FBXO15 antibodies are immunochemical reagents designed to bind specifically to the FBXO15 protein, a 49–55 kDa F-box protein involved in substrate recognition for ubiquitin-mediated proteolysis. These antibodies are essential for:
Detecting FBXO15 expression in tissues/cells via Western blot (WB), immunohistochemistry (IHC), and flow cytometry .
Studying FBXO15's regulatory roles in cancer metastasis, stem cell pluripotency, and mitochondrial function .
Breast Cancer Suppression: FBXO15 inhibits epithelial-mesenchymal transition (EMT) and cancer stem cell (CSC) phenotypes by promoting ubiquitination and degradation of SOX2 and STAT3. High FBXO15 expression correlates with improved survival in luminal-type breast cancer patients .
Drug Resistance: FBXO15 regulates P-glycoprotein (P-gp) expression via ubiquitination, influencing chemoresistance in cancer cells .
FBXO15 is highly expressed in pluripotent stem cells and testis tissues. It modulates mitochondrial morphology and cardiolipin synthesis, impacting cellular energy metabolism .
STAT3/SOX2 Degradation: FBXO15 binds to STAT3 and SOX2, enhancing their ubiquitination and proteasomal degradation, thereby suppressing EMT and metastasis in breast cancer .
P-glycoprotein Regulation: FBXO15 interacts with Ube2r1 to ubiquitinate P-gp, reducing its stability and mitigating drug resistance .
FBXO15 expression is suppressed by the EGFR/STAT3 pathway via miR-92a-3p, a microRNA upregulated in basal-type breast cancers. This axis creates a feedback loop that promotes cancer aggressiveness .
| Application | Dilution Range |
|---|---|
| Western Blot (WB) | 1:500 – 1:3000 |
| Immunohistochemistry (IHC) | 1:200 – 1:800 |
| Flow Cytometry | 10 µg/mL |
Prognostic Marker: Low FBXO15 expression correlates with poor survival in breast cancer patients, independent of subtype .
Therapeutic Target: Restoring FBXO15 activity could inhibit STAT3-driven metastasis or reverse chemoresistance by degrading P-gp .
FBXO15 (F-box protein 15) is a member of the F-box protein family that functions as a substrate recognition component within the SCF (Skp1-Cullin1-F-box) ubiquitin E3 ligase complex. F-box proteins are critical for the ubiquitin-mediated degradation of cellular regulatory proteins and are characterized by an approximately 40 amino acid F-box motif . FBXO15 plays significant roles in several cellular processes through its ability to target specific proteins for ubiquitination and subsequent proteasomal degradation.
Research has identified multiple substrates of FBXO15, including:
CLS1 (Cardiolipin Synthase 1), where FBXO15-mediated degradation impacts mitochondrial integrity and function
P-glycoprotein/ABCB1, where FBXO15 regulation affects multidrug resistance in cancer cells
Additionally, FBXO15 is expressed predominantly in undifferentiated embryonic stem cells and has been identified as a target of the pluripotency transcription factor Oct3/4, suggesting a potential role in stem cell biology .
FBXO15 antibodies have been validated for multiple research applications, with specific methodological considerations for each:
For all applications, it is recommended to titrate the antibody concentration to obtain optimal results for each specific experimental system and sample type .
Proper validation of FBXO15 antibody specificity is crucial for reliable experimental results. Implement the following methodological approaches:
Positive and negative controls:
Molecular weight verification:
Cross-reactivity assessment:
Alternative antibody comparison:
Compare results using antibodies targeting different epitopes of FBXO15
Correlate protein detection with mRNA expression data
Functional validation:
When investigating FBXO15's role in protein degradation pathways, consider these methodological approaches:
Ubiquitination assays:
Transfect cells with FLAG-tagged ubiquitin and/or HA-tagged FBXO15 constructs
Treat cells with proteasome inhibitors (e.g., MG132) 4-6 hours before harvesting to accumulate ubiquitinated proteins
Immunoprecipitate the substrate protein of interest and immunoblot for ubiquitin to detect ubiquitination
Protein stability assays:
Perform cycloheximide chase experiments to measure substrate protein half-life (t1/2)
Compare protein degradation kinetics between wild-type cells and those with FBXO15 overexpression or knockdown
Include proteasome inhibitors (MG132) and lysosomal inhibitors (leupeptin) to distinguish between degradation pathways
Substrate identification:
SCF complex analysis:
Investigate the association of FBXO15 with other SCF components (Skp1, Cullin1)
Use truncated mutants to identify domains required for substrate recognition and complex formation
Functional consequences:
Researchers encountering variable FBXO15 expression across experimental models should consider these methodological strategies:
Comprehensive expression profiling:
Regulatory mechanism investigation:
Alternative isoform detection:
Controlled expression systems:
Utilize inducible expression systems to achieve consistent FBXO15 levels across cell types
Consider viral transduction methods for cells that are difficult to transfect
Generate stable cell lines with uniform FBXO15 expression
Tissue-specific functionality:
Compare substrate targeting efficiency across cell types with different endogenous FBXO15 levels
Investigate cell-specific co-factors that might influence FBXO15 activity
For optimal FBXO15 detection in tissue sections, researchers should consider these methodological refinements:
Antigen retrieval optimization:
Antibody concentration titration:
Detection system selection:
Tissue preparation considerations:
Compare fresh-frozen versus formalin-fixed paraffin-embedded (FFPE) samples
Minimize fixation time to prevent excessive protein crosslinking
Consider performing section thickness optimization (4-8 μm)
Background reduction strategies:
Implement proper blocking steps with serum from the same species as the secondary antibody
Include endogenous peroxidase quenching step for chromogenic detection
Add avidin/biotin blocking for biotin-based detection systems
When encountering inconsistencies between different detection methods for FBXO15, implement these methodological approaches:
Epitope accessibility assessment:
Different applications (WB, IHC, ICC) expose different epitopes
Under reducing conditions (WB), epitopes may be more accessible than in fixed samples (IHC/ICC)
Test multiple antibodies targeting different regions of FBXO15
Sample preparation comparison:
Sensitivity threshold determination:
Establish detection limits for each method using serial dilutions of recombinant FBXO15
Consider signal amplification methods for samples with low expression
Use more sensitive detection methods (e.g., chemiluminescence for WB) for low-abundance samples
Protein modification interference:
Investigate if post-translational modifications affect antibody recognition
Include phosphatase or deubiquitinase treatments to remove modifications that might mask epitopes
Consider protein complex formation that might sequester epitopes
Methodological workflow standardization:
Implement consistent protocols across experiments
Document all variables that could affect detection (incubation times, temperatures, buffer compositions)
Maintain detailed records of antibody lot numbers and storage conditions
FBXO15 has multiple isoforms, and proper identification requires systematic analytical approaches:
Molecular weight profiling:
The calculated molecular weight of FBXO15 is 49 kDa, but it typically appears at approximately 55 kDa on SDS-PAGE
Different isoforms may appear as distinct bands (e.g., isoform 2 encompasses amino acids 298-434 of the full-length protein)
Use high-resolution gel systems (e.g., gradient gels) to resolve closely-migrating isoforms
Isoform-specific antibody selection:
Choose antibodies targeting regions that differ between isoforms
Consider using antibodies raised against specific isoforms when available
Validate antibody specificity with recombinant protein standards of each isoform
Transcript analysis correlation:
Genetic modification approaches:
Design isoform-specific siRNAs or shRNAs to selectively deplete individual variants
Express tagged versions of specific isoforms as positive controls
Use CRISPR/Cas9 to target isoform-specific exons
Mass spectrometry verification:
Perform immunoprecipitation followed by mass spectrometry
Identify isoform-specific peptides to confirm the presence of particular variants
Compare experimental spectra with theoretical peptide maps of known isoforms
Rigorous investigation of FBXO15's role in ubiquitination requires these control experiments:
Substrate specificity controls:
E3 ligase component controls:
Proteasome inhibition controls:
Genetic manipulation controls:
Ubiquitination assay controls:
Include samples without tagged ubiquitin as background controls
Use deubiquitinating enzyme inhibitors to preserve ubiquitin modifications
Perform experiments with ubiquitin mutants (e.g., K48R, K63R) to identify linkage types
FBXO15 has been implicated in mitochondrial function regulation through its targeting of CLS1, making it valuable for mitochondrial research:
Cardiolipin synthesis pathway investigation:
Mitochondrial morphology assessment:
Mitochondrial membrane potential analysis:
ATP production measurement:
Disease model applications:
Investigating tissue-specific and disease-related functions of FBXO15 requires these systematic approaches:
Comparative interactome analysis:
Perform immunoprecipitation of FBXO15 followed by mass spectrometry across different tissues
Compare protein interaction networks between normal and disease states
Validate tissue-specific interactions with co-immunoprecipitation and proximity ligation assays
Substrate competition assays:
Develop in vitro ubiquitination assays with purified components
Test multiple potential substrates simultaneously to assess preferential targeting
Investigate how tissue-specific factors might influence substrate selection
Domain mapping experiments:
Generate truncated or point-mutated FBXO15 constructs
Identify regions responsible for tissue-specific substrate recognition
Perform structural analysis of FBXO15-substrate complexes
Phosphorylation-dependent regulation:
Disease-specific expression profiling:
Compare FBXO15 levels across tissues in disease models (cancer, neurodegenerative disorders)
Correlate expression with disease progression markers
Develop tissue-specific conditional knockout models to assess function in vivo