establish baseline FBXO30 and EG5 expression profiles across cancer cell lines using Western blotting, qRT-PCR, and immunofluorescence. Next, implement a CRISPR-Cas9 approach to generate FBXO30 knockout cancer cell lines alongside rescue lines expressing either wild-type FBXO30 or ligase-dead mutants.
For functional analysis, employ live-cell imaging with fluorescently tagged tubulin to track mitotic spindle dynamics in real-time. Quantify multipolar spindle formation, spindle pole fragmentation, and chromosome segregation errors. Perform immunofluorescence co-staining for FBXO30, EG5, and γ-tubulin to analyze centrosome amplification and spindle morphology.
To establish causality, manipulate EG5 levels through siRNA knockdown or EG5 inhibitors (such as monastrol) in both FBXO30-normal and FBXO30-deficient cells. If abnormal spindle phenotypes in FBXO30-deficient cells are normalized through EG5 reduction, this confirms FBXO30's regulatory role through EG5. Additionally, perform cell cycle synchronization experiments using double thymidine block to analyze FBXO30 and EG5 oscillation patterns during mitosis, with specific attention to G2/M transition where aberrant EG5 activity leads to multipolar spindles and genomic instability .
For mammopoiesis studies, analyze:
Mammary epithelial cell populations using flow cytometry with markers CD24, CD29, CD49f, and Sca1
Mammosphere formation assays to assess stem cell function
Immunohistochemistry for lineage markers (Keratin 5 for myoepithelial cells)
Mitotic spindle analysis in mammary epithelial cells using β-tubulin and DAPI staining
Quantification of EG5 protein levels and cell cycle distribution
For neural development studies, focus on:
BMP signaling pathway components and their expression levels
RARγ degradation kinetics in neural progenitor cells
Neural tube closure in embryonic development
Retinol and retinoic acid level measurements
Neural progenitor cell proliferation and differentiation markers
Cross-tissue analysis should include ubiquitination assays comparing FBXO30's preferential substrates (EG5 versus RARγ) in each tissue type, and rescue experiments using tissue-specific expression of either substrate to determine if the phenotypes are reversible .
Developing phospho-specific antibodies for FBXO30 regulation studies requires careful consideration of multiple technical factors. First, perform in silico analysis using phosphorylation prediction algorithms (NetPhos, GPS, PhosphoSitePlus) to identify potential phosphorylation sites on FBXO30. Validate these sites using mass spectrometry following immunoprecipitation of FBXO30 from cells treated with phosphatase inhibitors.
For antibody development, synthetic phosphopeptides should span 10-15 amino acids centered on the phosphorylation site, with the following considerations:
Include a terminal cysteine for carrier protein conjugation if not naturally present
Ensure peptide solubility and minimize secondary structure formation
Create paired phosphorylated and non-phosphorylated peptides for screening
Consider multiple host species to maximize application versatility
Antibody validation should include:
ELISA testing against phosphorylated versus non-phosphorylated peptides
Western blotting with lysates from cells treated with kinase activators/inhibitors
Competition assays with free phosphopeptides
Testing on FBXO30 knockout samples (negative control)
Testing on samples treated with lambda phosphatase
For understanding FBXO30 regulation, particularly important is examining how phosphorylation affects FBXO30-substrate interactions. Unlike EG5, where phosphorylation at T927 or S1040 doesn't affect FBXO30-mediated ubiquitination, FBXO30's own phosphorylation state may determine substrate specificity between RARγ and EG5 in different cellular contexts .
Cell synchronization experiments for studying FBXO30 dynamics require precise timing and multiple synchronization methods for comprehensive analysis. Implement a double thymidine block protocol (2 mM thymidine for 18 hours, release for 9 hours, second thymidine treatment for 17 hours) to arrest cells at G1/S boundary. After release, collect samples every 2 hours for 24 hours to cover a complete cell cycle.
Alternative synchronization approaches include:
Nocodazole treatment (100 ng/mL for 12-16 hours) for M-phase arrest
Serum starvation (0.1% serum for 48 hours) for G0/G1 arrest
RO-3306 (10 μM for 18 hours) for G2/M arrest
For each time point, analyze:
FBXO30 protein levels by Western blotting
FBXO30 subcellular localization by immunofluorescence
FBXO30-substrate interactions by co-immunoprecipitation
Ubiquitination status of FBXO30 targets
Cell cycle distribution by flow cytometry with propidium iodide staining
Critical controls include asynchronous cell populations and validation of synchronization efficiency using established cell cycle markers (cyclin B1, phospho-histone H3). Based on findings from EG5 studies, pay particular attention to G2/M transition, where FBXO30-mediated regulation appears most critical for normal mitotic spindle formation and chromosome segregation .
Detecting FBXO30-mediated ubiquitination requires different approaches for in vitro and in vivo scenarios. For in vitro ubiquitination:
Reconstitute the complete ubiquitination cascade using purified components:
Recombinant E1 ubiquitin-activating enzyme
E2 ubiquitin-conjugating enzyme
FBXO30 immunoprecipitated from mammalian cells or recombinant
Purified substrate (RARγ or EG5)
Ubiquitin (consider using tagged versions for easier detection)
ATP and buffer components
Incubate the reaction at 30°C for 1-2 hours and analyze by SDS-PAGE followed by immunoblotting with anti-ubiquitin antibodies.
For in vivo ubiquitination:
Treat cells with proteasome inhibitors (MG132, 10 μM for 4-6 hours) to prevent degradation of ubiquitinated proteins
Perform denaturing immunoprecipitation to disrupt protein-protein interactions:
Lyse cells in buffer containing 1% SDS
Dilute lysate to 0.1% SDS before immunoprecipitation
Immunoprecipitate the substrate of interest
Analyze by Western blotting with anti-ubiquitin antibodies
Alternative approach using His-tagged ubiquitin:
Transfect cells with His-ubiquitin
Lyse cells under denaturing conditions
Purify ubiquitinated proteins using Ni-NTA resin
Detect specific substrates by immunoblotting
Controls should include FBXO30 knockout cells, substrate mutants lacking FBXO30 binding sites, and F-box domain mutants of FBXO30 that cannot form an active SCF complex .
To correlate FBXO30 expression with clinical outcomes in developmental disorders, researchers should implement a multi-layer analysis approach. Begin with tissue microarray construction from well-characterized patient cohorts with detailed clinical data and long-term follow-up. Perform immunohistochemistry with validated FBXO30 antibodies, using automated quantitative analysis (AQUA) to generate objective expression scores.
For neural tube defects specifically, establish a comprehensive assessment matrix:
| Parameter | Techniques | Clinical Correlation |
|---|---|---|
| FBXO30 protein levels | IHC, Western blot | Defect severity, long-term neurological outcomes |
| FBXO30 mRNA expression | qRT-PCR, RNA-seq | Genetic predisposition analysis |
| BMP pathway activity | Target gene expression panel | Developmental pathway dysregulation |
| Retinol/RA levels | HPLC analysis of serum | Maternal nutrition status |
| RARγ degradation | Western blot, half-life analysis | Pathway functionality |
| Genetic variants | Targeted sequencing of FBXO30 | Heritable risk factors |
Statistical analysis should include multivariate models adjusting for confounding factors such as gestational age, maternal nutrition, and genetic background. Longitudinal studies with consistent follow-up intervals are essential for correlating early FBXO30 expression patterns with long-term developmental outcomes. Case-control studies comparing NTD samples with matched controls allow for identification of FBXO30 expression thresholds that predict adverse outcomes .
A comprehensive workflow for analyzing FBXO30 knockout phenotypes across model systems should follow a systematic approach from molecular to organismal levels. Begin with validation of knockout efficiency through genomic PCR, Western blotting, and immunostaining to confirm absence of FBXO30 protein.
For cellular phenotyping:
Analyze cell cycle distribution using flow cytometry with propidium iodide staining
Examine mitotic spindle formation through immunofluorescence for β-tubulin
Quantify centrosome numbers using γ-tubulin staining
Assess genomic stability through karyotyping and micronuclei formation analysis
Measure proliferation rates using BrdU incorporation assays
For tissue-specific analysis:
In mammary tissue:
Flow cytometry for epithelial cell populations (CD24+CD29lo luminal and CD24+CD29hi myoepithelial)
Mammosphere formation assays
Whole-mount analysis of ductal development
Immunohistochemistry for lineage markers
In neural tissue:
Neural tube closure assessment in embryos
BMP signaling target gene expression
Neuroprogenitor proliferation and differentiation markers
For rescue experiments, introduce either wild-type FBXO30 or target substrate modulators (EG5 inhibitors or shRNA) to determine phenotype reversibility. Establish clear quantitative metrics for each phenotype to enable statistical comparison across experiments and between different model systems .
Successful multiplex immunofluorescence studies involving FBXO30 and its target proteins require careful antibody selection and validation. Begin with comprehensive antibody screening against single targets in positive control tissues before attempting multiplexing. Consider these key factors:
Host species compatibility:
Select primary antibodies raised in different host species (e.g., rabbit anti-FBXO30, mouse anti-EG5, rat anti-RARγ)
If using same-species antibodies, employ sequential staining with complete HRP inactivation between rounds
Antibody format selection:
For direct detection: Pre-conjugated primary antibodies with non-overlapping fluorophores
For indirect detection: Carefully matched secondary antibodies with minimal cross-reactivity
Signal amplification considerations:
For low abundance proteins: Tyramide signal amplification (TSA)
For co-localization studies: Standard indirect immunofluorescence to minimize spatial distortion
Multiplexing panel design:
| Target | Abundance | Recommended Approach | Signal Separation |
|---|---|---|---|
| FBXO30 | Low-moderate | Rabbit mAb + TSA | 488 nm (green) |
| EG5/KIF11 | Moderate | Mouse mAb + standard IF | 555 nm (red) |
| RARγ | Low | Goat pAb + TSA | 647 nm (far red) |
| Cellular context marker | Variable | Directly conjugated | 405 nm (blue) |
Validation controls:
Single staining controls for each antibody
Fluorescence minus one (FMO) controls
Peptide competition controls
FBXO30 knockout tissues as negative control
For optimal visualization of protein interactions, complement standard confocal microscopy with super-resolution techniques such as structured illumination microscopy (SIM) or stochastic optical reconstruction microscopy (STORM) for detailed co-localization analysis at the subcellular level .
Several cutting-edge technologies are emerging for real-time analysis of FBXO30-substrate interactions in living cells. Proximity ligation assays (PLA) offer visualization of protein interactions within 40 nm distance, providing spatial information about FBXO30-substrate complexes. For real-time dynamics, bimolecular fluorescence complementation (BiFC) can be employed, where split fluorescent proteins fused to FBXO30 and its substrate reconstitute fluorescence upon interaction.
Förster resonance energy transfer (FRET) approaches using FBXO30 and substrates tagged with compatible fluorophores (e.g., CFP-FBXO30 and YFP-EG5) enable quantitative assessment of protein-protein interactions with high temporal resolution during cell cycle progression. Lattice light-sheet microscopy combined with these fluorescent reporters offers superior spatial and temporal resolution with minimal phototoxicity for extended imaging.
For substrate fate tracking, fluorescent timer proteins fused to FBXO30 targets provide color-coded information about protein age and turnover rates. CRISPR-based gene tagging with split fluorescent proteins enables visualization of endogenous protein interactions without overexpression artifacts.
Correlative light and electron microscopy (CLEM) can bridge the resolution gap between fluorescence microscopy and ultrastructural analysis, particularly valuable for studying FBXO30's role in mitotic spindle formation and centrosome dynamics. Mass spectrometry imaging (MSI) is emerging as a label-free approach to map protein modifications, including ubiquitination patterns, across cellular compartments .
Computational approaches offer powerful tools for unraveling FBXO30 substrate recognition mechanisms. Structural modeling using homology-based techniques can predict the three-dimensional conformation of FBXO30's substrate-binding domain based on known F-box protein structures. Molecular docking simulations can then identify potential interaction interfaces between FBXO30 and substrates like RARγ and EG5.
Machine learning algorithms trained on known E3 ligase-substrate pairs can predict additional FBXO30 substrates by analyzing protein features such as degron motifs, disorder regions, and post-translational modification sites. Network analysis integrating protein-protein interaction data, co-expression patterns, and evolutionary conservation can reveal substrate candidates within FBXO30's functional pathways.
Molecular dynamics simulations offer insights into the conformational changes of FBXO30 upon substrate binding and how these changes affect ubiquitin transfer. These simulations can also elucidate how phosphorylation of either FBXO30 or its substrates alters binding affinity and specificity.
Text mining approaches can identify potential FBXO30-substrate relationships from the scientific literature that might have been overlooked. Combined with systems biology approaches modeling ubiquitin-proteasome dynamics, these computational methods provide testable hypotheses about FBXO30's role in maintaining proteostasis across different tissue contexts and developmental stages .
Generating reliable FBXO30 knockout and knockdown models requires careful selection of appropriate techniques based on experimental goals. For complete knockout models, CRISPR-Cas9 genome editing offers the most definitive approach. Design sgRNAs targeting early exons of FBXO30, preferably exon 1, to ensure complete protein elimination. Use multiple guide RNAs to increase editing efficiency, and screen clones by genomic PCR, Sanger sequencing, and Western blotting.
For conditional knockout models critical for tissue-specific studies:
Generate FBXO30-floxed mice with loxP sites flanking critical exons
Cross with tissue-specific Cre lines (e.g., MMTV-Cre for mammary, Nestin-Cre for neural)
Validate recombination efficiency by tissue-specific genomic PCR and immunoblotting
For transient knockdown applications:
siRNA approach: Design 3-4 siRNAs targeting different regions of FBXO30 mRNA
shRNA approach: Use inducible systems (Tet-On/Off) for temporal control
Validation criteria: >80% reduction in FBXO30 protein levels within 48-72 hours
Critical controls: Non-targeting siRNA/shRNA and rescue with siRNA-resistant FBXO30
Regardless of the approach, comprehensive validation is essential:
Genomic verification: PCR and sequencing of targeted region
Transcript analysis: RT-PCR and RNA-seq to confirm absence of FBXO30 mRNA
Protein validation: Western blotting and immunostaining with antibodies targeting different epitopes
Functional validation: Analysis of substrate accumulation (RARγ, EG5)
Phenotypic confirmation: Assessment of cellular processes known to be regulated by FBXO30 (mitotic spindle formation, BMP signaling) .
Implementing robust quality control measures for commercial FBXO30 antibodies is essential for reliable experimental outcomes. For any new antibody lot, perform Western blotting validation using positive controls (tissues known to express FBXO30 such as mammary epithelial cells) and negative controls (FBXO30 knockout tissues or cells with FBXO30 knocked down by siRNA/shRNA). Verify the antibody detects a single band of appropriate molecular weight (~30-35 kDa).
Application-specific quality control measures:
For immunohistochemistry/immunofluorescence:
Test multiple antibody dilutions to determine optimal signal-to-noise ratio
Include absorption controls using blocking peptides
Compare staining patterns across multiple FBXO30 antibodies targeting different epitopes
Include FBXO30 knockout tissues as negative controls
Validate subcellular localization against known patterns
For immunoprecipitation:
Verify pull-down efficiency by comparing input, unbound, and immunoprecipitated fractions
Confirm co-immunoprecipitation of known interaction partners (Skp1, Cul1)
Perform reverse immunoprecipitation with antibodies against known partners
Test specificity using structurally similar F-box proteins
For flow cytometry:
Establish appropriate fixation and permeabilization protocols
Perform fluorescence-minus-one controls
Validate with cells expressing fluorescently tagged FBXO30